17 research outputs found

    Serum level of galectin-3 in human bladder cancer

    Get PDF
    We examine serum level of galectin-3 in patients with bladder cancer. We used serum samples of 67 patients with urological diseases and classified these patients into bladder cancer group (n=43) and control group (n=24). Galectin-3 concentration was measured by ELISA (Human Galectin-3 Assay Kit, IBL). And we selected the patient with high serum galectin-3 concentration (Urothelial Carcinoma, G3, pT3a pN0M0), we performed immunohistochemical staining with the VECTASTAIN ABC (Avidin Biotinylated enzyme Complex) system. Median value of serum galectin-3 concentration was 1068 pg/ml (range 551-2028) in the cancer group vs 584 pg/ml (range 259-1262) in controls. Serum galectin-3 concentration of the bladder cancer patients was statistically higher than that of controls (p<0.0005). There was no apparent correlation in serum galectin-3 concentration with the clinico-pathological features such as stage and grade. Higher expression of galectin-3 was observed in bladder cancer tissue than in normal bladder tissue. We suggest the measurement of serum galectin-3 is useful for diagnosis of bladder cancer

    Effectiveness of Subcutaneous Growth Hormone in HIV-1 Patients with Moderate to Severe Facial Lipoatrophy

    Get PDF
    Objective: To evaluate effect of recombinant human growth hormone (rhGH) among HIV-infected adults with moderate to severe facial lipoatrophy as a side effect of long-term antiretroviral treatment. Design: A prospective open-label study Methods: Twenty-five HIV-1 patients with moderate to severe facial lipoatrophy who had been on antiretroviral treatment for more than 18 months were enrolled. rhGH (5 mg) was given every other day for 6 months. After treatment was completed, the participants were followed up for 6 months. Facial lipoatrophy was evaluated by computed tomography at months 0, 3, 6 and 12. Results: Nearly all participants (24 of 25) completed the study. The sum of bilateral soft tissue thickness at the level of zygomatics at months 0, 3, 6, 12 were 7.23, 8.59, 8.35, 8.60 mm, respectively. There was significant improvement from baseline in month 3 (p=0.009) and month 12 (p=0.021). In the 6 months of follow-up, the soft tissue showed no significant decrease. Several side effects including diarrhea, arthralgia, myalgia, mastalgia and hand numbness were seen, which were self-limited and transient. Conclusion: rhGH is effective and relatively safe for moderate to severe facial lipoatrophy. Its effect was sustained at least for 6 months after the cessation of rhGH

    Slow Turnover of HIV-1 Receptors on Quiescent CD4<sup>+</sup> T Cells Causes Prolonged Surface Retention of gp120 Immune Complexes <i>In Vivo</i>

    No full text
    <div><p>Peripheral blood CD4<sup>+</sup> T cells in HIV-1<sup>+</sup> patients are coated with Ig. However, the causes and consequences of the presence of Ig<sup>+</sup> CD4<sup>+</sup> T cells remain unknown. Previous studies have demonstrated the rapid turnover of viral receptors (VRs) on lymphoma and tumor cells. The present study investigates the turnover of VRs on peripheral quiescent CD4<sup>+</sup> T cells (qCD4s), which are the most abundant peripheral blood CD4<sup>+</sup> T cells. Utilizing pharmacological and immunological approaches, we found that the turnover of VRs on qCD4s is extremely slow. As a result, exposure to gp120 or HIV-1 virions <i>in vitro</i> causes gp120 to remain on the surface for a long period of time. It requires approximately three days for cell-bound gp120 on the surface to be reduced by 50%. In the presence of patient serum, gp120 forms surface immune complexes (ICs) that are also retained for a long time. Indeed, when examining the percentages of Ig<sup>+</sup> CD4<sup>+</sup> T cells at different stages of HIV-1 infection, approximately 70% of peripheral resting CD4<sup>+</sup> T cells (rCD4s) were coated with surface VRs bound to slow-turnover gp120-Ig. The levels of circulating ICs in patient serum were insufficient to form surface ICs on qCD4s, suggesting that surface ICs on qCD4s require much higher concentrations of HIV-1 exposure such as might be found in lymph nodes. In the presence of macrophages, Ig<sup>+</sup> CD4<sup>+</sup> T cells generated <i>in vitro</i> or directly isolated from HIV-1<sup>+</sup> patients were ultimately phagocytosed. Similarly, the frequencies and percentages of Ig<sup>+</sup> rCD4s were significantly increased in an HIV-1<sup>+</sup> patient after splenectomy, indicating that Ig<sup>+</sup> rCD4s might be removed from circulation and that non-neutralizing anti-envelope antibodies could play a detrimental role in HIV-1 pathogenesis. These findings provide novel insights for vaccine development and a rationale for using Ig<sup>+</sup> rCD4 levels as an independent clinical marker.</p></div

