23 research outputs found

    An approach to open virtual commissioning for component-based automation

    Get PDF
    Increasing market demands for highly customised products with shorter time-to-market and at lower prices are forcing manufacturing systems to be built and operated in a more efficient ways. In order to overcome some of the limitations in traditional methods of automation system engineering, this thesis focuses on the creation of a new approach to Virtual Commissioning (VC). In current VC approaches, virtual models are driven by pre-programmed PLC control software. These approaches are still time-consuming and heavily control expertise-reliant as the required programming and debugging activities are mainly performed by control engineers. Another current limitation is that virtual models validated during VC are difficult to reuse due to a lack of tool-independent data models. Therefore, in order to maximise the potential of VC, there is a need for new VC approaches and tools to address these limitations. The main contributions of this research are: (1) to develop a new approach and the related engineering tool functionality for directly deploying PLC control software based on component-based VC models and reusable components; and (2) to build tool-independent common data models for describing component-based virtual automation systems in order to enable data reusability. [Continues.

    Development of Low-Cost Porous Carbons through Alkali Activation of Crop Waste for CO<sub>2</sub> Capture

    No full text
    To achieve the ā€œdouble carbonā€ (carbon peak and carbon neutrality) target, low-cost CO2 capture at large CO2 emission points is of great importance, during which the development of low-cost CO2 sorbents will play a key role. Here, we chose peanut shells (P) from crop waste as the raw material and KOH and K2CO3 as activators to prepare porous carbons by a simple one-step activation method. Interestingly, the porous carbon showed a good adsorption capacity of 2.41 mmol/g for 15% CO2 when the mass ratio of K2CO3 to P and the activation time were only 0.5 and 0.5 h, respectively, and the adsorption capacity remained at 98.76% after 10 adsorptionā€“desorption cycle regenerations. The characterization results suggested that the activated peanut shell-based porous carbons were mainly microporous and partly mesoporous, and hydroxyl (Oā€“H), ether (Cā€“O), and pyrrolic nitrogen (N-5) functional groups that promoted CO2 adsorption were formed during activation. In conclusion, KOH- and K2CO3-activated P, especially K2CO3-activated P, showed good CO2 adsorption and regeneration performance. In addition, not only the use of a small amount of the activator but also the raw material of crop waste reduces the sorbent preparation costs and CO2 capture costs

    Trefoil Factor-3 (TFF3) Stimulates <i>De Novo</i> Angiogenesis in Mammary Carcinoma both Directly and Indirectly via IL-8/CXCR2

    No full text
    <div><p>Mammary carcinoma cells produce pro-angiogenic factors to stimulate angiogenesis and tumor growth. Trefoil factor-3 (TFF3) is an oncogene secreted from mammary carcinoma cells and associated with poor prognosis. Herein, we demonstrate that TFF3 produced in mammary carcinoma cells functions as a promoter of tumor angiogenesis. Forced expression of TFF3 in mammary carcinoma cells promoted proliferation, survival, invasion and <i>in vitro</i> tubule formation of human umbilical vein endothelial cells (HUVEC). MCF7-TFF3 cells with forced expression of TFF3 generated tumors with enhanced microvessel density as compared to tumors formed by vector control cells. Depletion of TFF3 in mammary carcinoma cells by siRNA concordantly decreased the angiogenic behavior of HUVEC. Forced expression of TFF3 in mammary carcinoma cells stimulated IL-8 transcription and subsequently enhanced IL-8 expression in both mammary carcinoma cells and HUVEC. Depletion of IL-8 in mammary carcinoma cells with forced expression of TFF3, or antibody inhibition of IL-8, partially abrogated mammary carcinoma cell TFF3-stimulated HUVEC angiogenic behavior <i>in vitro</i>, as did inhibition of the IL-8 receptor, CXCR2. Depletion of STAT3 by siRNA in MCF-7 cells with forced expression of TFF3 partially diminished the angiogenic capability of TFF3 on stimulation of cellular processes of HUVEC. Exogenous recombinant hTFF3 also directly promoted the angiogenic behavior of HUVEC. Hence, TFF3 is a potent angiogenic factor and functions as a promoter of <i>de novo</i> angiogenesis in mammary carcinoma, which may co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression.</p></div

    Mammary carcinoma cells with forced expression of TFF3 promoted tumor angiogenesis <i>in vivo</i>.

    No full text
    <p>(A) IHC analysis of CD31 and CD34 protein expressions in xenograft tumors formed by MCF-7 cells with forced expression of TFF3. (B) Microvessel density (CD31) was assessed by quantifying percentage area of CD31 labeled cells in xenografts formed by MCF-7 cells with forced expression of TFF3. (C) CD34 was assessed by quantifying percentage area of CD34 labeled cells in xenografts formed by MCF-7 cells with forced expression of TFF3. MCF-7 cells with empty vector (MCF7-Vec) used as control. **, <i>P</i> < <i>0</i>.<i>01</i>; ***, <i>P</i> < <i>0</i>.<i>001</i>; scale bar, 50 Ī¼m.</p

    Depletion of STAT3 in mammary carcinoma cells by siRNA partially diminished the ability of TFF3 to stimulate mammary carcinoma cell IL-8 promoter activity and IL-8 protein expression as well as HUVEC migration, invasion, and tubule formation <i>in vitro</i>.

