14 research outputs found

    Super-mini percutaneous nephrolithotomy (SMP) vs retrograde intrarenal surgery (RIRS) in the management of renal calculi <= 2 cm: A propensity matched study

    Get PDF
    Objective: To compare the effectiveness and safety of Super-Mini PCNL (SMP) and Retrograde Intrarenal Surgery (RIRS) in the management of renal calculi ≤ 2 cm. Patients and methods: A prospective, inter-institutional, observational study of patients presenting with renal calculi ≤ 2 cm. Patients underwent either SMP (Group 1) or RIRS (Group 2) and were performed by 2 experienced high-volume surgeons. Results: Between September 2018 and April 2019, 593 patients underwent PCNL and 239 patients had RIRS in two tertiary centers. Among them, 149 patients were included for the final analysis after propensity-score matching out of which 75 patients underwent SMP in one center and 74 patients underwent RIRS in the other. The stone-free rate (SFR) was statistically significantly higher in Group 1 on POD-1 (98.66% vs. 89.19%; p = 0.015), and was still higher in Group 1 on POD-30 (98.66% vs. 93.24%, p = 0.092) SFR on both POD-1 and POD-30 for lower pole calculi was higher in Group 1 (100 vs. 82.61%, p = 0.047 and 100 vs 92.61% p = 0.171). The mean (SD) operative time was significantly shorter in Group 1 at 36.43 min (14.07) vs 51.15 (17.95) mins (p < 0.0001). The mean hemoglobin drop was significantly less in Group 1 (0.31 vs 0.53 gm%; p = 0.020). There were more Clavien–Dindo complications in Group 2 (p = 0.021). The mean VAS pain score was significantly less in Group 2 at 6 and 12 h postoperatively (2.52 vs 3.67, 1.85 vs 2.40, respectively: p < 0.0001), whereas the mean VAS pain score was significantly less in Group 1 at 24 h postoperatively (0.31 vs 1.01, p < 0.0001). The mean hospital stay was significantly shorter in Group 1 (28.37 vs 45.70 h; p < 0.0001). Conclusion: SMP has significantly lower operative times, complication rates, shorter hospital stay, with higher stone-free rates compared to RIRS. SMP is associated with more early post-operative pain though.Manipal Academy of Higher Education, Manipa

    Landscapes of Urbanization and De-Urbanization: A Large-Scale Approach to Investigating the Indus Civilization's Settlement Distributions in Northwest India.

    Get PDF
    Survey data play a fundamental role in studies of social complexity. Integrating the results from multiple projects into large-scale analyses encourages the reconsideration of existing interpretations. This approach is essential to understanding changes in the Indus Civilization's settlement distributions (ca. 2600-1600 b.c.), which shift from numerous small-scale settlements and a small number of larger urban centers to a de-nucleated pattern of settlement. This paper examines the interpretation that northwest India's settlement density increased as Indus cities declined by developing an integrated site location database and using this pilot database to conduct large-scale geographical information systems (GIS) analyses. It finds that settlement density in northwestern India may have increased in particular areas after ca. 1900 b.c., and that the resulting landscape of de-urbanization may have emerged at the expense of other processes. Investigating the Indus Civilization's landscapes has the potential to reveal broader dynamics of social complexity across extensive and varied environments.ER

    Flow cytometric analysis of vaccine-specific and bystander CD4<sup>+</sup> T cell responses to TT recall vaccination detected by combined CD40L and cytokine intracellular staining.

