• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 28
  • 12
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 81
  • 81
  • 25
  • 22
  • 22
  • 18
  • 16
  • 13
  • 11
  • 9
  • 9
  • 9
  • 7
  • 7
  • 7
  • 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.
51

Overexpression of HSPA12B Protects Against Cerebral Ischemia/Reperfusion Injury via a PI3K/Akt-Dependent Mechanism

Ma, Yujie, Lu, Chen, Li, Chuanfu, Li, Rongrong, Zhang, Yangyang, Ma, He, Zhang, Xiaojin, Ding, Zhengnian, Liu, Li 01 January 2013 (has links)
Background and purpose: HSPA12B is a newly discovered member of the Hsp70 family proteins. This study investigated the effects of HSPA12B on focal cerebral ischemia/reperfusion (I/R) injury in mice. Methods: Transgenic mice overexpressing human HSPA12B (Tg) and wild-type littermates (WT) were subjected to 60. min of middle cerebral artery occlusion to induce ischemia and followed by reperfusion (I/R). Neurological deficits, infarct volumes and neuronal death were examined at 6 and 24. hrs after reperfusion. Blood-brain-barrier (BBB) integrity and activated cellular signaling were examined at 3. hrs after reperfusion. Results: After cerebral I/R, Tg mice exhibited improvement in neurological deficits and decrease in infarct volumes, when compared with WT I/R mice. BBB integrity was significantly preserved in Tg mice following cerebral I/R. Tg mice also showed significant decreases in cell injury and apoptosis in the ischemic hemispheres. We observed that overexpression of HSPA12B activated PI3K/Akt signaling and suppressed JNK and p38 activation following cerebral I/R. Importantly, pharmacological inhibition of PI3K/Akt signaling abrogated the protection against cerebral I/R injury in Tg mice. Conclusions: The results demonstrate that HSPA12B protects the brains from focal cerebral I/R injury. The protective effect of HSPA12B is mediated though a PI3K/Akt-dependent mechanism. Our results suggest that HSPA12B may have a therapeutic potential against ischemic stroke.
52

HSPA12B Inhibits Lipopolysaccharide-Induced Inflammatory Response in Human Umbilical Vein Endothelial Cells

Wu, Jun, Li, Xuehan, Huang, Lei, Jiang, Surong, Tu, Fei, Zhang, Xiaojin, Ma, He, Li, Rongrong, Li, Chuanfu, Li, Yuehua, Ding, Zhengnian, Liu, Li 01 January 2015 (has links)
Heat shock protein A12B (HSPA12B) is a newly discovered member of the HSP70 protein family. This study investigated the effects of HSPA12B on lipopolysaccharide (LPS)-induced inflammatory responses in human umbilical vein endothelial cells (HUVECs) and the possible mechanisms involved. A HUVECs inflammatory model was induced by LPS. Overexpression of HSPA12B in HUVECs was achieved by infection with recombinant adenoviruses encoding green fluorescence protein-HSPA12B. Knockdown of HSPA12B was achieved by siRNA technique. Twenty four hours after virus infection or siRNA transfection, HUVECs were stimulated with 1 μg/ml LPS for 4 hrs. Endothelial cell permeability ability was determined by transwell permeability assay. The binding rate of human neutrophilic polymorphonuclear leucocytes (PMN) with HUVECs was examined using myeloperoxidase assay. Cell migrating ability was determined by the wound-healing assay. The mRNA and protein expression levels of interested genes were analyzed by RT-qPCR and Western blot, respectively. The release of cytokines interleukin-6 and tumour necrosis factor-α was measured by ELISA. HSPA12B suppressed LPS-induced HUVEC permeability and reduced PMN adhesion to HUVECs. HSPA12B also inhibited LPS-induced up-regulation of adhesion molecules and inflammatory cytokine expression. By contrast, knockdown of HSPA12B enhanced LPS-induced increases in the expression of adhesion molecules and inflammatory cytokines. Moreover, HSPA12B activated PI3K/Akt signalling pathway and pharmacological inhibition of this pathway by Wortmannin completely abrogated the protection of HSPA12B against inflammatory response in HUVECs. Our results suggest that HSPA12B attenuates LPS-induced inflammatory responses in HUVECs via activation of PI3K/Akt signalling pathway.
53

The Toll-Like Receptor 9 Agonist, CpG-Oligodeoxynucleotide 1826, Ameliorates Cardiac Dysfunction After Trauma-Hemorrhage

