11 research outputs found

    image_1.tif

    No full text
    <p>The requirements for effector and memory CD8 T cell development are incompletely understood. Recent work has revealed a role for G-protein coupled receptor 18 (GPR18) in establishment of the intestinal CD8αα intraepithelial lymphocyte compartment. Here, we report that GPR18 is also functionally expressed in conventional CD8αβ T cells. When the receptor is lacking, mice develop fewer CD8<sup>+</sup> KLRG1<sup>+</sup> Granzyme B<sup>+</sup> effector-memory cells. Bone marrow chimera studies show that the GPR18 requirement is CD8 T cell intrinsic. GPR18 is not required for T-bet expression in KLRG1<sup>+</sup> CD8 T cells. Gene transduction experiments confirm the functional activity of GPR18 in CD8 T cells. In summary, we describe a novel GPCR requirement for establishment or maintenance of the CD8 KLRG1<sup>+</sup> effector-memory T cell compartment. These findings have implications for methods to augment CD8 effector cell numbers.</p

    image_3.tif

    No full text
    <p>The requirements for effector and memory CD8 T cell development are incompletely understood. Recent work has revealed a role for G-protein coupled receptor 18 (GPR18) in establishment of the intestinal CD8αα intraepithelial lymphocyte compartment. Here, we report that GPR18 is also functionally expressed in conventional CD8αβ T cells. When the receptor is lacking, mice develop fewer CD8<sup>+</sup> KLRG1<sup>+</sup> Granzyme B<sup>+</sup> effector-memory cells. Bone marrow chimera studies show that the GPR18 requirement is CD8 T cell intrinsic. GPR18 is not required for T-bet expression in KLRG1<sup>+</sup> CD8 T cells. Gene transduction experiments confirm the functional activity of GPR18 in CD8 T cells. In summary, we describe a novel GPCR requirement for establishment or maintenance of the CD8 KLRG1<sup>+</sup> effector-memory T cell compartment. These findings have implications for methods to augment CD8 effector cell numbers.</p

    image_6.tif

    No full text
    <p>The requirements for effector and memory CD8 T cell development are incompletely understood. Recent work has revealed a role for G-protein coupled receptor 18 (GPR18) in establishment of the intestinal CD8αα intraepithelial lymphocyte compartment. Here, we report that GPR18 is also functionally expressed in conventional CD8αβ T cells. When the receptor is lacking, mice develop fewer CD8<sup>+</sup> KLRG1<sup>+</sup> Granzyme B<sup>+</sup> effector-memory cells. Bone marrow chimera studies show that the GPR18 requirement is CD8 T cell intrinsic. GPR18 is not required for T-bet expression in KLRG1<sup>+</sup> CD8 T cells. Gene transduction experiments confirm the functional activity of GPR18 in CD8 T cells. In summary, we describe a novel GPCR requirement for establishment or maintenance of the CD8 KLRG1<sup>+</sup> effector-memory T cell compartment. These findings have implications for methods to augment CD8 effector cell numbers.</p

    image_2.tif

    No full text
    <p>The requirements for effector and memory CD8 T cell development are incompletely understood. Recent work has revealed a role for G-protein coupled receptor 18 (GPR18) in establishment of the intestinal CD8αα intraepithelial lymphocyte compartment. Here, we report that GPR18 is also functionally expressed in conventional CD8αβ T cells. When the receptor is lacking, mice develop fewer CD8<sup>+</sup> KLRG1<sup>+</sup> Granzyme B<sup>+</sup> effector-memory cells. Bone marrow chimera studies show that the GPR18 requirement is CD8 T cell intrinsic. GPR18 is not required for T-bet expression in KLRG1<sup>+</sup> CD8 T cells. Gene transduction experiments confirm the functional activity of GPR18 in CD8 T cells. In summary, we describe a novel GPCR requirement for establishment or maintenance of the CD8 KLRG1<sup>+</sup> effector-memory T cell compartment. These findings have implications for methods to augment CD8 effector cell numbers.</p

    image_4.tif

    No full text
    <p>The requirements for effector and memory CD8 T cell development are incompletely understood. Recent work has revealed a role for G-protein coupled receptor 18 (GPR18) in establishment of the intestinal CD8αα intraepithelial lymphocyte compartment. Here, we report that GPR18 is also functionally expressed in conventional CD8αβ T cells. When the receptor is lacking, mice develop fewer CD8<sup>+</sup> KLRG1<sup>+</sup> Granzyme B<sup>+</sup> effector-memory cells. Bone marrow chimera studies show that the GPR18 requirement is CD8 T cell intrinsic. GPR18 is not required for T-bet expression in KLRG1<sup>+</sup> CD8 T cells. Gene transduction experiments confirm the functional activity of GPR18 in CD8 T cells. In summary, we describe a novel GPCR requirement for establishment or maintenance of the CD8 KLRG1<sup>+</sup> effector-memory T cell compartment. These findings have implications for methods to augment CD8 effector cell numbers.</p

    CTSB expression was up-regulated in dermal vasculature of Fli1<sup>+/−</sup> mice and in Fli1 siRNA-treated HDMECs.