    No full text
    <p>After a short term (6h) <i>in vitro</i> culture in the absence (CTRL) or in the presence of either TT (10μg/ml), PPD (15μg/ml) or <i>C</i>.<i>Alb</i> (10μg/ml), PBMNC were first stained for surface CD3 and CD4, then permeabilized and stained intracellularly with fluorescent antibodies specific for CD40L and the cytokines IL-2 and IFN-γ. (A) Data from one of the individuals (subject 1) show the gating strategy (top three dot plots) and the detection of vaccine-specific (TT) and bystander (PPD and <i>C</i>.<i>Alb</i>) CD4<sup>+</sup> T cell responses before (pre-vaccination) and one week after (week 1) a booster injection of TT. Percentages indicate the frequency of positive events within the CD3<sup>+</sup>CD4<sup>+</sup> lymphocyte population. (B) Kinetics of vaccine-specific and bystander CD4<sup>+</sup> T cell responses detected by intracellular CD40L and cytokine staining in the same individual who received a booster vaccination (Wk 0) with TT. Data indicate the frequency of positive cells within the CD3<sup>+</sup>CD4<sup>+</sup> population, obtained from the antigen-stimulated samples after subtracting the frequency of events in the control cultures. Responses were considered positive if they met the criteria described in Materials and Methods. The dotted line shows the cut off value of 0.01%.</p

    Analyis of activation (CD38, HLA-DR) and proliferation (Ki-67) markers on vaccine-specific (TT) and bystander (PPD and <i>C</i>.<i>Alb</i>) CD4<sup>+</sup>CD40L<sup>+</sup> T cells.

    No full text
    <p>After a short term (6h) <i>in vitro</i> culture in the absence (control) or in the presence of either TT (10μg/ml), PPD (15μg/ml) or <i>C</i>.<i>Alb</i> (10μg/ml), PBMNC were first stained for surface CD3, CD4, CD38 and HLA-DR, then permeabilized and stained intracellularly for CD40L and Ki-67. (A) Data indicate the percentage of CD38, HLA-DR, Ki-67 single positive and CD38<sup>+</sup>Ki-67<sup>+</sup> double positive cells within the CD3<sup>+</sup>CD4<sup>+</sup>CD40L<sup>+</sup> population in five healthy subjects one week after receiving a booster vaccination with TT. Median values are indicated by a bar. Significance of the difference between vaccine specific and bystander populations are indicated (NS—non-significant, * = P<0.01, ** = P<0.001), as analysed using non-parametric Mann-Whitney U-test. (B) Representative dot plots from subject 5, showing how at week 1 activated and proliferating (CD38<sup>+</sup>Ki-67<sup>+</sup>) cells are only found among the TT-specific cells, but not among the bystander PPD- and <i>C</i>.<i>Alb</i>-specific cells. Percentages of positive cells in the CD3<sup>+</sup>CD4<sup>+</sup>CD40L<sup>+</sup> gated population are indicated in each quadrant. Gates in dot plots were set using the appropriate isotype-matched controls. (C) Analysis of expression (%) of CD38, HLA-DR, Ki-67 single positive and CD38<sup>+</sup>Ki-67<sup>+</sup> double positive cells within the TT- and PPD-specific CD4<sup>+</sup>CD40L<sup>+</sup> T cell populations before (Wk 0) and at various time points after TT booster vaccination. Data indicate the mean ± standard deviation calculated from the individuals showing detectable responses. P-values indicate the significant changes in expression over baseline for all time points, calculated using paired T-test and confirmed using Friedman’s test; non-significant changes are unlabeled. At week 0, responses to TT and PPD were detected in subjects 4 and 5; at week 2, responses to PPD were detected in subjects 2 and 4; for all the remaining time points, responses to TT and PPD were detected in all subjects.</p

    Percentages (A) and absolute numbers (B) of relevant immune cell populations before (Wk 0) and up to 8 weeks after TT booster vaccination.

