Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
Cell ; 143(7): 1072-83, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21183071

ABSTRACT

The heart has the ability to grow in size in response to exercise, but little is known about the transcriptional mechanisms underlying physiological hypertrophy. Adult cardiomyocytes have also recently been proven to hold the potential for proliferation, a process that could be of great importance for regenerative medicine. Using a unique RT-PCR-based screen against all transcriptional components, we showed that C/EBPß was downregulated with exercise, whereas the expression of CITED4 was increased. Reduction of C/EBPß in vitro and in vivo resulted in a phenocopy of endurance exercise with cardiomyocyte hypertrophy and proliferation. This proliferation was mediated, at least in part, by the increased CITED4. Importantly, mice with reduced cardiac C/EBPß levels displayed substantial resistance to cardiac failure upon pressure overload. These data indicate that C/EBPß represses cardiomyocyte growth and proliferation in the adult mammalian heart and that reduction in C/EBPß is a central signal in physiologic hypertrophy and proliferation.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Heart/physiology , Physical Conditioning, Animal , Animals , Cell Proliferation , Cells, Cultured , Embryo, Nonmammalian/metabolism , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Myocardium/cytology , Myocytes, Cardiac/metabolism , Rats , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology
2.
Exp Cell Res ; 439(2): 114097, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38796135

ABSTRACT

Leucine-rich α2-glycoprotein-1 (LRG1) is overexpressed in various cancers, including non-small cell lung cancer (NSCLC), but its role in NSCLC cell metastasis is not well understood. In this study, NSCLC cell exosomes were analyzed using different techniques, and the impact of exosomal LRG1 on NSCLC cell behavior was investigated through various assays both in vitro and in vivo. The study revealed that LRG1, found abundantly in NSCLC cells and exosomes, enhanced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). Exosomal LRG1 was shown to promote NSCLC cell metastasis in animal models. Additionally, the interaction between LRG1 and fibronectin 1 (FN1) in the cytoplasm was identified. It was observed that FN1 could counteract the effects of LRG1 knockdown on cell regulation induced by exosomes derived from NSCLC cells. Overall, the findings suggest that targeting exosomal LRG1 or FN1 may hold therapeutic potential for treating NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Cell Proliferation , Exosomes , Fibronectins , Glycoproteins , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Exosomes/metabolism , Exosomes/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Cell Proliferation/genetics , Fibronectins/metabolism , Fibronectins/genetics , Animals , Glycoproteins/metabolism , Glycoproteins/genetics , Cell Movement/genetics , Mice , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Mice, Nude , Neoplasm Metastasis , Mice, Inbred BALB C , Gene Expression Regulation, Neoplastic , A549 Cells
3.
Arterioscler Thromb Vasc Biol ; 43(2): 330-349, 2023 02.
Article in English | MEDLINE | ID: mdl-36453275

ABSTRACT

BACKGROUND: Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFß/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS: We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS: Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS: Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.


Subject(s)
Atherosclerosis , Fatty Liver , Humans , Animals , Mice , Liver/metabolism , Inflammation/genetics , Inflammation/prevention & control , Inflammation/metabolism , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Activins/genetics , Activins/metabolism , Fatty Liver/genetics , Fatty Liver/prevention & control , Cholesterol/metabolism , Metabolic Networks and Pathways , Receptors, LDL/genetics , Receptors, LDL/metabolism , Mice, Knockout , Mice, Inbred C57BL
4.
Circulation ; 145(16): 1218-1233, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35114812