    Schematic figure summarizes the causes and consequences of sIC<sup>+</sup> rCD4s.

    No full text
    <p>rCD4s continuously travel between the blood stream and LNs over a period of approximately 1 d. Because a large proportion of HIV-1 is produced in the LNs, the target T cells that migrate to the LNs are exposed to high concentrations of HIV-1, gp120, or ICs as well as anti-env Abs. Prolonged retention of gp120-VR complexes on rCD4s causes the retention of sICs in a manner that reflects the levels of HIV-1 exposure in the LNs. sIC<sup>+</sup> rCD4s are removed from circulation through ADCP or ADCC by macrophages or NK cells, respectively. The sIC<sup>+</sup> rCD4s that are not removed from circulation remigrate to the LNs to be exposed to a high concentration of gp120/HIV-1. The percentages and amounts of sICs on rCD4s in the blood reflect a balance of five factors, namely, the levels of virus production in lymphoid tissues, the levels of anti-env Abs, the turnover dynamics of sICs on rCD4s, the duration of repeated exposure by continuous migration to the lymphoid organs, and the levels of immunological elimination of sIC<sup>+</sup> rCD4s.</p

    HIV-1/gp120 remains on the surface of qCD4s for a long period of time due to slow VR turnover.

    No full text
    <p>(<b>a, b</b>) Time course of surface VR expression (<b>a</b>) and representative FACS of CXCR4/CCR5 expression on qCD4s (<b>b</b>) following a variety of activation stimuli. (<b>c</b>) The effect of BFA (10 ”g/ml), cycloheximide (50 ”g/ml) and ActD (20 ”g/ml) on the surface expression of CXCR4 (left) and CD4 (right) on qCD4s. (<b>d, e</b>) Confocal micrographs of CD4, CXCR4, and gp120 in qCD4s that were exposed or not exposed to the indicated strain of gp120 or HIV-1 before (<b>d</b>) or after (<b>e</b>) 16 h of anti-CD3 Ab exposure. qCD4s with (<b>e</b>) or without (<b>d</b>) permeabilization were stained with anti-CD4 goat polyclonal Abs (Cy3, red), anti-CXCR4 mouse mAbs (Qdot 655, blue), and anti-gp120 rabbit antiserum (Cy2, green). (<b>f</b>) Time course of cell-bound gp120, sICs (left panel), or surface CD4 expression (right panel) on gp120<sub>IIIB</sub>-pulsed or untreated qCD4s. The gp120<sub>IIIB</sub>-pulsed qCD4s were further incubated with HIV-1<sup>+</sup> Pt serum (Pt-serum) to form sICs or untreated and cultured in the absence or presence of anti-CD3 Abs. The effect of T22 pre-exposure on cell-bound gp120<sub>IIIB</sub> in anti-CD3 Ab stimulation (αCD3+T22) was also examined. (<b>g</b>) Time course of cell-bound gp120, sICs, or surface CD4 expression on HIV-1<sub>Lai</sub> (Lai)<b>-</b> (<b>left</b>), HIV-1<sub>BaL</sub> (BaL)<b>-</b>, or clinical isolate (Clinical)<b>-</b> (<b>right</b>) pulsed qCD4s. HIV-1-pulsed qCD4s were further incubated with HIV-1<sup>+</sup> Pt serum (<b>Pt-serum</b>) to form sICs or untreated and cultured in the absence or presence of anti-CD3 Abs. The effect of enfuvirtide (<b>Enf</b>) exposure was also examined. (<b>h</b>) The amount and location of cell-bound gp120 in gp120<sub>IIIB</sub>-pulsed qCD4s that were cultured in the absence or presence of anti-CD3 Abs were assessed by confocal microscopy (<b>upper</b>) or by western blotting (<b>bottom</b>). The lower numbers indicate the value by densitometry. (<b>i</b>) Time course of chemotaxis inhibition on gp120<sub>IIIB</sub>-, gp120<sub>BaL</sub>- (<b>upper</b>), or HIV-1<sub>Lai</sub>- (<b>bottom</b>) pulsed qCD4s. Chemotaxis of gp120- or HIV-1-pulsed or non-pulsed qCD4s toward the indicated chemokines was evaluated using a transwell assay. Bars, SD. The data here are representative of at least three independent experiments.</p