    No full text
    <p>(A) Percentages of CD3<sup>+</sup>, CD3<sup>+</sup>CD4<sup>+</sup>, CD3<sup>+</sup>CD8<sup>+</sup> and CD3<sup>+</sup>CD4<sup>-</sup>CD8<sup>-</sup> cells were determined on viable PBMNC prior to cell culture by flow cytometry, gating on singlets first, and then on the lymphocyte population. A minimum of 100000 events were acquired. Combined staining for CD45RA and CCR7 allowed discrimination of Naïve (CD45RA<sup>+</sup> CCR7<sup>+</sup>), Memory (CD45RA<sup>-</sup>), central memory, T<sub>CM</sub> (CD45RA<sup>-</sup> CCR7<sup>+</sup>) and effector memory, T<sub>EM</sub> (CD45RA<sup>-</sup> CCR7<sup>-</sup>) CD3<sup>+</sup>CD4<sup>+</sup> cells. (B) The absolute number of cells was calculated from the blood lymphocyte counts available for subjects 1, 2 and 3 (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0136717#pone.0136717.t001" target="_blank">Table 1</a>), and the phenotypic data.</p

    Distribution of vaccine-specific (TT) and bystander (PPD, <i>C</i>.<i>Alb</i>) CD4<sup>+</sup> CD40L<sup>+</sup> and cytokine positive T cells according to the expression of CD45RA and CCR7.

    No full text
    <p>After a short term (6h) <i>in vitro</i> culture in the absence (control) or in the presence of either TT (10μg/ml), PPD (15μg/ml) or <i>C</i>.<i>Alb</i> (10μg/ml), antigen-specific CD4<sup>+</sup> T cells, identified by CD40L expression and cytokine (IL-2, IFN-γ) production (see <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0136717#pone.0136717.g002" target="_blank">Fig 2A</a>), were stained with CD45RA and CCR7 and the percentages of naïve (T<sub>N</sub>, CD45RA<sup>+</sup>CCR7<sup>+</sup>), central memory (T<sub>CM</sub>, CD45RA<sup>-</sup>CCR7<sup>+</sup>), effector memory (T<sub>EM</sub>, CD45RA<sup>-</sup>CCR7<sup>-</sup>) and terminally differentiated (T<sub>TD</sub>, CD45RA<sup>+</sup>CCR7<sup>-</sup>) were calculated among the CD4<sup>+</sup>CD40L<sup>+</sup>, CD4<sup>+</sup>CD40L<sup>+</sup>IL-2<sup>+</sup> and CD4<sup>+</sup>CD40L<sup>+</sup>IFN-γ<sup>+</sup> cells. (A) Representative data from subject 1 showing the phenotype of vaccine-specific (TT) and bystander (PPD, <i>C</i>.<i>Alb</i>) CD4<sup>+</sup> T cells one week after booster vaccination with TT. The percentages of events in each quadrant are indicated. Gates in dot plots were set using the appropriate isotype-matched controls. Responding cells are CD45RA<sup>-</sup> memory type cells, but whilst the TT-specific are in their vast majority T<sub>EM</sub>, bystander (PPD and <i>C</i>.<i>Alb</i>-specific) cells are mainly T<sub>CM</sub>. N.D. Responses not detected. (B) Cumulative data (mean percentages) from five subjects, one week after TT vaccination showing the differential phenotype between vaccine-specific and bystander responses. The numbers on the bars indicate the mean percentages ± SD of T<sub>EM</sub> cells among the TT-specific, and of T<sub>CM</sub> cells among the PPD- and <i>C</i>.<i>Alb</i>-specific populations, respectively. <i>C</i>.<i>Alb</i>-specific CD40L<sup>+</sup> cells were detected only in subjects 1, 4 and 5; <i>C</i>.<i>Alb</i>-specific CD40L<sup>+</sup>IL-2<sup>+</sup> cells were detected only in subject 1; <i>C</i>.<i>Alb</i>-specific CD40L<sup>+</sup>IFN-γ<sup>+</sup> cells were not detected in any subjects (N.D). (C) Kinetics of T<sub>CM</sub> to T<sub>EM</sub> ratio in vaccine-specific (TT) and bystander (PPD) CD4<sup>+</sup> CD40L<sup>+</sup> T cells in subjects 4 before TT vaccination (Wk 0) and during follow up. Black asterisks indicate time points where TT-specific responses were not detectable.</p

    Vaccination expands antigen-specific cd4+ memory T cells and mobilizes bystander central memory T cells