ABSTRACT

BACKGROUND: The heart grows in response to pathological and physiological stimuli. The former often precedes cardiomyocyte loss and heart failure; the latter paradoxically protects the heart and enhances cardiomyogenesis. The mechanisms underlying these differences remain incompletely understood. Although long noncoding RNAs (lncRNAs) are important in cardiac development and disease, less is known about their roles in physiological hypertrophy or cardiomyogenesis. METHODS: RNA sequencing was applied to hearts from mice after 8 weeks of voluntary exercise-induced physiological hypertrophy and cardiomyogenesis or transverse aortic constriction for 2 or 8 weeks to induce pathological hypertrophy or heart failure. The top lncRNA candidate was overexpressed in hearts with adeno-associated virus vectors and inhibited with antisense locked nucleic acid-GapmeRs to examine its function. Downstream effectors were identified through promoter analyses and binding assays. The functional roles of a novel downstream effector, dachsous cadherin-related 2 (DCHS2), were examined through transgenic overexpression in zebrafish and cardiac-specific deletion in Cas9-knockin mice. RESULTS: We identified exercise-regulated cardiac lncRNAs, called lncExACTs. lncExACT1 was evolutionarily conserved and decreased in exercised hearts but increased in human and experimental heart failure. Cardiac lncExACT1 overexpression caused pathological hypertrophy and heart failure; lncExACT1 inhibition induced physiological hypertrophy and cardiomyogenesis, protecting against cardiac fibrosis and dysfunction. lncExACT1 functioned by regulating microRNA-222, calcineurin signaling, and Hippo/Yap1 signaling through DCHS2. Cardiomyocyte DCHS2 overexpression in zebrafish induced pathological hypertrophy and impaired cardiac regeneration, promoting scarring after injury. In contrast, murine DCHS2 deletion induced physiological hypertrophy and promoted cardiomyogenesis. CONCLUSIONS: These studies identify lncExACT1-DCHS2 as a novel pathway regulating cardiac hypertrophy and cardiomyogenesis. lncExACT1-DCHS2 acts as a master switch toggling the heart between physiological and pathological growth to determine functional outcomes, providing a potentially tractable therapeutic target for harnessing the beneficial effects of exercise.


Subject(s)
Cadherin Related Proteins/metabolism , Heart Failure , MicroRNAs , RNA, Long Noncoding , Animals , Cardiomegaly/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Zebrafish/genetics
5.
Opt Express ; 30(7): 11563-11571, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35473098

ABSTRACT

We report electrically pumped continuous-wave (CW) InAs/GaAs quantum dot lasers directly grown on planar exact silicon (001) with asymmetric waveguide structures. Surface hydrogen-annealing for the GaAs/ Si (001) templates and low-temperature growth for GaInP upper cladding layers were combined in the growth of the laser structure to achieve a high slope efficiency. For the broad-stripe edge-emitting lasers with 2-mm cavity length and 20-µm stripe width made from the above laser structure, a threshold current density of 203.5 A/cm2 and a single-facet slope efficiency of 0.158 W/A are achieved at ∼1.31 µm band under CW conditions. The extrapolated mean-time-to-failure reaches up to 21000 hours at room temperature, which is deduced from the data measured from C-mount packaged devices. Importantly, these results can provide a practical strategy to realize 1.3 µm wavelength band distributed feedback lasers directly on planar exact Si (001) templates with thin buffer layers.

6.
Lasers Med Sci ; 38(1): 10, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36539630

ABSTRACT

Aiming to the personalized laser therapy of nevus of Ota (NO), a local thermal non-equilibrium model was employed to optimize laser wavelength, pulse duration, and energy density under different melanin depth and volume fraction. According to our simulation, the optimal pulse duration is between 15 and 150 ns to limit heat transfer inside the hyperplastic melanin, and 50 ns is recommended to decrease the energy absorption by normal melanin in epidermis. Correlations of the minimum and the maximum energy densities are proposed with respect to melanin depth and volume fraction for the 755-nm and 1064-nm lasers. For the same NO type, the therapy window of the 755-nm laser is larger than that of 1064-nm. For NO with shallow depth or low volume fraction, the 755-nm laser is recommended to make the treatment more stable owing to its lager therapy window. For deeper depth or higher volume fraction, the 1064-nm laser is recommended to avoid thermal damage of epidermis. Through comparison with clinical data, the optimized laser parameters are proved practicable since high cure rate can be achieved when energy density falls into the range of predicted therapy window. With developing of non-invasive measurement technology of melanin content and distribution, personalized treatment of NO maybe possible in the near future.


Subject(s)
Laser Therapy , Low-Level Light Therapy , Nevus of Ota , Skin Neoplasms , Humans , Nevus of Ota/radiotherapy , Nevus of Ota/surgery , Melanins , Skin Neoplasms/radiotherapy , Skin Neoplasms/surgery
8.
Cardiovasc Res ; 120(3): 262-272, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38084908

