19 research outputs found

    Effects of telmisartan on fat distribution: a meta-analysis of randomized controlled trials

    No full text
    <p><b>Objectives</b>: Several meta-analyses have confirmed the positive metabolic effects of telmisartan, an angiotensin II receptor blocker that can also act as a partial peroxisome proliferator-activated receptor-γ agonist, compared to those of other angiotensin II receptor blockers. These effects include decreased fasting glucose, glycosylated hemoglobin, interleukin-6, and tumor necrosis factor-α levels. However, no systemic analysis of telmisartan’s effects on body fat distribution has been performed. We performed a meta-analysis of randomized controlled telmisartan trials to investigate its effects on body weight, fat distribution, and visceral adipose reduction. <b>Research design and methods</b>: A literature search was performed using Embase, MEDLINE, and the Cochrane Library between January 1966 and November 2013. Randomized controlled trials in English and meeting the following criterion were included: random assignment of hypertensive participants with overweight/obesity, metabolic syndrome, or glucose intolerance to telmisartan or control therapy group. <b>Results</b>: Of 651 potentially relevant reports, 15 satisfied the inclusion criterion. While visceral fat area was significantly lower in the telmisartan group than in the control group (weighted mean difference = −18.13 cm<sup>2</sup>, 95% C.I. = −27.16 to −9.11, <i>P<sub>χ</sub></i><sup>2</sup> = 0.19, <i>I</i><sup>2</sup> = 41%), subcutaneous fat area was similar (weighted mean difference =2.94 cm<sup>2</sup>, 95% C.I. = −13.01 to 18.89, <i>P<sub>χ</sub></i><sup>2</sup> = 0.30, <i>I</i><sup>2</sup> = 17%). Total cholesterol levels were significantly different between the groups (standardized mean difference = −0.24, 95% C.I. = −0.45 to −0.03, <i>P<sub>χ</sub></i><sup>2</sup> = 0.0002, <i>I</i><sup>2</sup> = 67%). <b>Limitations</b>: Limitations include: (1) limited number of studies, especially those evaluating fat distribution; (2) different imaging modalities to assess visceral fat area (V.F.A.) and subcutaneous fat area (S.F.A.); (3) observed heterogeneity. <b>Conclusion</b>: The findings suggest that telmisartan affected fat distribution, inducing visceral fat reduction, and thus could be useful in hypertensive patients with obesity/overweight, metabolic syndrome, or glucose intolerance.</p

    MTD-mediated parkin delivery to the brain.

    No full text
    <p>(<b>A–B</b>) Immunoblotting of parkin proteins in the cerebellum. Sagittal sections through the cerebellum were immunostatined with anti-parkin (<b>A</b>) or anti-MTD10 (<b>B</b>) antibody 2 hrs after IP injection of 200 µg of diluent alone or His-tagged parkin proteins without (HP) or with the MTD13 or MTD10 sequences (HPM<sub>13</sub> or PM<sub>10</sub>) (<b>C–D</b>) Western blot analysis of brain parkin. Lysates were prepared from brain samples 2 hrs (<b>C</b>) and 30 hrs (<b>D</b>) after IV administration of diluent alone or 200 µg His-tagged parkin proteins without (HP) or with the MTD01 (HPM<sub>01</sub>) or MTD13 (HPM<sub>13</sub>) sequences (<b>C</b>) or with untagged parkin protein containing MTD10 (PM<sub>10</sub>) (<b>D</b>) and analyzed by western blotting using anti-parkin and anti-β-actin antibodies.</p

    CP-Parkin stimulates dopamine expression in MPTP-lesioned mice.

    No full text
    <p>(<b>A</b>) Experimental design. 8-week-old C57BL/6 female mice received three doses of MPTP on days 1 and 2 and were injected IP on days 3 through 7 with diluent alone, or with 10 mg/kg of parkin proteins (IP) with (HPM<sub>13</sub> and PM<sub>10</sub>) or without (HP) a MTD sequence. Urine and brain dopamine levels, gross motor function and brain lesions (TH immunostaining) were analyzed on subsequent days as indicated. (<b>B</b>) Dopamine levels in the urine of MPTP-lesioned mice. Urine dopamine levels in MPTP-lesioned mice were measured by ELISA 1, 2, 4, 6 and 8 hrs after HP and HPM<sub>13</sub> protein treatment. Values from 5 mice are presented as means ± S.D. Experimental differences between groups were assessed by a Student’s two-paired <i>t</i>-test (*<i>p</i><0.005). (<b>C</b>) Striatal dopamine levels in MPTP-lesioned mice. Dopamine levels in striatal biopsies were determined by ELISA in lesioned mice without protein treatment or after daily treatments with PM<sub>10</sub> as shown in panel A. Dopamine levels in groups of 5 mice are presented as means ± S.D. Experimental differences between groups were assessed by a Student’s two-paired <i>t</i>-test (*<i>p</i><0.01 and **<i>p</i><0.05).</p

    CP-Parkin reduces acute MPTP-induced neurotoxicity and preserves motor function.