Zhang, Xia, Gao, Ming, Ha, Tuanzhu, Kalbfleisch, John H., Williams, David L., Li, Chuanfu, Kao, Race L. 01 August 2012 (has links)
Cardiovascular collapse is the major factor contributing to the mortality of trauma-hemorrhage (T-H) patients. Toll-like receptors (TLRs) play a critical role in T-H-induced cardiac dysfunction. This study evaluated the role of TLR9 agonist, CpG-oligodeoxynucleotide (ODN) 1826, in cardiac functional recovery after T-H. Trauma-hemorrhage was induced in a murine model by soft tissue injury and blood withdrawals from the jugular vein to a mean arterial pressure of 35 ± 5 mmHg. Mice were treated with CpG-ODN 1826 (10 μg/30 g body weight) by intraperitoneal injection 1 h before T-H (n = 5-8/group). Hemodynamic parameters were measured before, during hemorrhage, and at 60 min after T-H. Trauma-hemorrhage significantly decreased the mean arterial pressure and left ventricular pressure compared with sham controls. In contrast, CpG-ODN administration significantly attenuated the decrease in arterial pressure and left ventricular pressure due to T-H. Trauma-hemorrhage markedly decreased myocardial levels of phosphorylated Akt by 57.9%. However, CpG-ODN treatment significantly blunted the decrement in phospho-Akt by activating the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. The PI3K inhibitor LY294002 partially abolished CpG-induced cardioprotection, indicating that additional signaling pathways are involved in the protective effect of CpG-ODN after T-H. We observed that CpG-ODN treatment also significantly attenuated the decrease in myocardial phospho-ERK levels after T-H. Inhibition of ERK by U0126 also partially abolished the cardioprotective effect of CpG-ODN after T-H. Our data suggest that CpG-ODN significantly attenuates T-H-induced cardiac dysfunction. The mechanisms involve activation of both PI3K/Akt and ERK signaling pathways. The TLR9 agonist, CpG-ODN 1826, may provide a novel treatment strategy for preventing or managing cardiac dysfunction and enhancing recovery in T-H patients.
54

MEK/ERKs Signaling Is Essential for Lithium-Induced Neurite Outgrowth in N2a Cells

Wang, Zhuyao, Wang, Juan, Li, Jingjin, Wang, Xiaohui, Yao, Yuzhen, Zhang, Xiaojin, Li, Chuanfu, Cheng, Yunlin, Ding, Guoxian, Liu, Li, Ding, Zhengnian 01 June 2011 (has links)
Lithium, a drug used for the treatment of bipolar disorder, has been shown to affect different aspects of neuronal development such as neuritogenesis, neurogenesis and survival. The underlying mechanism responsible for lithium's influence on neuronal development, however, still remains to be elucidated. In the present study, we demonstrate that lithium increases the phosphorylation of extracellular-signal regulated kinases (ERKs) and protein kinase B (Akt) and promotes neurite outgrowth in mouse N2a neuroblastoma cells (N2a). The inactivation of mitogen-activated protein kinase kinase (MEK)/ERKs signaling with a MEK inhibitor inhibits neurite outgrowth, but it enhances Akt activation in lithium-treated N2a cells. Furthermore, the inactivation of phosphoinositide-3-kinase (PI3K)/Akt signaling with a PI3K inhibitor increases both lithium-induced ERKs activation and lithium-induced neurite outgrowth. Taken together, our study suggests that lithium-induced neurite outgrowth in N2a cells is regulated by cross-talk between the MEK/ERKs and PI3K/Akt pathways and requires the activation of the MEK/ERKs signaling.
55

TLR2 Ligands Induce Cardioprotection Against Ischaemia/Reperfusion Injury Through a PI3K/Akt-Dependent Mechanism

Ha, Tuanzhu, Hu, Yulong, Liu, Li, Lu, Chen, McMullen, Julie R., Kelley, Jim, Kao, Race L., Williams, David L., Gao, Xiang, Li, Chuanfu 01 September 2010 (has links)
Aims Toll-like receptor (TLR)-mediated signalling pathways have been implicated in myocardial ischaemia/reperfusion (I/R) injury. Activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway protects the myocardium from ischaemic injury. We hypothesized that the modulation of TLR2 would induce cardioprotection against I/R injury via activation of the PI3K/Akt signalling. Methods and results Mice were treated with TLR2 ligands, peptidoglycan (PGN) or Pam3CSK4, respectively, 1 h before the hearts were subjected to ischaemia (1 h), followed by reperfusion (4 h). Infarct size was determined by triphenyltetrazolium chloride staining. Cardiac function and haemodynamic performance were evaluated. Infarct size was significantly reduced in PGN-or Pam3CSK4-treated mice compared with untreated I/R mice. Administration of TLR2 ligands improved cardiac function following I/R. PGN treatment increased the levels of phospho-Akt and phospho-GSK-3β (glycogen synthase kinase-3β), compared with untreated I/R hearts. PGN stimulation increased TLR2 tyrosine phosphorylation and association of the p85 subunit of PI3K with TLR2. To investigate the role of PI3K/Akt signalling in PGN-induced cardioprotection, we administered the PI3K inhibitor, Wortmannin, to the mice 15 min before PGN treatment. We also administered PGN to kinase-deficient Akt (kdAkt) transgenic mice 1 h before myocardial I/R. Both PI3K inhibition and kdAkt mice abolished the cardioprotection induced by PGN. To examine the role of TLR2 in PGN-induced cardioprotection, we administrated PGN to TLR2 knockout mice 1 h before the hearts were subjected to I/R. PGN-induced cardioprotection was lost in TLR2-deficient mice. Conclusion These results demonstrate that TLR2 ligands induced cardioprotection, which is mediated through a TLR2/PI3K/Akt-dependent mechanism.
56