ABSTRACT

AIMS: Physiological cardiac hypertrophy occurs in response to exercise and can protect against pathological stress. In contrast, pathological hypertrophy occurs in disease and often precedes heart failure. The cardiac pathways activated in physiological and pathological hypertrophy are largely distinct. Our prior work demonstrated that miR-222 increases in exercised hearts and is required for exercise-induced cardiac hypertrophy and cardiomyogenesis. Here, we sought to define the role of miR-222 in pathological hypertrophy. METHODS AND RESULTS: We found that miR-222 also increased in pathological hypertrophy induced by pressure overload. To assess its functional significance in this setting, we generated a miR-222 gain-of-function model through cardiac-specific constitutive transgenic miR-222 expression (TgC-miR-222) and used locked nucleic acid anti-miR specific for miR-222 to inhibit its effects. Both gain- and loss-of-function models manifested normal cardiac structure and function at baseline. However, after transverse aortic constriction (TAC), miR-222 inhibition accelerated the development of pathological hypertrophy, cardiac dysfunction, and heart failure. Conversely, miR-222-overexpressing mice had less pathological hypertrophy after TAC, as well as better cardiac function and survival. We identified p53-up-regulated modulator of apoptosis, a pro-apoptotic Bcl-2 family member, and the transcription factors, Hmbox1 and nuclear factor of activated T-cells 3, as direct miR-222 targets contributing to its roles in this context. CONCLUSION: While miR-222 is necessary for physiological cardiac growth, it inhibits cardiac growth in response to pressure overload and reduces adverse remodelling and cardiac dysfunction. These findings support the model that physiological and pathological hypertrophy are fundamentally different. Further, they suggest that miR-222 may hold promise as a therapeutic target in pathological cardiac hypertrophy and heart failure.


Subject(s)
Heart Diseases , Heart Failure , MicroRNAs , Mice , Animals , MicroRNAs/genetics , Cardiomegaly/metabolism , Heart Failure/metabolism , Heart , Heart Diseases/pathology , Myocytes, Cardiac/metabolism , Disease Models, Animal , Homeodomain Proteins/metabolism
9.
Circulation ; 126(18): 2208-19, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23019294

ABSTRACT

BACKGROUND: Heart failure is a growing cause of morbidity and mortality. Cardiac phosphatidylinositol 3-kinase signaling promotes cardiomyocyte survival and function, but it is paradoxically activated in heart failure, suggesting that chronic activation of this pathway may become maladaptive. Here, we investigated the downstream phosphatidylinositol 3-kinase effector, serum- and glucocorticoid-regulated kinase-1 (SGK1), in heart failure and its complications. METHODS AND RESULTS: We found that cardiac SGK1 is activated in human and murine heart failure. We investigated the role of SGK1 in the heart by using cardiac-specific expression of constitutively active or dominant-negative SGK1. Cardiac-specific activation of SGK1 in mice increased mortality, cardiac dysfunction, and ventricular arrhythmias. The proarrhythmic effects of SGK1 were linked to biochemical and functional changes in the cardiac sodium channel and could be reversed by treatment with ranolazine, a blocker of the late sodium current. Conversely, cardiac-specific inhibition of SGK1 protected mice after hemodynamic stress from fibrosis, heart failure, and sodium channel alterations. CONCLUSIONS: SGK1 appears both necessary and sufficient for key features of adverse ventricular remodeling and may provide a novel therapeutic target in cardiac disease.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Heart Failure/enzymology , Immediate-Early Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Ventricular Remodeling/physiology , Acetanilides/therapeutic use , Animals , Cardiomegaly, Exercise-Induced , Consensus Sequence , Disease Models, Animal , Electrocardiography , Enzyme Induction , Humans , Hypertension/complications , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Ion Channel Gating/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Piperazines/therapeutic use , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Ranolazine , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Tachycardia, Ventricular/enzymology , Tachycardia, Ventricular/etiology
10.
Angiogenesis ; 16(4): 773-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23666122