    No full text
    <p>(<b>A</b>) PM<sub>10</sub> Reduces MPTP-induced dopaminergic toxicity. MPTM-lesioned mice were treated with PM<sub>10</sub> for 5 days as shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0102517#pone-0102517-g005" target="_blank">Figure 5A</a> (IP, 10 mg/kg) and loss/preservation of dopaminergic neurons was determined by tyrosine hydroxylase (TH) staining (left panel). (<b>B</b>) HPM<sub>13</sub> preserves gross motor function of MPTM-lesioned mice. 9 hrs after the last MPTP treatment (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0102517#pone-0102517-g005" target="_blank">Figure 5A</a>) mice were treated for 3 hrs with 200 µg proteins (IP, HP or HPM<sub>13</sub>), and motor ability was assessed by placing the animals in a water bath and video recording subsequent movements. The percentage of time of the mice in each treatment group were engaged in 4 legged motion is presented as means ± S.D. The number of mice in each group was as follows: Diluent, 16; MPTP only, 11; MPTP+HP, 7; MPTP+HPM<sub>13</sub>, 13. Experimental differences between groups were assessed by ANOVA (*<i>p</i><0.05, **<i>p</i><0.005, ***<i>p</i><0.001).</p

    Auto-ubiquitination activity of recombinant parkin proteins.

    No full text
    <p>E3 ligase activity of purified recombinant parkin proteins was assessed by an auto-ubiquitination assay. 1 µg of each of the indicated proteins was incubated for 1 hr at 37°C with 1 µM E1, 50 µM E2, 1 mM histidine-tagged Ubiquitin and 10 mM Mg-ATP, and the reaction products were fractionated by SDS PAGE and immunoblotted against an anti-Ubiquitin antibody. Parkin proteins (P, HP, PM<sub>10</sub>, PM<sub>13</sub>, PM<sub>151</sub> and PM<sub>174</sub>) are described in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0102517#pone-0102517-g001" target="_blank">Figure 1</a>. Samples without individual components or containing an unrelated protein, 6xHis-NM23 (HN), were used as negative controls.</p

    Structure and expression of MTD-parkin fusion proteins.

    No full text
    <p>(<b>A</b>) Structures of parkin fusion proteins. 6xHis-tagged parkin proteins (left) contained His-tag (HP) only or together with MTD01 (HPM<sub>01</sub>) or MTD13 (HPM<sub>13</sub>) sequences. Parkin proteins without the 6xHis tag (right) included native parkin (P) and proteins containing C-terminal MTD10 (PM<sub>10</sub>), MTD13 (PM<sub>13</sub>) MTD151 (PM<sub>151</sub>) and MTD174 (PM<sub>174</sub>) sequences. (<b>B–C</b>) Protein expression in <i>E. coli</i>. SDS PAGE analysis of cell lysates before (−) and after (+) IPTG induction; aliquots of Ni<sup>2+</sup> affinity purified proteins (P); and molecular weight standards (M). The size (number of amino acids), yield (mg/L) and solubility of each recombinant protein are indicated. Solubility was scored on a 4-point scale from highly soluble, with little tendency to precipitate (++++), to largely insoluble proteins (+).</p

    Cell-Permeable Parkin Proteins Suppress Parkinson Disease-Associated Phenotypes in Cultured Cells and Animals

    No full text
    <div><p>Parkinson’s disease (PD) is a neurodegenerative disorder of complex etiology characterized by the selective loss of dopaminergic neurons, particularly in the substantia nigra. Parkin, a tightly regulated E3 ubiquitin ligase, promotes the survival of dopaminergic neurons in both PD and Parkinsonian syndromes induced by acute exposures to neurotoxic agents. The present study assessed the potential of cell-permeable parkin (CP-Parkin) as a neuroprotective agent. Cellular uptake and tissue penetration of recombinant, enzymatically active parkin was markedly enhanced by the addition of a hydrophobic macromolecule transduction domain (MTD). The resulting CP-Parkin proteins (HPM<sub>13</sub> and PM<sub>10</sub>) suppressed dopaminergic neuronal toxicity in cells and mice exposed to 6-hydroxydopamine (6-OHDH) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). These included enhanced survival and dopamine expression in cultured CATH.a and SH-SY5Y neuronal cells; and protection against MPTP-induced damage in mice, notably preservation of tyrosine hydroxylase-positive cells with enhanced dopamine expression in the striatum and midbrain, and preservation of gross motor function. These results demonstrate that CP-Parkin proteins can compensate for intrinsic limitations in the parkin response and provide a therapeutic strategy to augment parkin activity in vivo.</p></div

    CP-Parkin protects neuronal cells from 6-OHDA-induced apoptosis.