α-Lipoic Acid Prevents Bupivacaine-Induced Neuron Injury in Vitro Through a PI3K/Akt-Dependent Mechanism

Wang, Xiaohui, Zhang, Xiaojin, Cheng, Yunlin, Li, Chuanfu, Zhang, Wenbo, Liu, Li, Ding, Zhengnian 01 January 2010 (has links)
Background: Bupivacaine is an amide type local anesthetic which is widely used for epidural anesthesia and nerve blockade in patients. However, local administration of bupivacaine could cause neuron injury showing transient neurologic symptoms. α-Lipoic acid (LA) was shown to protect nerve cells from substance-induced injury. We hypothesized that LA administration could attenuate bupivacaine-induced neurotoxicity. Methods: To evaluate our hypothesis, we treated mouse neuroblastoma N2a cells with LA 30 min before the cells were exposed to bupivacaine. We evaluated cellular injury by examination of cell viability, morphology changes, nuclear condensation, and Annexin V staining. We also examined the levels of intracellular reactive oxygen species (ROS) and activation of PI3K/Akt signaling pathway. In a separate experiment, we determined the effect of Akt inhibition on cell viability in the presence of LA and bupivacaine. Results: Bupivacaine treatment significantly induced cell injury as evidenced by decreased cell viability, increased nuclear condensation and Annexin V staining. Administration of LA significantly attenuated bupivacaine-induced cell injury. In addition, LA treatment increased the levels of phospho-Akt and phospho-GSK3β and attenuated bupivacaine decreased the levels of ROS. More significantly, pharmacological inhibition of Akt abolished the LA-induced protection from bupivacaine-caused cell injury. Conclusions: Our findings suggest that pretreatment of neuroblastoma cells with LA protected neural cells from bupivacaine-induced injury. The mechanisms involve activation of the PI3K/Akt signaling pathway.
57

Attenuation of Cardiac Hypertrophy by Inhibiting Both mTOR and NFκB Activation in Vivo

Ha, Tuanzhu, Li, Yuehua, Gao, Xiang, McMullen, Julie R., Shioi, Tetsuo, Izumo, Seigo, Kelley, Jim L., Zhao, Aiqiu, Haddad, Georges E., Williams, David L., Browder, I. William, Kao, Race L., Li, Chuanfu 15 December 2005 (has links)
A role for the PI3K/Akt/mTOR pathway in cardiac hypertrophy has been well documented. We reported that NFκB activation is needed for cardiac hypertrophy in vivo. To investigate whether both NFκB activation and PI3K/Akt/mTOR signaling participate in the development of cardiac hypertrophy, two models of cardiac hypertrophy, namely, induction in caAkt-transgenic mice and by aortic banding in mice, were employed. Rapamycin (2 mg/kg/daily), an inhibitor of the mammalian target of rapamycin, and the antioxidant pyrrolidine dithiocarbamate (PDTC; 120 mg/kg/daily), which can inhibit NFκB activation, were administered to caAkt mice at 8 weeks of age for 2 weeks. Both rapamycin and PDTC were also administered to the mice immediately after aortic banding for 2 weeks. Administration of either rapamycin or PDTC separately or together to caAkt mice reduced the ratio of heart weight/body weight by 21.54, 32.68, and 42.07% compared with untreated caAkt mice. PDTC administration significantly reduced cardiac NFκB activation by 46.67% and rapamycin significantly decreased the levels of p70S6K by 34.20% compared with untreated caAkt mice. Similar results were observed in aortic-banding-induced cardiac hypertrophy in mice. Our results suggest that both NFκB activation and the PI3K/Akt signaling pathway participate in the development of cardiac hypertrophy in vivo.
58

Reduced Cardiac Hypertrophy in Toll-Like Receptor 4-Deficient Mice Following Pressure Overload