ABSTRACT

Cell-based therapies to restore heart function after infarction have been tested in pre-clinical models and clinical trials with mixed results, and will likely require both contractile cells and a vascular network to support them. We and others have shown that human endothelial colony forming cells (ECFC) combined with mesenchymal progenitor cells (MPC) can be used to "bio-engineer" functional human blood vessels. Here we investigated whether ECFC + MPC form functional vessels in ischemic myocardium and whether this affects cardiac function or remodeling. Myocardial ischemia/reperfusion injury (IRI) was induced in 12-week-old immunodeficient rats by ligation of the left anterior descending coronary artery. After 40 min, myocardium was reperfused and ECFC + MPC (2 × 10(6) cells, 2:3 ratio) or PBS was injected. Luciferase assays after injection of luciferase-labeled ECFC + MPC showed that 1,500 ECFC were present at day 14. Human ECFC-lined perfused vessels were directly visualized by femoral vein injection of a fluorescently-tagged human-specific lectin in hearts injected with ECFC + MPC but not PBS alone. While infarct size at day 1 was no different, LV dimensions and heart weight to tibia length ratios were lower in cell-treated hearts compared with PBS at 4 months, suggesting post-infarction remodeling was ameliorated by local cell injection. Fractional shortening, LV wall motion score, and fibrotic area were not different between groups at 4 months. However, pressure-volume loops demonstrated improved cardiac function and reduced volumes in cell-treated animals. These data suggest that myocardial delivery of ECFC + MPC at reperfusion may provide a therapeutic strategy to mitigate LV remodeling and cardiac dysfunction after IRI.


Subject(s)
Cord Blood Stem Cell Transplantation , Endothelium, Vascular/physiopathology , Hematopoietic Stem Cells/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Myocardial Reperfusion Injury/physiopathology , Neovascularization, Physiologic/physiology , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling , Adult , Animals , Endothelial Cells/cytology , Feasibility Studies , Genes, Reporter , Hemodynamics , Heterografts , Humans , Microvessels/ultrastructure , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/surgery , Rats , Rats, Nude , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology
11.
Am J Physiol Heart Circ Physiol ; 303(1): H75-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22561297

ABSTRACT

Cardiac mammalian target of rapamycin (mTOR) is necessary and sufficient to prevent cardiac dysfunction in pathological hypertrophy. However, the role of cardiac mTOR in heart failure after ischemic injury remains undefined. To address this question, we used transgenic (Tg) mice with cardiac-specific overexpression of mTOR (mTOR-Tg mice) to study ischemia-reperfusion (I/R) injury in two animal models: 1) in vivo I/R injury with transient coronary artery ligation and 2) ex vivo I/R injury in Langendorff-perfused hearts with transient global ischemia. At 28 days after I/R, mortality was lower in mTOR-Tg mice than littermate control mice [wild-type (WT) mice]. Echocardiography and MRI demonstrated that global cardiac function in mTOR-Tg mice was preserved, whereas WT mice exhibited significant cardiac dysfunction. Masson's trichrome staining showed that 28 days after I/R, the area of interstitial fibrosis was smaller in mTOR-Tg mice compared with WT mice, suggesting that adverse left ventricular remodeling is inhibited in mTOR-Tg mice. In the ex vivo I/R model, mTOR-Tg hearts demonstrated improved functional recovery compared with WT hearts. Perfusion with Evans blue after ex vivo I/R yielded less staining in mTOR-Tg hearts than WT hearts, indicating that mTOR overexpression inhibited necrosis during I/R injury. Expression of proinflammatory cytokines, including IL-6 and TNF-α, in mTOR-Tg hearts was lower than in WT hearts. Consistent with this, IL-6 in the effluent post-I/R injury was lower in mTOR-Tg hearts than in WT hearts. These findings suggest that cardiac mTOR overexpression in the heart is sufficient to provide substantial cardioprotection against I/R injury and suppress the inflammatory response.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , TOR Serine-Threonine Kinases/physiology , Animals , Autophagy , Blotting, Western , Coronary Vessels/physiology , DNA/genetics , DNA/isolation & purification , Fibrosis , In Vitro Techniques , Inflammation/genetics , Inflammation/pathology , Ligation , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Necrosis , Perfusion , Real-Time Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics , Ultrasonography
12.
Life Sci Alliance ; 4(12)2021 12.
Article in English | MEDLINE | ID: mdl-34663679

ABSTRACT

Extracellular vesicles (EVs) mediate intercellular signaling by transferring their cargo to recipient cells, but the functional consequences of signaling are not fully appreciated. RBC-derived EVs are abundant in circulation and have been implicated in regulating immune responses. Here, we use a transgenic mouse model for fluorescence-based mapping of RBC-EV recipient cells to assess the role of this intercellular signaling mechanism in heart disease. Using fluorescent-based mapping, we detected an increase in RBC-EV-targeted cardiomyocytes in a murine model of ischemic heart failure. Single cell nuclear RNA sequencing of the heart revealed a complex landscape of cardiac cells targeted by RBC-EVs, with enrichment of genes implicated in cell proliferation and stress signaling pathways compared with non-targeted cells. Correspondingly, cardiomyocytes targeted by RBC-EVs more frequently express cellular markers of DNA synthesis, suggesting the functional significance of EV-mediated signaling. In conclusion, our mouse model for mapping of EV-recipient cells reveals a complex cellular network of RBC-EV-mediated intercellular communication in ischemic heart failure and suggests a functional role for this mode of intercellular signaling.


