• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 81
  • 35
  • 24
  • 11
  • 10
  • 8
  • 6
  • 4
  • 3
  • Tagged with
  • 216
  • 55
  • 52
  • 50
  • 35
  • 25
  • 24
  • 24
  • 24
  • 23
  • 21
  • 20
  • 18
  • 18
  • 17
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
111

Effects of Cccp-Induced Mitochondrial Uncoupling and Cyclosporin a on Cell Volume, Cell Injury and Preconditioning Protection of Isolated Rabbit Cardiomyocytes

Ganote, Charles E., Armstrong, Stephen C. 01 July 2003 (has links)
Cell swelling may contribute to acute cell injury subsequent to ischemia/reperfusion. The potential role of mitochondrial uncoupling and the resultant mitochondrial swelling, due to opening of the mitochondrial permeability transition pore (MPTP), were examined in an in vitro ischemically pelleted isolated rabbit cardiomyocyte model using the protonophore, carbonyl cyanide m-chlorophenylhydrazone (CCCP) to uncouple mitochondria. Cyclosporin A (CsA) was employed to inhibit MPTP opening. Cell volume was determined by a cell-flotation, density-gradient assay, using bromododecane. Cell viability, subsequent to an osmotic stress, was determined by trypan blue permeability. Ischemic preconditioning (IPC) facilitated volume regulation following an osmotic stress. Ischemic-cell swelling was reduced by IPC. IPC protected ischemically pelleted cells, but CsA had no significant effects on injury or IPC protection. CCCP ischemia accelerated rates of ischemic contracture and injury, and abolished IPC protection. IPC protection was restored by CsA. In CCCP-ischemic-uncoupled cells, subjected to a reduced (170 mOsm) osmotic stress, CsA and IPC afforded independent and additive protection. Chelerythrine and 5-hydroxydecanoate (5-HD) blocked IPC, but did not reduce CsA protection. Electron microscopy confirmed that CCCP ischemia induced mitochondrial matrix swelling that was reduced by CsA. Cardioprotection by IPC and CsA was accompanied by proportional reductions in cell swelling. Morphometric analysis of the electron photomicrographs demonstrated that the mitochondrial volume fractions were significantly reduced in the CsA/CCCP (29.8 ± 2.3%, P < 0.004) and IPC/CsA/CCCP (31.5 ± 1.7%, P < 0.0008) groups as compared to the CCCP-ischemic group (40.5 ± 1.7%) The IPC/CCCP group (39.5 ± 4.2%) was not significantly different from the CCCP-ischemic group. NIM 811, a CsA analogue MPTP blocker with no calcineurin inhibitory activity, afforded protection similar to CsA. The results suggest that CsA protection may, in part, be mediated by reduction of mitochondrial swelling.
112

Differential Translocation or Phosphorylation of Alpha B Crystallin Cannot Be Detected in Ischemically Preconditioned Rabbit Cardiomyocytes

Armstrong, Stephen C., Shivell, Christine L., Ganote, Charles E. 01 January 2000 (has links)
Alpha B Crystallin (αBC) is a putative effector protein of ischemic preconditioning (IPC). that is phosphorylated on Ser 45 by ERK1/2 and Set 59 by the p38 MAPK substrate, MAPKAPK-2. Translocation and phosphorylation of αBC was determined in cytosolic and cytoskeletal fractions by 1D SDS-PAGE and IEF, or using Ser 45 and Set 59 phospho-specific antibodies in: (1) control rabbit cardiomyocytes; (2) cells preconditioned by 10 min in vitro ischemia; or after pre-treatment with specific inhibitors of (3) Ser/Thr protein phosphatase 1/2A (calyculin A); (4) p38 MAPK (SB203580); or (5) ERK 1/2 (PD98059); all prior to 180 min ischemia. Ischemia induced a cytosolic to cytoskeletal translocation of αBC, which was similar in all the groups. Highly phosphorylated isoforms (D1/2) of αBC were present in cytosolic but not cytoskeletal fractions at 0 min ischemia. By 60-90 min ischemia. D1/2 isoforms had translocated to the cytoskeletal fraction. Calyculin A maintained D1/2 levels throughout prolonged ischemia. SB203580 decreased αBC phosphorylation. Neither PD98059 nor IPC altered αBC phosphorylation during prolonged ischemia. It is concluded that αBC phosphorylation during ischemia is regulated by p38 MAPK but not by ERK 1/2. The inability to detect a correlation between IPC protection and either αBC translocation or phosphorylation suggests that the proteins in the highly phosphorylated isoform bands of αBC quantitated in this study are not protective end effectors of classical IPC.
113

Sphingosylphosphorylcholine Promotes the Differentiation of Resident Sca-1 Positive Cardiac Stem Cells to Cardiomyocytes Through Lipid raft/JNK/STAT3 and β-catenin Signaling Pathways