    No full text
    <p>(<b>A</b>) Suppression of apoptosis in dopaminergic CATH.a cells. CATH.a cells at 5% (Low) or 70% (High) confluence were incubated with 50 µM 6-hydroxydopamine (6-OHDA, Agonist) for 1 hr, treated for 2.5 hrs with 2.5 µM HP or HPM<sub>13</sub> and assessed for apoptosis by TUNEL staining. The micrographs (left panels) are representative of three independent experiments, plotted (right panels) as means ± S.D. Experimental differences between groups were assessed by a Student’s two-paired <i>t</i>-test (*<i>p</i><0.001). (<b>B</b>) Suppression of apoptosis in SH-SY5Y cells. Apoptosis in SH-SY5Y treated with 6-OHDA with and without PM<sub>10</sub> was assessed as described in (<b>A</b>). (<b>C</b>) HPM<sub>13</sub> enhances dopamine release from CATH.a cells. The cells were incubated with 80 µM tyrosine for 24 hrs, treated for 5 hrs with 2.5 µM HP or HPM<sub>13</sub>, and levels of secreted dopamine were measured by ELISA. The data are presented as means ± S.D. of 4 independent experiments. Experimental differences between groups were assessed by a Student’s two-paired <i>t</i>-test (*<i>p</i><0.01 and **<i>p</i> 0.05).</p

    Identification of long noncoding RNAs involved in muscle differentiation

    No full text
    <div><p>Long noncoding RNAs (lncRNAs) are a large class of regulatory RNAs with diverse roles in cellular processes. Thousands of lncRNAs have been discovered; however, their roles in the regulation of muscle differentiation are unclear because no comprehensive analysis of lncRNAs during this process has been performed. In the present study, by combining diverse RNA sequencing datasets obtained from public database, we discovered lncRNAs that could behave as regulators in the differentiation of smooth or skeletal muscle cells. These analyses confirmed the roles of previously reported lncRNAs in this process. Moreover, we discovered dozens of novel lncRNAs whose expression patterns suggested their possible involvement in the phenotypic switch of vascular smooth muscle cells. The comparison of lncRNA expression change suggested that many lncRNAs have common roles during the differentiation of smooth and skeletal muscles, while some lncRNAs may have opposite roles in this process. The expression change of lncRNAs was highly correlated with that of their neighboring genes, suggesting that they may function as cis-acting lncRNAs. Furthermore, within the lncRNA sequences, there were binding sites for miRNAs with expression levels inversely correlated with the expression of corresponding lncRNAs during differentiation, suggesting a possible role of these lncRNAs as competing endogenous RNAs. The lncRNAs identified in this study will be a useful resource for future studies of gene regulation during muscle differentiation.</p></div

    Identification of long noncoding RNAs (lncRNAs) involved in phenotypic change of vascular smooth muscle cells (VSMCs).

    No full text
    <p>(A) The VSMCs with the synthetic phenotype can be differentiated into less the proliferative and contractile phenotypic by the overexpression of myocardin (MYOCD) or by treatment with transforming growth factor beta (TGFβ). The cells with the contractile phenotype can be converted into the more proliferative and synthetic phenotype by treatment with platelet-derived growth factor (PDGF). (B) Expression level of representative genes previously known to be involved in phenotypic change of VSMCs. The RNA-seq data of PDGF-treated venous smooth muscle cells, MYOCD-overexpressing human coronary artery smooth muscle (HCASM) cells, and TGFβ-treated HCASM cells were obtained from the Gene Expression Omnibus (GEO) (see <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0193898#sec002" target="_blank">Methods</a>). The fragments per kilobase of exon model per million mapped fragments (FPKM) values of a protein-coding gene (<i>CNN1</i>) and a long noncoding RNA (<i>SMILR</i>) are depicted in the y-axis. Error bars indicate standard errors from four samples for the PDGF set, or deviation from two samples for the MYOCD and TGFβ sets. (C) Expression profiles of lncRNAs during phenotypic change of VSMCs were analyzed. The calculation of lncRNAs’ expression changes is described in Method section. The gene clusters with prominent changes between the synthetic and contractile phenotypes are indicated with boxes. Detailed expression changes of genes from those clusters are depicted in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0193898#pone.0193898.s001" target="_blank">S1</a> and <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0193898#pone.0193898.s002" target="_blank">S2</a> Figs.</p
    corecore