    rCD4s from HIV-1<sup>+</sup> Pts are coated with gp120.

    No full text
    <p>(<b>a</b>) Representative FACS data from rCD4s purified from healthy controls (H1, H2) or chronic asymptomatic patients (Pt-f2r1, Pt-f2r2) stained with Leu3a and CD4v4 (numbers in FACS plots indicate percentages of MFIs of Leu3a/CD4v4). (<b>b</b>) Summary of results of percentages of MFIs of Leu3a/CD4v4 in purified rCD4s from healthy controls (H) and chronic asymptomatic patients (C; CD4 counts: 420±84.6 (± SD); IgG<sup>+</sup> rCD4s: 75.5±12.6% (± SD)). (<b>c, d</b>) Detection of cell-bound gp120 on rCD4s in a patient with low anti-gp120 Ab levels. (<b>c</b>) Western blot test results for the HIV-1<sup>+</sup> Pt (NDA-01) at initial admission and three months after. N, negative control; Pt, patient serum; Po, positive control. HIV-1 infection was defined as detectable amounts of plasma HIV-1 RNA (1.5×10<sup>5</sup> copies/ml at initial admission), a positive antibody test (HIV1/2 ELISA), and low CD4<sup>+</sup> T cell counts (38 cells/”l). Plasma HIV-1 env and gag region sequences revealed that the patient was infected with a clade B HIV-1. (<b>d</b>) FACS data from rCD4s stained with anti-IgG (upper left), anti-IgM (upper right), anti-gp120 (rabbit anti-gp120 antiserum) (lower left), or purified IgG from pooled serum from HIV-1<sup>+</sup> Pts (lower right).</p

    sICs on qCD4s trigger Fc-mediated effector systems.

    No full text
    <p>(<b>a–e</b>) Autologous macrophages phagocytose qCD4s with sICs. Orange-CMTMR-labeled macrophages (red) cocultured with CFSE-labeled autologous qCD4s (green) exposed to the indicated concentrations of gp120<sub>IIIB</sub>, HIV-1<sub>Lai</sub>, or medium followed by incubation with HI patient serum (Pt-serum), non-HI patient serum, or medium for 1 h before coculture. (<b>a–c</b>) Confocal micrographs of representative data (<b>a</b>) and summary of phagocytosis assays (<b>b, c</b>) shown as percentages of macrophages containing 1–2, 3–5, or >6 qCD4s. The numbers in (<b>a</b>) denote percentages of macrophages containing at least one qCD4; inset shows a macrophage containing >10 qCD4s. (<b>d</b>) Time course of TUNEL assay on sIC<sup>+</sup> qCD4s that were phagocytosed by macrophages. Confocal images of macrophages (arrowheads, red), TUNEL<sup>+</sup> (green) and cell nuclei (Topro-3, blue). The numbers indicate the percentage of TUNEL<sup>+</sup> phagocytosed qCD4s/total phagocytosed qCD4s. (<b>e</b>) Summary of the time course of phagocytosis assays. (<b>f</b>) Summary of apoptotic qCD4s in the NK cell-mediated ADCC assay. CFSE-labeled NK cells incubated with autologous qCD4s (2∶1), which were exposed to the indicated concentrations of gp120<sub>IIIB</sub>, gp120<sub>BaL</sub>, HIV-1<sub>Lai</sub>, HIV-1<sub>BaL</sub>, or medium. (G) Summary of effects of IL-2 (50 ng/ml) or IL-15 (20 ng/ml) treatment on NK cell-mediated ADCC. Bars, SD. The data presented here are representative of at least three independent experiments.</p
    corecore