Li, Wenjing, Liu, Honghong, Liu, Pingping, Yin, Deling, Zhang, Shangli, Zhao, Jing 01 July 2016 (has links)
Resident cardiac Sca-1-positive (+) stem cells may differentiate into cardiomyocytes to improve the function of damaged hearts. However, little is known about the inducers and molecular mechanisms underlying the myogenic conversion of Sca-1+ stem cells. Here we report that sphingosylphosphorylcholine (SPC), a naturally occurring bioactive lipid, induces the myogenic conversion of Sca-1+ stem cells, as evidenced by the increased expression of cardiac transcription factors (Nkx2.5 and GATA4), structural proteins (cardiac Troponin T), transcriptional enhancer (Mef2c) and GATA4 nucleus translocation. First, SPC activated JNK and STAT3, and the JNK inhibitor SP600125 or STAT3 inhibitor stattic impaired the SPC-induced expression of cardiac transcription factors and GATA4 nucleus translocation, which suggests that JNK and STAT3 participated in SPC-promoted cardiac differentiation. Moreover, STAT3 activation was inhibited by SP600125, whereas JNK was inhibited by β-cyclodextrin as a lipid raft breaker, which indicates a lipid raft/JNK/STAT3 pathway involved in SPC-induced myogenic transition. β-Catenin, degraded by activated GSK3β, was inhibited by SPC. Furthermore, GSK3β inhibitors weakened but the β-catenin inhibitor promoted SPC-induced differentiation. We found no crosstalk between the lipid raft/JNK/STAT3 and β-catenin pathway. Our study describes a lipid, SPC, as an endogenic inducer of myogenic conversion in Sca-1+ stem cells with low toxicity and high efficiency for uptake.
114

Sarcolemmal Blebs and Osmotic Fragility as Correlates of Irreversible Ischemic Injury in Preconditioned Isolated Rabbit Cardiomyocytes

Armstrong, Stephen C., Shivell, Christine L., Ganote, Charles E. 01 January 2001 (has links)
The hypothesis that irreversible ischemic injury is related to sub-sarcolemmal blebbing and an inherent osmotic fragility of the blebs was tested by subjecting isolated control and ischemically preconditioned (IPC) or calyculin A (CalA)-pretreated (protected) rabbit cardiomyocytes to ischemic pelleting followed by resuspension in 340, 170 or 85 mosmol medium containing trypan blue. At time points from 0-240 min, osmotic fragility was assessed by the percentage of trypan blue permeable cells. Membrane blebs were visualized with India ink preparations. Bleb formation, following acute hypo-osmotic swelling, developed by 75 min and increased with longer periods of ischemia. Osmotic fragility developed only after 75 min. Cells resuspended in 340 mosmol media did not form blebs and largely retained the ability to exclude trypan blue, even after 240 min ischemia. Although the latent tendency for osmotic blebbing preceded the development of osmotic fragility, most osmotically fragile cells became permeable without evident sarcolemmal bleb formation. The onset of osmotic fragility was delayed in protected cells, but protection did not reduce the bleb formation. It is concluded that blebbing and osmotic fragility are independent manifestations of ischemic injury. The principal locus of irreversible ischemic injury and the protection provided by IPC may lie within the sarcolemma rather than at sarcolemmal attachments to underlying adherens junctions.
115

β-Cyclodextrin Induces the Differentiation of Resident Cardiac Stem Cells to Cardiomyocytes Through Autophagy

Shi, Xingxing, Li, Wenjing, Liu, Honghong, Yin, Deling, Zhao, Jing 01 August 2017 (has links)
Cardiac stem cells (CSCs) have emerged as promising cell candidates to regenerate damaged hearts, because of the potential in differentiating to cardiomyocytes. However, the differentiation is difficult to trigger without inducers. Here we reported that β-cyclodextrin (β-CD) increased the expression of cardiac transcription factors (Nkx2.5 and GATA4), structural proteins (cardiac Troponin T, cTnt), transcriptional enhancer (Mef2c) and induced GATA4 nucleus translocation in adult resident CSCs, thus β-CD could be used to enhance myogenic transition. As the differentiation process was accompanied by autophagy, we constructed the Atg5 knockdown cell line by using the Atg5 siRNA lentivirus, and the myogenic conversion was blocked in Atg5 knockdown cells, which suggested that β-CD induces the cardiomyocytes transition of resident CSCs through autophagy. Furthermore, we found that JNK/STAT3 and GSK3β/β-catenin was the downstream pathways of β-CD-induced autophagy and differentiation using the inhibitors. Moreover, β-CD performed its functions through improving intracellular cholesterol levels and affecting cholesterol efflux. Collectively, our results reveal that β-CD as a novel tool to induce myogenic transition of CSCs, which could mobilize the resident CSCs or used together with CSCs to enhance the therapy effects of CSCs on damaged hearts. In addition, the clarified molecular mechanisms supported the new targets for inducing cardiomyocyte differentiation.
116

Phosphoinositide-3-kinase/akt - Dependent Signaling is Required for Maintenance of [Ca<sup>2+</sup>]<sub>I,</sub>I<sub>Ca</sub>, and Ca<sup>2+</sup> Transients in HL-1 Cardiomyocytes

