14 research outputs found

    Gastrocutaneous Closure Device

    Get PDF
    A gastrocutaneous closure device allows gastrocutaneous fistula closure from external abdominal access through the fistula site. Access through the fistula ensures accurate closure placement on the interior lumen wall of the stomach. A closure or clip has a plurality of prongs defined by a deformable material, such that the prongs extend radially from a central hub in an arcuate or curved, semicircular shape. The arcuate shape converges towards a central point or axis at a distal end, and the proximate end of the prongs attaches to the central hub such that the prongs radiate from the hub and the distal end curves back toward the axis through the hub. The deformable prongs may therefore radially compress or retract to define a larger or smaller diameter. The fistula lies on the axis such that the biased, inserted prongs pull the inner stomach wall closed around the healing fistula

    Analgesic Efficacy of Pfannenstiel Incision Infiltration with Ropivacaine 7.5 mg/mL for Caesarean Section

    Get PDF
    Background. Pain after Caesarean delivery is partly related to Pfannenstiel incision, which can be infiltrated with local anaesthetic solutions. Methods. A double- blind randomized control trial was designed to assess the analgesic efficacy of 7.5 mg/mL ropivacaine solution compared to control group, in two groups of one hundred and forty four parturients for each group, who underwent Caesarean section under spinal anaesthesia: group R (ropivacaine group) and group C (control group). All parturients also received spinal sufentanil (2.5 μg). Results. Ropivacaine infiltration in the Pfannenstiel incision for Caesarean delivery before wound closure leads to a reduction of 30% in the overall consumption of analgesics (348 550 mg for group R versus 504 426 mg for group C with P < .05), especially opioids in the first 24 hours, but also significantly increases the time interval until the first request for an analgesic (4 h 20 min ± 2 h 26 for group R versus 2 h 42 ± 1 h 30 for group C). The P values for the two groups were: P < .0001 for paracetamol, P < .0001 for ketoprofen and P for nalbuphine which was the most significant. There is no significant difference in the threshold of VAS in the two series. Conclusion. This technique can contribute towards a programme of early rehabilitation in sectioned mothers, with earlier discharge from the post-labour suite

    Inhibition of Acetyl-CoA Carboxylase 1 (ACC1) and 2 (ACC2) Reduces Proliferation and <i>De Novo</i> Lipogenesis of EGFRvIII Human Glioblastoma Cells

    No full text
    <div><p>Tumor cell proliferation and migration processes are regulated by multiple metabolic pathways including glycolysis and de novo lipogenesis. Since acetyl-CoA carboxylase (ACC) is at the junction of lipids synthesis and oxidative metabolic pathways, we investigated whether use of a dual ACC inhibitor would provide a potential therapy against certain lipogenic cancers. The impact of dual ACC1/ACC2 inhibition was investigated using a dual ACC1/ACC2 inhibitor as well as dual siRNA knock down on the cellular viability and metabolism of two glioblastoma multiform cancer cell lines, U87 and a more aggressive form, U87 EGFRvIII. We first demonstrated that while ACCi inhibited DNL in both cell lines, ACCi preferentially blunted the U87 EGFRvIII cellular proliferation capacity. Metabolically, chronic treatment with ACCi significantly upregulated U87 EGFRvIII cellular respiration and extracellular acidification rate, a marker of glycolytic activity, but impaired mitochondrial health by reducing maximal respiration and decreasing mitochondrial ATP production efficiency. Moreover, ACCi treatment altered the cellular lipids content and increased apoptotic caspase activity in U87 EGFRvIII cells. Collectively these data indicate that ACC inhibition, by reducing DNL and increasing cellular metabolic rate, may have therapeutic utility for the suppression of lipogenic tumor growth and warrants further investigation.</p></div

    Inhibition of ACC leads to decreased de novo lipogenesis in both U87 and U87 EGFRvIII but reduced the number of U87 EGFRvIII cells to a greater extent.