Subject(s)
Erythrocytes/metabolism , Extracellular Vesicles/metabolism , Heart Failure/blood , Myocardial Infarction/blood , Myocardium/metabolism , RNA, Nuclear/genetics , RNA-Seq/methods , Signal Transduction/genetics , Single-Cell Analysis/methods , Animals , Cell Communication/genetics , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Female , Healthy Volunteers , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism
13.
Am J Physiol Cell Physiol ; 299(6): C1256-66, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861467

ABSTRACT

Previous studies have suggested that inhibition of the mammalian target of rapamycin (mTOR) by rapamycin suppresses myocardial hypertrophy. However, the role of mTOR in the progression of cardiac dysfunction in pathological hypertrophy has not been fully defined. Interestingly, recent reports indicate that the inflammatory response, which plays an important role in the development of heart failure, is enhanced by rapamycin under certain conditions. Our aim in this study was to determine the influence of mTOR on pathological hypertrophy and to assess whether cardiac mTOR regulates the inflammatory response. We generated transgenic mice with cardiac-specific overexpression of wild-type mTOR (mTOR-Tg). mTOR-Tg mice were protected against cardiac dysfunction following left ventricular pressure overload induced by transverse aortic constriction (TAC) (P < 0.01) and had significantly less interstitial fibrosis compared with littermate controls (WT) at 4 wk post-TAC (P < 0.01). In contrast, TAC caused cardiac dysfunction in WT. At 1 wk post-TAC, the proinflammatory cytokines interleukin (IL)-1ß and IL-6 were significantly increased in WT mice but not in mTOR-Tg mice. To further characterize the effects of mTOR activation, we exposed HL-1 cardiomyocytes transfected with mTOR to lipopolysaccharide (LPS). mTOR overexpression suppressed LPS-induced secretion of IL-6 (P < 0.001), and the mTOR inhibitors rapamycin and PP242 abolished this inhibitory effect of mTOR. In addition, mTOR overexpression reduced NF-κB-regulated transcription in HL-1 cells. These data suggest that mTOR mitigates adverse outcomes of pressure overload and that this cardioprotective effect of mTOR is mediated by regulation of the inflammatory reaction.


Subject(s)
Cardiomegaly/physiopathology , Heart/physiopathology , Myocytes, Cardiac/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/enzymology , Cardiomegaly/pathology , Female , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Interleukin-1beta/analysis , Interleukin-6/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myocytes, Cardiac/pathology , Rats , TOR Serine-Threonine Kinases/genetics
14.
J Am Heart Assoc ; 9(11): e014199, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32458746

ABSTRACT

Background Delivery of hydrogels to the heart is a promising strategy for mitigating the detrimental impact of myocardial infarction (MI). Challenges associated with the in vivo delivery of currently available hydrogels have limited clinical translation of this technology. Gelatin methacryloyl (GelMA) bioadhesive hydrogel could address many of the limitations of available hydrogels. The goal of this proof-of-concept study was to evaluate the cardioprotective potential of GelMA in a mouse model of MI. Methods and Results The physical properties of GelMA bioadhesive hydrogel were optimized in vitro. Impact of GelMA bioadhesive hydrogel on post-MI recovery was then assessed in vivo. In 20 mice, GelMA bioadhesive hydrogel was applied to the epicardial surface of the heart at the time of experimental MI. An additional 20 mice underwent MI but received no GelMA bioadhesive hydrogel. Survival rates were compared for GelMA-treated and untreated mice. Left ventricular function was assessed 3 weeks after experimental MI with transthoracic echocardiography. Left ventricular scar burden was measured with postmortem morphometric analysis. Survival rates at 3 weeks post-MI were 89% for GelMA-treated mice and 50% for untreated mice (P=0.011). Left ventricular contractile function was better in GelMA-treated than untreated mice (fractional shortening 37% versus 26%, P<0.001). Average scar burden in GelMA-treated mice was lower than in untreated mice (6% versus 22%, P=0.017). Conclusions Epicardial GelMA bioadhesive application at the time of experimental MI was performed safely and was associated with significantly improved post-MI survival compared with control animals. In addition, GelMA treatment was associated with significantly better preservation of left ventricular function and reduced scar burden.