Graves, Bridget M., Simerly, Thomas, Li, Chuanfu, Williams, David L., Wondergem, Robert 22 June 2012 (has links)
The phosphoinositide 3-kinases (PI3K/Akt) dependent signaling pathway plays an important role in cardiac function, specifically cardiac contractility. We have reported that sepsis decreases myocardial Akt activation, which correlates with cardiac dysfunction in sepsis. We also reported that preventing sepsis induced changes in myocardial Akt activation ameliorates cardiovascular dysfunction. In this study we investigated the role of PI3K/Akt on cardiomyocyte function by examining the role of PI3K/Akt-dependent signaling on [Ca 2+]i, Ca2+ transients and membrane Ca2+ current, ICa, in cultured murine HL-1 cardiomyocytes. LY294002 (120 μM), a specific PI3K inhibitor, dramatically decreased HL-1 [Ca 2+]i, Ca2+ transients and ICa. We also examined the effect of PI3K isoform specific inhibitors, i.e. α (PI3-kinase α inhibitor 2; 28 nM); ? (TGX-221; 100 nM) and γ (AS-252424; 100 nM), to determine the contribution of specific isoforms to HL-1 [Ca 2+]i regulation. Pharmacologic inhibition of each of the individual PI3K isoforms significantly decreased [Ca2+]i, and inhibited Ca 2+ transients. Triciribine (120 μM), which inhibits AKT downstream of the PI3K pathway, also inhibited [Ca2+]i, and Ca 2+ transients and ICa. We conclude that the PI3K/Akt pathway is required for normal maintenance of [Ca2+]i in HL-1 cardiomyocytes. Thus, myocardial PI3K/Akt-PKB signaling sustains [Ca 2+]i required for excitation-contraction coupling in cardiomyoctyes.
117

β-arrestin2/miR-155/GSK3β Regulates Transition of 5'-Azacytizine-Induced Sca-1-Positive Cells to Cardiomyocytes

Zhao, Jing, Feng, Yimin, Yan, Hui, Chen, Yangchao, Wang, Jinlan, Chua, Balvin, Stuart, Charles, Yin, Deling 01 January 2014 (has links)
Stem-cell antigen 1-positive (Sca-1+) cardiac stem cells (CSCs), a vital kind of CSCs in humans, promote cardiac repair in vivo and can differentiate to cardiomyocytes with 5'-azacytizine treatment in vitro. However, the underlying molecular mechanisms are unknown. b-arrestin2 is an important scaffold protein and highly expressed in the heart. To explore the function of b-arrestin2 in Sca-1+ CSC differentiation, we used b-arrestin2-knockout mice and overexpression strategies. Real-time PCR revealed that b-arrestin2 promoted 5'-azacytizine-induced Sca-1+ CSC differentiation in vitro. Because the microRNA 155 (miR-155) may regulate b-arrestin2 expression, we detected its role and relationship with b-arrestin2 and glycogen synthase kinase 3 (GSK3β), another probable target of miR-155. Real-time PCR revealed that miR-155, inhibited by b-arrestin2, impaired 5'-azacytizine-induced Sca-1+ CSC differentiation. On luciferase report assay, miR-155 could inhibit the activity of b-arrestin2 and GSK3β, which suggests a loop pathway between miR-155 and b-arrestin2. Furthermore, b-arrestin2-knockout inhibited the activity of GSK3β. Akt, the upstream inhibitor of GSK3β, was inhibited in b-arrestin2-Knockout mice, so the activity of GSK3β was regulated by b-arrestin2 not Akt. We transplanted Sca-1+ CSCs from b-arrestin2-knockout mice to mice with myocardial infarction and found similar protective functions as in wild-type mice but impaired arterial elastance. Furthermore, low level of b-arrestin2 agreed with decreased phosphorylation of AKT and increased phophorylation of GSK3β, similar to in vitro findings. The β-arrestin2/miR-155/GSK3β pathway may be a new mechanism with implications for treatment of heart disease.
118

Analysis of catecholamine-induced beta-adrenergic signaling in TTS by patient-specific pluripotent stem cell-derived cardiomyocytes

Guessoum, Celina Isabelle 09 June 2020 (has links)
No description available.
119

Nano-structural Analysis of Engrafted Human Induced Pluripotent Stem Cell-derived Cardiomyocytes in Mouse Hearts Using a Genetic-probe APEX2 / 遺伝子プローブAPEX2を用いたマウス心臓における生着ヒトiPS細胞由来心筋細胞のナノ構造解析

Hatani, Takeshi 23 January 2019 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(医学) / 甲第21455号 / 医博第4422号 / 新制||医||1033(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 山下 潤, 教授 湊谷 謙司, 教授 浅野 雅秀 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
120

Investigating Cardiac Metabolism in Barth Syndrome Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes

Fatica, Erica Marie 02 May 2019 (has links)
No description available.

Page generated in 0.0624 seconds