28 research outputs found

    Analysis of the Hepatic Functional Reserve, Portal Hypertension, and Prognosis of Patients With Human Immunodeficiency Virus/Hepatitis C Virus Coinfection Through Contaminated Blood Products in Japan

    Get PDF
    Background As the survival of human immunodeficiency virus (HIV)-infected individuals has improved due to the widespread use of antiretroviral therapy, the mortality rate due to hepatitis C virus (HCV)-related liver disease has increased in HIV/HCV-coinfected patients. Aim The aims of this study were to establish the appropriate therapeutic strategy for HIV/HCV-coinfected patients by evaluating the liver function, including the hepatic functional reserve and portal hypertension, and to investigate the prognosis of HIV/HCV-coinfected patients in Japan. Patients and Methods In addition to regular liver function tests, the hepatic functional reserve of 41 patients with HIV/HCV coinfection was evaluated using the indocyanine green retention rate and liver galactosyl serum albumin-scintigraphy. The data for 146 patients with HIV/HCV coinfection through blood products were extracted from 4 major HIV centers in Japan. In addition to liver function tests, the platelet counts (PLT) were evaluated as a marker of portal hypertension. Results In spite of the relatively preserved general liver function test results, approximately 40% of the HIV/HCV-coinfected patients had an impaired hepatic functional reserve. In addition, while the albumin and bilirubin levels were normal, the PLT was <150,000/μL in 17 patients. Compared with HCV mono-infected patients with a PLT <150,000/μL, the survival of HIV/HCV-coinfected patients was shorter (HCV, 5 years, 97%; 10 years, 86% and HIV/HCV, 5 years, 87%; 10 years, 73%; P <.05). Conclusion These results must be taken into account to establish an optimal therapeutic strategy, including the appropriate timing of liver transplantation in HIV/HCV-coinfected patients in Japan

    DOCK2 is involved in the host genetics and biology of severe COVID-19

    Get PDF
    「コロナ制圧タスクフォース」COVID-19疾患感受性遺伝子DOCK2の重症化機序を解明 --アジア最大のバイオレポジトリーでCOVID-19の治療標的を発見--. 京都大学プレスリリース. 2022-08-10.Identifying the host genetic factors underlying severe COVID-19 is an emerging challenge. Here we conducted a genome-wide association study (GWAS) involving 2, 393 cases of COVID-19 in a cohort of Japanese individuals collected during the initial waves of the pandemic, with 3, 289 unaffected controls. We identified a variant on chromosome 5 at 5q35 (rs60200309-A), close to the dedicator of cytokinesis 2 gene (DOCK2), which was associated with severe COVID-19 in patients less than 65 years of age. This risk allele was prevalent in East Asian individuals but rare in Europeans, highlighting the value of genome-wide association studies in non-European populations. RNA-sequencing analysis of 473 bulk peripheral blood samples identified decreased expression of DOCK2 associated with the risk allele in these younger patients. DOCK2 expression was suppressed in patients with severe cases of COVID-19. Single-cell RNA-sequencing analysis (n = 61 individuals) identified cell-type-specific downregulation of DOCK2 and a COVID-19-specific decreasing effect of the risk allele on DOCK2 expression in non-classical monocytes. Immunohistochemistry of lung specimens from patients with severe COVID-19 pneumonia showed suppressed DOCK2 expression. Moreover, inhibition of DOCK2 function with CPYPP increased the severity of pneumonia in a Syrian hamster model of SARS-CoV-2 infection, characterized by weight loss, lung oedema, enhanced viral loads, impaired macrophage recruitment and dysregulated type I interferon responses. We conclude that DOCK2 has an important role in the host immune response to SARS-CoV-2 infection and the development of severe COVID-19, and could be further explored as a potential biomarker and/or therapeutic target

    2. Progress in Management of HIV Infection

    No full text

    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

    sICs of IgG or IgM on purified rCD4s from HIV-1<sup>+</sup> Pts is molecularly linked to surface CD4 and shows slow turnover.

    No full text
    <p>(a) Summary of the percentages of IgG<sup>+</sup> rCD4s or IgM<sup>+</sup> rCD4s in healthy individuals (<i>H</i>), acute HIV-1<sup>+</sup> individuals (<i>A</i>), and chronic pre-symptomatic HIV-1<sup>+</sup> individuals (<i>C</i>) before, after 3 yrs of complete suppression of VL (<50 copies/ml) with ART (Treated), or untreated for 3 yrs (Untreated). Bars, SD. (b) Representative FACS of IgG expression on rCD4s from HIV-1<sup>+</sup> Pts. (c) Anti-IgG Ab immunoblotting of purified HIV-1<sup>+</sup> Pt rCD4 lysates. For the comparison of IgG binding levels, MFI values of IgG on rCD4s of the lysate samples are denoted above. IgG, positive IgG control; H, rCD4 lysate from an HIV-1-seronegative healthy donor. (d) Three-dimensional reconstitution confocal micrographs of Igs (Qdot655, green) and CD4 (Cy2, red) in rCD4s from an HIV-1<sup>+</sup> Pt. (e, f) Representative time course of FACS (e) and calculated half-life of sICs (f) in purified rCD4s from an HIV-1<sup>+</sup> Pt. Bar, SD. (g) Percentage of Ig<sup>+</sup> cells in purified HIV-1<sup>+</sup> Pt rCD4s without (none) or with 10 min of 0.05% trypsinization (Tryp). (h) Percentage of Ig<sup>+</sup> cells in purified HIV-1<sup>+</sup> Pt rCD4s before (0 h), after 58 h of culture (58 h), or 58 h of culture with exposure to HIV-1<sup>+</sup> Pt serum (58 h+Pt-serum). (i) Changes in percentages of IgM<sup>+</sup> or IgG<sup>+</sup> rCD4s in blood, plasma VL, and CD4 lymphocyte counts during ART in the four HIV-1<sup>+</sup> Pts. Two patients discontinued therapy after substantial suppression of VLs (left panels). HIV-1 RNA levels in two other patients were suppressed to undetectable levels for approximately 2 yr with ART (right panels). (j, k) Summary of the percentages (k) and representative FACS (j) of Igs on purified HIV-1<sup>+</sup> Pt rCD4s before and after 6 h of PMA (0.3 ng/ml) exposure. (l) Fluorescence and DIC images of purified HIV-1<sup>+</sup> Pt rCD4s that were stained with anti-Ig Abs (Cy2, green) and goat polyclonal anti-CD4 (Cy3, red) before and after 6 h of PMA exposure. Data in d and l are representative of five independent experiments.</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 µg/ml), cycloheximide (50 µg/ml) and ActD (20 µg/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/µl). 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
    corecore