Subject(s)
Gelatin/administration & dosage , Methacrylates/administration & dosage , Myocardial Infarction/drug therapy , Myocardium/pathology , Tissue Adhesives/administration & dosage , Animals , Disease Models, Animal , Drug Compounding , Fibrosis , Gelatin/chemistry , Hydrogels , Methacrylates/chemistry , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Proof of Concept Study , Tissue Adhesives/chemistry , Ventricular Function, Left
15.
Sci Transl Med ; 11(482)2019 03 06.
Article in English | MEDLINE | ID: mdl-30842316

ABSTRACT

Activin type II receptor (ActRII) ligands have been implicated in muscle wasting in aging and disease. However, the role of these ligands and ActRII signaling in the heart remains unclear. Here, we investigated this catabolic pathway in human aging and heart failure (HF) using circulating follistatin-like 3 (FSTL3) as a potential indicator of systemic ActRII activity. FSTL3 is a downstream regulator of ActRII signaling, whose expression is up-regulated by the major ActRII ligands, activin A, circulating growth differentiation factor-8 (GDF8), and GDF11. In humans, we found that circulating FSTL3 increased with aging, frailty, and HF severity, correlating with an increase in circulating activins. In mice, increasing circulating activin A increased cardiac ActRII signaling and FSTL3 expression, as well as impaired cardiac function. Conversely, ActRII blockade with either clinical-stage inhibitors or genetic ablation reduced cardiac ActRII signaling while restoring or preserving cardiac function in multiple models of HF induced by aging, sarcomere mutation, or pressure overload. Using unbiased RNA sequencing, we show that activin A, GDF8, and GDF11 all induce a similar pathologic profile associated with up-regulation of the proteasome pathway in mammalian cardiomyocytes. The E3 ubiquitin ligase, Smurf1, was identified as a key downstream effector of activin-mediated ActRII signaling, which increased proteasome-dependent degradation of sarcoplasmic reticulum Ca2+ ATPase (SERCA2a), a critical determinant of cardiomyocyte function. Together, our findings suggest that increased activin/ActRII signaling links aging and HF pathobiology and that targeted inhibition of this catabolic pathway holds promise as a therapeutic strategy for multiple forms of HF.


Subject(s)
Activin Receptors, Type II/metabolism , Aging/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Activins/blood , Adult , Aged , Aged, 80 and over , Aging/blood , Animals , Constriction, Pathologic , Disease Models, Animal , Follistatin-Related Proteins/metabolism , Frailty , Heart Failure/blood , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Ligands , Male , Mice, Inbred C57BL , Middle Aged , Myocytes, Cardiac/metabolism , Pressure , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Severity of Illness Index , Systole
16.
EBioMedicine ; 32: 172-181, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29779700

ABSTRACT

Despite substantial declines in mortality following myocardial infarction (MI), subsequent left ventricular remodeling (LVRm) remains a significant long-term complication. Extracellular small non-coding RNAs (exRNAs) have been associated with cardiac inflammation and fibrosis and we hypothesized that they are associated with post-MI LVRm phenotypes. RNA sequencing of exRNAs was performed on plasma samples from patients with "beneficial" (decrease LVESVI ≥ 20%, n = 11) and "adverse" (increase LVESVI ≥ 15%, n = 11) LVRm. Selected differentially expressed exRNAs were validated by RT-qPCR (n = 331) and analyzed for their association with LVRm determined by cardiac MRI. Principal components of exRNAs were associated with LVRm phenotypes post-MI; specifically, LV mass, LV ejection fraction, LV end systolic volume index, and fibrosis. We then investigated the temporal regulation and cellular origin of exRNAs in murine and cell models and found that: 1) plasma and tissue miRNA expression was temporally regulated; 2) the majority of the miRNAs were increased acutely in tissue and at sub-acute or chronic time-points in plasma; 3) miRNA expression was cell-specific; and 4) cardiomyocytes release a subset of the identified miRNAs packaged in exosomes into culture media in response to hypoxia/reoxygenation. In conclusion, we find that plasma exRNAs are temporally regulated and are associated with measures of post-MI LVRm.