    No full text
    <p>Immunodetection of CTSB proteins in the skin sections of 3 month-old wild type (A) and Fli1<sup>+/−</sup> (B) mice (original magnification was ×40) by Vectastain ABC kit according to the manufacturer's instruction. Insets (original magnification was ×40) depict representative arterioles (panel 1), venules (panel 2), and capillaries (panel 3; red arrowheads), respectively. Representative results in 5 wild type and 5 Fli1<sup>+/−</sup> mice are shown. (C) HDMECs were seeded shortly before transfection. The cells were transfected with 10 nM of Fli1 and scrambled non-silencing siRNA (Santa Cruz) using HiPerfect transfection reagent (Qiagen, Valencia, CA, USA) for 72 hours. Cells were then serum starved for the last 24 hours. mRNA levels of Fli1 and CTSB genes were examined by quantitative real-time PCR and normalized to the levels of human 18S rRNA gene. Results of controls or relative value compared with the controls are expressed as means ± SD of 3 independent experiments. Statistical analysis was carried out with a 2-tailed paired t-test. *P<0.05, **P<0.005.</p

    Serum pro-CTSB levels in dcSSc patients further classified into subgroups based on disease duration.

    No full text
    <p>dcSSc patients were divided into 3 subgroups: those with disease duration of <1 years, those with disease duration of 1 to 6 years, and those with disease duration of >6 years. Serum pro-CTSB levels were determined by a specific ELISA. The horizontal bars indicate the mean value in each group. Statistical analysis was carried out with a Kruskal-Wallis test and a Steel-Dwass' test for multiple comparison. *P<0.05.</p

    Correlation of serum pro-cathepsin B levels with clinical features in patients with dcSSc and lcSSc.

    No full text
    <p>Unless noted otherwise, values are percentages. dcSSc, diffuse cutaneous systemic sclerosis; lcSSc, limited cutaneous systemic sclerosis; MRSS, modified Rodnan total skin thickness score; DLco, diffuse capacity for carbon monoxide; VC, vital capacity; RVSP; right ventricular systolic pressure. Patients were evaluated for the presence of esophageal, pulmonary, cardiac, renal, or muscle involvements, as follows. Esophagus hypomotility was defined as distal esophageal hypomotility on barium-contrast radiography. Interstitial lung disease (ILD) was defined as bibasilar interstitial fibrosis on chest radiographs, and in patients with no abnormalities on chest radiographs early ILD was defined as alveolitis on high-resolution computer tomography. Elevated right ventricular systolic pressure (RVSP) was defined as 35 mmHg or more on echocardiogram. Cardiac involvement was defined as any of the following: symptomatic pericarditis, clinical evidence of left ventricular congestive heart failure, or arrhythmias requiring treatment. Scleroderma renal crisis was defined as malignant hypertension and/or rapidly progressive renal failure. Skeletal muscle involvement was defined as proximal muscle weakness and elevated serum creatine kinase levels, plus abnormal electromyographic findings consistent with myopathy and/or histopathologic changes in inflammatory myopathy. Disease onset was defined as the first clinical event of SSc other than Raynaud's phenomenon. Disease duration was defined as the interval between the onset and the time the blood samples were drawn. Statistical analysis was carried out with Fisher's exact probability test.</p><p>*P<0.05.</p

    CTSB expression was up-regulated in dermal vasculatures of SSc patients compared to those in controls.

    No full text
    <p>CTSB expression levels in dermal vasculatures were determined by immunohistochemistry in skin section from 8 healthy control subjects (A, B) and 8 SSc patients (C, D). Representative results are shown. Original magnification was ×200 (A, C) and ×400 (B, D). Analysis of CTSB expression levels in vessel walls is included in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0032272#pone-0032272-t002" target="_blank">Table 2</a>.</p

    Cathepsin B levels in dermal vasculature in normal and systemic sclerosis skin.

    No full text
    <p>NS, normal skin; SSc, systemic sclerosis; dcSSc, diffuse cutaneous systemic sclerosis; lcSSc, limited cutaneous systemic sclerosis. We used the following grading system: −, no staining; +, slight staining; ++, moderate staining; +++, strong staining.</p
    corecore