    No full text
    <p>(A) mRNA expression of ACC1 (left panel) and ACC2 (right panel) in both U87 and U87 EGFRvIII cells after treatment with a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2) or scrambled control siRNAs as assessed by qPCR. Data are normalized to housekeeping genes expression. Each data point represents mean +/- sem, n = 3. *p<0.05, **p<0.01, *** p<0.001. (B) U87 and U87 EGFRvIII cells were treated with a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2) or scrambled control siRNAs for 144 hours. Extracted proteins were assessed by western blot (top panel). ACC1 protein (lower left panel) and ACC2 protein (lower right panel) were quantified by measurement of band intensity volume normalized to α-tubulin loading control. N = 3 experiments with representative blots shown. **** p<0.0001. (C) Inhibition of de novo lipogenesis in both U87 and U87 EGFRvIII cells as assessed by measurement of <sup>14</sup>C-acetate incorporation into neutral lipids after treatment with a dose range of ACCi or DMSO control. Data are expressed as percent (%) of DMSO control. Each data point represents mean +/- sem, n = 3. (D) Total cellular proteins were assessed after 72 hours treatment with either a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2), scrambled control siRNAs, 30 μM ACCi or DMSO control. Data are represented as fold change from transfection reagent only control (Control) or DMSO control. Each data point represents mean +/- sem, n = 3. *** p<0.001, **** p<0.0001. (E) Total cellular proteins were assessed after 144 hours treatment with either a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2), scrambled control siRNAs, 30 μM ACCi or DMSO control. Data are represented as fold change from either transfection reagent only control (Control) or DMSO control. Each data point represents mean +/- sem, n = 3. *** p<0.001, **** p<0.0001.</p

    Inhibition of ACC affects lipids content, cell viability and triggers U87 and U87 EGFRvIII cell death.

    No full text
    <p>(A) Essential fatty acid (C18:2), left panel, and de novo fatty acids (C16:0, C16:1 and C18:0), right panel, in U87 and U87 EGFRvIII cells treated with 30 μM ACCi or DMSO control for 144 hours as assessed by UPLC MS/MS. Data are represented as nmol/mg protein. Each data point represents mean +/- sem, n = 3. *p<0.05. (B) Effect of 144 hours treatment with 30 μM ACCi (patterned bars) or DMSO control (solid bars) on total cellular lipid species in U87 (top panel) and U87 EGFRvIII (lower panel) cells as assessed by UPLC-MS/MS. Data are expressed as nmol/mg protein. Each data point represents mean +/- sem, n = 3. *p<0.05, **p<0.01. (C) Cellular cytotoxicity after 144 hours treatment with either 30 μM ACCi, DMSO control, a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2) or scrambled siRNA control on U87 and U87 EGFRvIII as assessed by measurement of lactate dehydrogenase release into the assay media. Control samples were treated with transfection reagent only. Media samples were not treated with transfection reagent or DMSO. Data are represented as absorbance units per milligram of total protein per well. Each data point represents mean +/- sem, n = 3. *p<0.05, ****p<0.0001. (D) Cellular caspase activity after 144 hours treatment with either 30 μM ACCi, DMSO control, a combination of siRNAs targeted to ACC1 and ACC2 (siACC1/2) or scrambled siRNA control on U87 and U87 EGFRvIII cells as assessed by measurement of caspase 3/7 activity. Control samples were treated with transfection reagent only. Media samples were not treated with transfection reagent or DMSO. Data are represented as luminescence per milligram of total protein per well. Each data point represents mean +/- sem, n = 3. *p<0.05, **p<0.01, ****p<0.0001.</p

    Inhibition of ACC alters the metabolic response of U87 and U87 EGFRvIII cells to fatty acids and glucose.

    No full text
    <p>(A) Oxygen Consumption Rate (OCR) and Extracellular Acidification Rate (ECAR) values for U87 cells after treatment with 30 μM ACCi or DMSO control for 72 hours followed by acute treatment with BSA or 100 μM palmitate and 10 mM glucose as assessed by Seahorse XF96e system. Data are expressed as pmol/min/mg protein (OCR) and mpH/min/mg protein (ECAR). Each data point represents mean +/- sem, n = 3. * p<0.05, ****p<0.0001. (B) Oxygen Consumption Rate (OCR) and Extracellular Acidification Rate (ECAR) values for U87 EGFRvIII cells after treatment with 30 μM ACCi or DMSO control for 72 hours followed by acute treatment with BSA or 100 μM palmitate and 10 mM glucose as assessed by Seahorse XF96e system. Data are expressed as pmol/min/mg protein (OCR) and mpH/min/mg protein (ECAR). Each data point represents mean +/- sem, n = 3. * p<0.05, ****p<0.0001. (C) Experimental timeline and media compositions for Figs 4A and 4B.</p

    U87 EGFRvIII cells display higher proliferation, de novo lipogenesis and metabolic activity.