Subject(s)
Cell-Free Nucleic Acids/blood , Fibrosis/diet therapy , Fish Oils/administration & dosage , Myocardial Infarction/diet therapy , Adult , Aged , Contrast Media/therapeutic use , Female , Fibrosis/blood , Fibrosis/diagnostic imaging , Fibrosis/pathology , Humans , Magnetic Resonance Imaging , Male , MicroRNAs/blood , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , RNA, Small Untranslated/genetics , Stroke Volume/genetics , Ventricular Function, Left/genetics , Ventricular Remodeling/drug effects
17.
JAMA Cardiol ; 3(9): 871-876, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30090932

ABSTRACT

Importance: Mortality is high among patients heart failure (HF) who are receiving treatment, and therefore identifying new pathways rooted in preclinical cardiac remodeling phenotypes may afford novel biomarkers and therapeutic avenues. Circulating extracellular RNAs (ex-RNAs) are an emerging class of biomarkers with target-organ epigenetic effects relevant to myocardial biology, although large human investigations remain limited. Objective: To measure the association of highly expressed circulating ex-RNAs with left ventricular remodeling and incident HF in a community-based cohort. Design, Setting, and Participants: This is a prospective observational cohort study of individuals who were included in the eighth examination of the Framingham Offspring Cohort (2005-2008). Collected data include measurements of the left ventricle via electrocardiography, determination of circulating ex-RNAs in plasma, and incidence of heart failure. Data analysis was completed from December 2016 to June 2018. Exposures: A total of 398 circulating ex-RNA molecules in plasma were measured by reverse transcription polymerase chain reaction; disease ontology analysis was also performed. Main Outcomes and Measures: Echocardiographic indices of left ventricular (LV) remodeling and incident heart failure. Results: A total of 2763 participants of the Framingham Heart Study with measured ex-RNAs (mean [SD] age, 66.3 [9.0] years; 1499 [54.3%] female) were included in this study. Of this sample, 2429 to 2432 individuals had echocardiographic measures recorded (depending on the measurement). A total of 2681 individuals had HF status determined, of whom 116 (4.3%) experienced HF (median [interquartile range] follow-up, 7.7 [6.6-8.6] years). We identified 12 ex-RNAs associated with LV mass and at least 1 other echocardiographic phenotype (LV end-diastolic volume or left atrial dimension). Of these 12 ex-RNAs, 3 micro RNAs (miR-17, miR-20a, and miR-106b) were associated with a 15% reduction in long-term incident HF per 2-fold increase in circulating level during the follow-up period, after adjustments for age, sex, established HF risk factors, and prevalent or interim myocardial infarction. These 3 RNAs shared sequence homology and targeted a shared group of messenger RNAs that specified pathways relevant to HF (eg, transforming growth factor-ß signaling, growth/cell cycle, and apoptosis), and shared a disease association with hypertension in disease ontology analysis. Conclusions and Relevance: This study identifies a group of circulating, noncoding RNAs associated with echocardiographic phenotypes, long-term incident HF, and pathways relevant to myocardial remodeling in a large community-based sample. Further investigations into the functional biology of these ex-RNAs are warranted for surveillance for HF prevention.


Subject(s)
Heart Failure/epidemiology , Heart Ventricles/diagnostic imaging , MicroRNAs/blood , Myocardial Infarction/epidemiology , Ventricular Remodeling , Aged , Echocardiography , Female , Heart Failure/genetics , Humans , Incidence , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Prospective Studies , Survival Analysis
18.
Circulation ; 112(1): 84-92, 2005 Jul 05.
Article in English | MEDLINE | ID: mdl-15983244

ABSTRACT

BACKGROUND: In the heart, the expressions of several types of prostanoid receptors have been reported. However, their roles in cardiac hypertrophy in vivo remain unknown. We intended to clarify the roles of these receptors in pressure overload-induced cardiac hypertrophy using mice lacking each of their receptors. METHODS AND RESULTS: We used a model of pressure overload-induced cardiac hypertrophy produced by banding of the transverse aorta in female mice. In wild-type mice subjected to the banding, cardiac hypertrophy developed during the observation period of 8 weeks. In mice lacking the prostaglandin (PG) I2 receptor (IP(-/-)), however, cardiac hypertrophy and cardiomyocyte hypertrophy were significantly greater than in wild-type mice at 2 and 4 weeks but not at 8 weeks, whereas there was no such augmentation in mice lacking the prostanoid receptors other than IP. In addition, cardiac fibrosis observed in wild-type hearts was augmented in IP(-/-) hearts, which persisted for up to 8 weeks. In IP(-/-) hearts, the expression level of mRNA for atrial natriuretic peptide, a representative marker of cardiac hypertrophy, was significantly higher than in wild-type hearts. In vitro, cicaprost, an IP agonist, reduced platelet-derived growth factor-induced proliferation of wild-type noncardiomyocytes, although it could not inhibit cardiotrophin-1-induced hypertrophy of cardiomyocytes. Accordingly, cicaprost increased cAMP concentration efficiently in noncardiomyocytes. CONCLUSIONS: IP plays a suppressive role in the development of pressure overload-induced cardiac hypertrophy via the inhibition of both cardiomyocyte hypertrophy and cardiac fibrosis. Both effects have been suggested as originating from the action on noncardiomyocytes rather than cardiomyocytes.