    No full text
    <p>(A) IL-8 promoter reporter activity in MCF7-Vec and MCF7-TFF3 with depletion of STAT3 transiently transfected with an IL-8 promoter reporter vector (full length, -4800 to + 104 bp) and a pRL-CMV control reporter vector. Scrambled control siRNA was used as control. MCF7-Vec transiently transfected with scrambled control siRNA was as baseline. (B) ELISA analysis of IL-8 protein secreted to the medium by MCF7-Vec and MCF7-TFF3 transiently transfected with pcDNA vector containing STAT3 siRNA or control siRNA. *, <i>P < 0</i>.<i>05</i>; **, <i>P</i> < <i>0</i>.<i>01</i> as compared MCF7-Vec or MCF7-TFF3 transiently transfected with control siRNA. (C) HUVEC migration after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 transiently transfected with control siRNA and STAT3 siRNA. (D) HUVEC invasion after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 transiently transfected with control siRNA and STAT3 siRNA. (E) and (F) HUVEC tubule formation <i>in vitro</i> in the Matrigel after 12 hours co-culture with MCF-7 cells with forced expression of TFF3 transiently transfected with control siRNA and STAT3 siRNA in serum-free conditions. Total tubule length (E) and tubule number (F) was assessed. **, <i>P</i> < <i>0</i>.<i>01</i> as compared to MCF7-Vec or MCF7-TFF3 transiently transfected with control siRNA, respectively; Scrambled control siRNA was used as control; MCF7-Vec transiently transfected with scrambled control siRNA was a baseline.</p

    Depletion of TFF3 by siRNA in mammary carcinoma cells decreased angiogenic behavior of HUVEC.

    No full text
    <p><b>(</b>A) Semi-quantitative RT-PCR analysis of TFF3 mRNA level in MCF-7 cells with depletion of TFF3 (MCF7-siTFF3) and control siRNA vector cells (MCF7-siVec) after 24 hours transient transfection. (B) Western blot analysis of TFF3 protein in MCF-7 cells with depletion of TFF3 and control siRNA vector cells after 48 and 72 hours transient transfection. (C) Monolayer proliferation of HUVEC after co-culture with MCF-7 cells with depletion of TFF3 in 10% FBS conditions. (D) Monolayer proliferation of HUVEC after co-culture with MCF-7 cells with depletion of TFF3 in 0.2% FBS conditions. (E) HUVEC cell cycle progression after 24 hours co-culture with MCF-7 cells with depletion of TFF3 in serum-free and 10% FBS conditions. (F) HUVEC apoptotic cell death after 24 hours co-culture with MCF-7 cells with depletion of TFF3 in serum-free and 10% FBS conditions. (G) HUVEC migration after 24 hours co-culture with MCF-7 cells with depletion of TFF3 in serum free conditions. (H) HUVEC invasion after 24 hours co-culture with MCF-7 cells with depletion of TFF3 in serum free conditions. (I) and (J) HUVEC tubule formation <i>in vitro</i> in the Matrigel after 12 hours co-culture with MCF-7 cells with depletion of TFF3 in serum-free conditions. Total tubule length (I) and tubule number (J) were assessed after 12 hours incubation. (K) Representative light photomicrographs of HUVEC tubule formation <i>in vitro</i> in the Matrigel after 12 hours co-culture with MCF-7 cells with depletion of TFF3. MCF-7 cells with control siRNA vector (MCF7-siVec) was used as control. Ī²-ACTIN was used as input control in semi-quantitative RT-PCR and Western blot analyses. *, <i>P < 0</i>.<i>05</i>; **, <i>P</i> < <i>0</i>.<i>01</i>; ***, <i>P</i> < <i>0</i>.<i>001</i>; scale bar, 200 Ī¼m.</p

    Exogenous recombinant hTFF3 increased HUVEC monolayer proliferation, migration, invasion, and tubule formation <i>in vitro</i>.