    No full text
    <p>(A) U87 EGFRvIII cells have reduced mRNA expression of WT EGFR compared to U87 cells (upper panel) and higher total EGFR including the mutated EGFRvIII expression (lower panel) as assessed by qPCR. Data are normalized to the expression of housekeeping genes. Each data point represents mean +/- sem, n = 3–4. * p<0.05, *** p = 0.0003. (B) U87 and U87 EGFRvIII cellular proliferation rates as assessed by luciferase measurement. Data are expressed as fold change from time 0 hours. Each data point represents mean +/- sem, n = 3. * p<0.05, **** p<0.0001. (C) Basal de novo lipogenesis of U87 and U87 EGFRvIII cells as assessed by measurement of the incorporation of <sup>14</sup>C-acetate into neutral lipids. Data are expressed as <sup>14</sup>C counts, DPM. Each data point represents mean +/- sem, n = 3. **** p<0.0001. (D) Total triacylglycerides and diacylglycerides content of U87 and U87 EGFRvIII cells at basal as assessed by UPLC-MS/MS measurement. Data are expressed as nmol/mg protein. Each data point represents mean +/- sem, n = 3. (E) Basal OCR and ECAR measurements from U87 and U87 EGFRvIII cells as assessed using Seahorse XF96e system. Data are expressed as pmol/min/mg protein (OCR) and mpH/min/mg protein (ECAR). Each data point represents mean +/- sem, n = 5. ****p<0.0001. (F) Bioenergetics profile of U87 and U87 EGFRvIII as assessed using Seahorse XF96e system. Injections A, B and C are oligomycin, FCCP and a mixture of rotenone and antimycin A respectively (top panel). Data are represented as % of baseline respiration, fold change from basal respiration and % of basal respiration. Each data point represents mean +/- sem, n = 5. *p<0.05, **p<0.01. (G) Media compositions for Fig 1E and 1F. Proliferation refers to media used for cell maintenance and growth before the bioenergetics experiments.</p

    FGF21 does not require adipocyte AMP-activated protein kinase (AMPK) or the phosphorylation of acetyl-CoA carboxylase (ACC) to mediate improvements in whole-body glucose homeostasis

    No full text
    Objective: Fibroblast growth factor 21 (FGF21) shows great potential for the treatment of obesity and type 2 diabetes, as its long-acting analogue reduces body weight and improves lipid profiles of participants in clinical studies; however, the intracellular mechanisms mediating these effects are poorly understood. AMP-activated protein kinase (AMPK) is an important energy sensor of the cell and a molecular target for anti-diabetic medications. This work examined the role of AMPK in mediating the glucose and lipid-lowering effects of FGF21. Methods: Inducible adipocyte AMPK β1β2 knockout mice (iβ1β2AKO) and littermate controls were fed a high fat diet (HFD) and treated with native FGF21 or saline for two weeks. Additionally, HFD-fed mice with knock-in mutations on the AMPK phosphorylation sites of acetyl-CoA carboxylase (ACC)1 and ACC2 (DKI mice) along with wild-type (WT) controls received long-acting FGF21 for two weeks. Results: Consistent with previous studies, FGF21 treatment significantly reduced body weight, adiposity, and liver lipids in HFD fed mice. To add, FGF21 improved circulating lipids, glycemic control, and insulin sensitivity. These effects were independent of adipocyte AMPK and were not associated with changes in browning of white (WAT) and brown adipose tissue (BAT). Lastly, we assessed whether FGF21 exerted its effects through the AMPK/ACC axis, which is critical in the therapeutic benefits of the anti-diabetic medication metformin. ACC DKI mice had improved glucose and insulin tolerance and a reduction in body weight, body fat and hepatic steatosis similar to WT mice in response to FGF21 administration. Conclusions: These data illustrate that the metabolic improvements upon FGF21 administration are independent of adipocyte AMPK, and do not require the inhibitory action of AMPK on ACC. This is in contrast to the anti-diabetic medication metformin and suggests that the treatment of obesity and diabetes with the combination of FGF21 and AMPK activators merits consideration. Keywords: FGF21, AMPK, ACC, Adipocyte, Brown fat, Obesity, Diabete
    corecore