Ha, Tuanzhu, Li, Yuehua, Hua, Fang, Ma, Jinag, Gao, Xiang, Kelley, Jim, Zhao, Aiqiu, Haddad, Georges E., Williams, David L., Browder, I. William, Kao, Race L., Li, Chuanfu 01 November 2005 (has links)
Objective: We have previously demonstrated that nuclear factor kappa B (NFκB) activation is needed for the development of cardiac hypertrophy in vivo. NFκB is a downstream transcription factor in the Toll-like receptor (TLR)-mediated signaling pathway; therefore, we investigated a role of TLR4 in cardiac hypertrophy in vivo. Methods: TLR4-deficient mice (C.C3H-Tlr4 lps-d, n = 6), wild-type (WT) genetic background mice (BALB/c, n = 6), TLR4-deleted strain (C57BL/10ScCr, n = 8), and WT controls (C57BL/10ScSn, n = 8) were subjected to aortic banding for 2 weeks. Age-matched surgically operated mice served as controls. In a separate experiment, rapamycin (2 mg/kg, daily) was administered to TLR4-deficient mice and WT mice immediately following aortic banding. The ratio of heart weight/body weight (HW / BW) was calculated, and cardiac myocyte size was examined by FITC-labeled wheat germ agglutinin staining of membranes. NFκB binding activity and the levels of phospho-p70S6K in the myocardium were also examined. Results: Aortic banding significantly increased the ratio of HW / BW by 33.9% (0.601 ± 0.026 vs. 0.449 ± 0.004) and cell size by 68.4% in WT mice and by 10.00% (0.543 ± 0.011 vs. 0.495 ± 0.005) and by 11.8% in TLR4-deficient mice, respectively, compared with respective sham controls. NFκB binding activity and phospho-p70S6K levels were increased by 182.6% and 115.2% in aortic-banded WT mice and by 78.0% and 162.0% in aortic-banded TLR4-deficient mice compared with respective sham controls. In rapamycin-treated aortic-banded mice, the ratio of HW / BW was increased by 18.0% in WT mice and by 3.5% in TLR4-deficient mice compared with respective sham controls. Conclusion: Our results demonstrate that TLR4 is a novel receptor contributing to the development of cardiac hypertrophy in vivo and that both the TLR4-mediated pathway and PI3K/Akt/mTOR signaling are involved in the development of cardiac hypertrophy in vivo.
59

CHARACTERIZATION OF SIPL1-MEDIATED PTEN INACTIVATION DURING TUMORIGENESIS / INACTIVATION OF PTEN BY SIPL1

De Melo, Jason Anthony 11 1900 (has links)
As the primary antagonist to the tumorigenic PI3K/AKT pathway, PTEN is classified as a tumor suppressor. The inactivation of PTEN through genetic or post-translational modifications is a critical step in the tumorigenesis of many breast cancers (BCs). SIPL1 is a novel protein which was identified as a PTEN negative regulator. To further explore SIPL1-mediated PTEN inactivation, we analyzed 17 datasets covering 3484 BC cases and 228 normal individuals. SIPL1 gene amplification and increased mRNA expression correlates with the progression and poor prognosis of ER and/or PR positive tumors. Furthermore, examination of a BC tissue microarray containing 224 tumor cases revealed elevated SIPL1 protein expression in ER+ and PR+ tumors and was associated with greater AKT activation. Additionally, ectopic expression of SIPL1 in CHO-K1 cells resulted in increased AKT activation and cell proliferation, and cytoskeleton reorganization alongside with PTEN downregulation. SIPL1 contributes to the linear polyubiquitination of NEMO, suggesting a role for SIPL1 in PTEN ubiquitination. Indeed, it was SIPL1, not the SIPL1-∆UBL (a PTEN-binding defective mutant) which robustly induced PTEN polyubiquitination in a lysine (K) 63-dependent but K48-independent manner. While K48-linked polyubiquitin chains direct protein degradation, K63-linked chains regulate a variety of protein functions. SIPL1 binds polyubiquitinated PTEN with significantly higher affinity than non-ubiquitinated PTEN. A SIPL1 mutant, SIPL1-TFLV, is unable to cause PTEN ubiquitination but is capable of PTEN association. Collectively, our results reveal that SIPL1 interacts with PTEN with a low affinity, which results in PTEN polyubiquitination, and that the modification may stabilize the association between SIPL1 and PTEN. We propose a model where SIPL1 mediates the K63-linked ubiquitination of PTEN inactivating it. The downregulation of PTEN, when paired with the tumor-promoting effects of ER and/or PR, stimulates breast tumorigenesis. SIPL1 is an important BC marker and future research should focus on its potential as a therapeutic target. / Thesis / Doctor of Philosophy (PhD)
60

Keratinocyte growth factor as a survival factor in human breast cancer

Chang, Hsiang-Lin 02 December 2005 (has links)
No description available.

Page generated in 0.0329 seconds