Subject(s)
Cardiomegaly/etiology , Hypertension/complications , Receptors, Epoprostenol/physiology , Animals , Biomarkers/analysis , Cardiomegaly/pathology , Cell Enlargement , Cyclic AMP/blood , Disease Models, Animal , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Fibrosis , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , RNA, Messenger/analysis , Receptors, Epoprostenol/deficiency , Receptors, Epoprostenol/genetics
19.
Front Biosci (Landmark Ed) ; 21(3): 479-86, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26709788

ABSTRACT

Clinical factors and liver biopsy cannot accurately predict the risk of developing cirrhosis in chronic hepatitis B (CHB).This study was to develop a predictive gene signature for cirrhosis in CHB patients. A total of 183 untreated CHB patients were enrolled. GeneChip, significant analysis of microarray (SAM) and prediction analysis of microarray (PAM) were used to select predictor genes (PGs) in liver tissues. The Cirrhosis Risk Score (CRS) was calculated based on 6 PG variables and the predictive value of CRS was evaluated. Firstly differentially expressed genes were filtered from a genome scan and SAM, and 87 significant genes were selected for the signature building. Secondly a signature consisting of 6 PGs (CD24, CXCL6, EHF, ITGBL1, LUM and SOX9) most predictive for cirrhosis risk in CHB patients was developed in the selection set (n=40) by use of PAM and PCR approach. Finally the CRS was calculated to estimate the risk of developing cirrhosis and then tested in validation cohort (n=143). The area under the ROC curves (AUROC) of the CRS was 0.944 and exceeded to 6 PGs and clinical factors. A low CRS cutoff of 6.43 to identify low-risk patients would misclassify only 8.16% of high-risk patients, while a high cutoff of 8.32 to identify high-risk patients would misclassify 0% of low-risk patients. So CRS is a better predictor than clinical factors in differentiating high-risk versus low-risk for cirrhosis and application of CRS in clinical practice could help to reduce the rate of liver biopsy in patients with CHB.


Subject(s)
Genetic Predisposition to Disease , Hepatitis B, Chronic/genetics , Liver Cirrhosis/genetics , Algorithms , Gene Expression Profiling , Humans , Risk Factors
20.
JCI Insight ; 1(9)2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27430023

ABSTRACT

The mechanisms by which exercise mediates its multiple cardiac benefits are only partly understood. Prior comprehensive analyses of the cardiac transcriptional components and microRNAs dynamically regulated by exercise suggest that the CBP/p300-interacting protein CITED4 is a downstream effector in both networks. While CITED4 has documented functional consequences in neonatal cardiomyocytes in vitro, nothing is known about its effects in the adult heart. To investigate the impact of cardiac CITED4 expression in adult animals, we generated transgenic mice with regulated, cardiomyocyte-specific CITED4 expression. Cardiac CITED4 expression in adult mice was sufficient to induce an increase in heart weight and cardiomyocyte size with normal systolic function, similar to the effects of endurance exercise training. After ischemia-reperfusion, CITED4 expression did not change initial infarct size but mediated substantial functional recovery while reducing ventricular dilation and fibrosis. Forced cardiac expression of CITED4 also induced robust activation of the mTORC1 pathway after ischemic injury. Moreover, pharmacological inhibition of mTORC1 abrogated CITED4's effects in vitro and in vivo. Together, these data establish CITED4 as a regulator of mTOR signaling that is sufficient to induce physiologic hypertrophy at baseline and mitigate adverse ventricular remodeling after ischemic injury.

SELECTION OF CITATIONS
SEARCH DETAIL