    No full text
    <p>(A) HUVEC monolayer proliferation at different concentrations of recombinant hTFF3 in 10% FBS condition. (B) HUVEC monolayer proliferation at different concentrations of recombinant hTFF3 in 0.2% FBS conditions. HUVEC treated with different concentrations of recombinant hTFF3 (0.1, 1.0, 2.5, 5, 10 ng/mL of recombinant hTFF3) and BSA control (10 ng/mL). BSA was used as control. HUVEC treated with BSA control was a baseline. (C) HUVEC migration after 24 hours co-cultured with different concentration of recombinant hTFF3 or BSA control. (D) HUVEC invasion after 24 hours co-cultured with different concentration of recombinant hTFF3 or BSA control. (E) HUVEC tubule formation <i>in vitro</i>, in which HUVEC treated with different concentration of recombinant hTFF3 or BSA control were plated in the Matrigel. Total tubule length was assessed using ImageJ analysis software. (F) Representative light photomicrographs of HUVEC tubule formation <i>in vitro</i>, in which HUVEC treated with different concentrations of recombinant hTFF3 or BSA control. *, <i>P</i> < <i>0</i>.<i>05</i>; **, <i>P</i> < <i>0</i>.<i>01</i>; ***, <i>P</i> < <i>0</i>.<i>001</i> as compared with BSA control. Scale bar, 200 Ī¼m.</p

    Mammary carcinoma cells with forced expression of TFF3 increased angiogenic behavior of HUVEC.

    No full text
    <p>(A) Monolayer proliferation of HUVEC after co-culture with MCF-7 cells with forced expression of TFF3 in 10% FBS conditions. (B) Monolayer proliferation of HUVEC after co-culture with MCF-7 cells with forced expression of TFF3 in 0.2% FBS conditions. (C) HUVEC cell cycle progression after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 in serum-free (SF) and 10% FBS conditions. (D) HUVEC apoptotic cell death after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 in serum-free and 10% FBS conditions. (E) HUVEC migration after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 in serum-free conditions. (F) HUVEC invasion after 24 hours co-culture with MCF-7 cells with forced expression of TFF3 in serum-free conditions. (G) and (H) HUVEC tubule formation <i>in vitro</i> in the Matrigel after 12 hours co-culture with MCF-7 cells with forced expression of TFF3. Total tubule length (G) and total tubule number (H) were assessed. (I) Representative light photomicrographs of HUVEC tubule formation <i>in vitro</i> in the Matrigel after 12 hours co-culture with MCF-7 cells with forced expression of TFF3. MCF-7 cells with empty vector (MCF7-Vec) was used as control. Ī²-ACTIN was used as input control in semi-quantitative RT-PCR and Western blot analyses. *, <i>P < 0</i>.<i>05</i>; **, <i>P</i> < <i>0</i>.<i>01</i>; ***, <i>P</i> < <i>0</i>.<i>001</i>; scale bar, 200 Ī¼m.</p

    TFF3 enhanced IL-8 expression in mammary carcinoma cells and HUVEC.

    No full text
    <p>(A) IL-8 promoter reporter activity (full length, -4800 to + 104 bp) in MCF-7 cells with forced expression of TFF3 and control vector cells. MCF-7 cells with empty vector (MCF7-Vec) was used as control. (B) Semi-quantitative RT-PCR analysis of IL-8 mRNA level in MCF-7 with forced expression of TFF3 and control vector cells. (C) ELISA analysis of IL-8 protein secreted to the medium by MCF-7 cells with forced expression of TFF3 and control vector cells. (D) IHC analysis of IL-8 protein expression in xenograft tumors formed by MCF-7 cells with forced expression of TFF3. (E) Percentage of IL-8 labeled cells in xenograft tumors formed by MCF-7 cells with forced expression of TFF3 and control vector cells. (F) IL-8 promoter reporter activity (full length, -4800 to + 104 bp) in MCF-7 cells with depletion of TFF3 and control siRNA vector cells. MCF-7 cells with control siRNA vector (MCF7-siVec) was used as control. (G) Semi-quantitative RT-PCR analysis of IL-8 mRNA level in MCF-7 with depletion of TFF3 and control siRNA vector cells. (H) ELISA analysis of IL-8 protein secreted to the medium by MCF-7 cells with depletion of TFF3 and control siRNA vector cells. (I) Semi-quantitative RT-PCR analysis of IL-8 mRNA level in HUVEC co-cultured with MCF-7 cells with forced expression of TFF3 and control vector cells. (J) ELISA analysis of IL-8 protein secreted to the medium by HUVEC co-cultured MCF-7 with forced expression of TFF3 and control vector cells. Ī²-ACTIN was used as input control in semi-quantitative RT-PCR and Western blot analyses. **, <i>P</i> < <i>0</i>.<i>01</i>; ***, <i>P</i> < <i>0</i>.<i>001</i>; scale bar, 50 Ī¼m.</p

    TFF3 is a promoter of angiogenesis in mammary carcinoma.

    No full text
    <p>TFF3 secreted from mammary carcinoma cells indirectly stimulated angiogenic behavior of endothelial cells to promote angiogenesis in mammary carcinoma via an IL-8/CXCR2 axis. STAT3 is one transcription factor responsible for the increased expression of IL-8 by TFF3. TFF3 also promotes angiogenesis by direct functional effects on endothelial cellular processes promoting angiogenesis. TFF3 stimulates angiogenesis to co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression and dissemination.</p
    corecore