    No full text
    CD4+ T helper memory (Thmem) cells influence both natural and vaccine-boosted immunity, but mechanisms for their maintenance remain unclear. Pro-survival signals from the common gamma-chain cytokines, in particular IL-7, appear important. Previously we showed in healthy volunteers that a booster vaccination with tetanus toxoid (TT) expanded peripheral blood TT-specific Thmem cells as expected, but was accompanied by parallel increase of Thmem cells specific for two unrelated and non cross-reactive common recall antigens. Here, in a new cohort of healthy human subjects, we compare blood vaccine-specific and bystander Thmem cells in terms of differentiation stage, function, activation and proliferative status. Both responses peaked 1 week post-vaccination. Vaccine-specific cytokine-producing Thmem cells were predominantly effector memory, whereas bystander cells were mainly of central memory phenotype. Importantly, TT-specific Thmem cells were activated (CD38High HLA-DR+), cycling or recently divided (Ki-67+), and apparently vulnerable to death (IL-7R?Low and Bcl-2 Low). In contrast, bystander Thmem cells were resting (CD38Low HLA-DR- Ki-67-) with high expression of IL-7R? and Bcl-2. These findings allow a clear distinction between vaccine-specific and bystander Thmem cells, suggesting the latter do not derive from recent proliferation but from cells mobilized from as yet undefined reservoirs. Furthermore, they reveal the interdependent dynamics of specific and bystander T-cell responses which will inform assessments of responses to vaccines

    Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy

    Get PDF
    21siBACKGROUND: Vascular endothelial growth factor (VEGF)-B activates cytoprotective/antiapoptotic and minimally angiogenic mechanisms via VEGF receptors. Therefore, VEGF-B might be an ideal candidate for the treatment of dilated cardiomyopathy, which displays modest microvascular rarefaction and increased rate of apoptosis. OBJECTIVES: This study evaluated VEGF-B gene therapy in a canine model of tachypacing-induced dilated cardiomyopathy. METHODS: Chronically instrumented dogs underwent cardiac tachypacing for 28 days. Adeno-associated virus serotype 9 viral vectors carrying VEGF-B167 genes were infused intracoronarily at the beginning of the pacing protocol or during compensated heart failure. Moreover, we tested a novel VEGF-B167 transgene controlled by the atrial natriuretic factor promoter. RESULTS: Compared with control subjects, VEGF-B167 markedly preserved diastolic and contractile function and attenuated ventricular chamber remodeling, halting the progression from compensated to decompensated heart failure. Atrial natriuretic factor-VEGF-B167 expression was low in normally functioning hearts and stimulated by cardiac pacing; it thus functioned as an ideal therapeutic transgene, active only under pathological conditions. CONCLUSIONS: Our results, obtained with a standard technique of interventional cardiology in a clinically relevant animal model, support VEGF-B167 gene transfer as an affordable and effective new therapy for nonischemic heart failure.reservedmixedWoitek, Felix; Zentilin, Lorena; Hoffman, Nicholas E.; Powers, Jeffery C.; Ottiger, Isabel; Parikh, Suraj; Kulczycki, Anna M.; Hurst, Marykathryn; Ring, Nadja; Wang, Tao; Shaikh, Farah; Gross, Polina; Singh, Harinder; Kolpakov, Mikhail A.; Linke, Axel; Houser, Steven R.; Rizzo, Victor; Sabri, Abdelkarim; Madesh, Muniswamy; Giacca, Mauro; Recchia, Fabio A.Woitek, Felix; Zentilin, Lorena; Hoffman, Nicholas E.; Powers, Jeffery C.; Ottiger, Isabel; Parikh, Suraj; Kulczycki, Anna M.; Hurst, Marykathryn; Ring, Nadja; Wang, Tao; Shaikh, Farah; Gross, Polina; Singh, Harinder; Kolpakov, Mikhail A.; Linke, Axel; Houser, Steven R.; Rizzo, Victor; Sabri, Abdelkarim; Madesh, Muniswamy; Giacca, Mauro; Recchia, Fabio A
    corecore