565 research outputs found

    Stabilin receptors clear LPS and control systemic inflammation

    Get PDF
    Lipopolysaccharides (LPSs) cause lethal endotoxemia if not rapidly cleared from blood circulation. Liver sinusoidal endothelial cells (LSEC) systemically clear LPS by unknown mechanisms. We discovered that LPS clearance through LSEC involves endocytosis and lysosomal inactivation via Stabilin-1 and 2 (Stab1 and Stab2) but does not involve TLR4. Cytokine production was inversely related to clearance/endocytosis of LPS by LSEC. When exposed to LPS, Stabilin double knockout mice (Stab DK) and Stab1 KO, but not Stab2 KO, showed significantly enhanced systemic inflammatory cytokine production and early death compared with WT mice. Stab1 KO is not significantly different from Stab DK in circulatory LPS clearance, LPS uptake and endocytosis by LSEC, and cytokine production. These data indicate that (1) Stab1 receptor primarily facilitates the proactive clearance of LPS and limits TLR4-mediated inflammation and (2) TLR4 and Stab1 are functionally opposing LPS receptors. These findings suggest that endotoxemia can be controlled by optimizing LPS clearance by Stab1

    A proteomic approach based on multiple parallel separation for the unambiguous identification of an antibody cognate antigen

    No full text
    Autoantibodies obtained from cancer patients have been identified as useful tools for cancer diagnostics, prognostics, and as potential targets for immunotherapy. Serological proteome analysis in combination with 2-DE is a classic strategy for identification of tumor-associated antigens in the serum of cancer patients. However, serological proteome analysis cannot always indicate the true antigen out of a complex proteome identified from a single protein spot because the most abundant protein is not always the most antigenic. To address this problem, we utilized multiple parallel separation (MPS) for proteome separation. The common identities present in the fractions obtained using different separation methods were regarded as the true antigens. The merit of our MPS technique was validated using anti-ARPC2 and anti-PTEN antibodies. Next, we applied the MPS technique for the identification of glycyl-tRNA synthetase as the cognate antigen for an autoantibody that was overexpressed in the plasma of breast cancer patients. These results reveal that MPS can unambiguously identify an antibody cognate antigen by reducing false-positives. Therefore, MPS could be used for the characterization of diagnostic antibodies raised in laboratory animals as well as autoantibodies isolated from diseased patients.Desmetz C, 2009, J PROTEOMICS, V72, P982, DOI 10.1016/j.jprot.2009.06.004Achilli F, 2009, DIS MODEL MECH, V2, P359, DOI 10.1242/dmm.002527Mou ZR, 2009, CANCER LETT, V278, P123, DOI 10.1016/j.canlet.2008.09.009Kim HJ, 2009, J PROTEOME RES, V8, P1368, DOI 10.1021/pr8007573Waller LN, 2008, J PROTEOME RES, V7, P4577, DOI 10.1021/pr8001518Caron M, 2007, MOL CELL PROTEOMICS, V6, P1115, DOI 10.1074/mcp.R600016-MCP200Pardo M, 2007, J PROTEOME RES, V6, P2802, DOI 10.1021/pr070021tCanelle L, 2006, ELECTROPHORESIS, V27, P1609, DOI 10.1002/elps.200500712Gires O, 2004, CELL MOL LIFE SCI, V61, P1198, DOI 10.1007/s00018-004-4045-8Imafuku Y, 2004, DIS MARKERS, V20, P149Stulik J, 2003, PROTEOMICS, V3, P951, DOI 10.1002/pmic.200300370Lichtenfels R, 2003, BBA-PROTEINS PROTEOM, V1646, P21, DOI 10.1016/S1570-9639(02)00547-2Wasenius VM, 2003, CLIN CANCER RES, V9, P68Ros A, 2002, PROTEOMICS, V2, P151Brichory FM, 2001, P NATL ACAD SCI USA, V98, P9824Scandurro AB, 2001, INT J ONCOL, V19, P129Klade CS, 2001, PROTEOMICS, V1, P890Chong BE, 2001, RAPID COMMUN MASS SP, V15, P291Soussi T, 2000, CANCER RES, V60, P1777Ward RL, 1999, HUM IMMUNOL, V60, P510Leslie RDG, 1999, DIABETOLOGIA, V42, P3Figeys D, 1998, ELECTROPHORESIS, V19, P1811Yamamoto A, 1997, INTERNAL MED, V36, P724Hirakata M, 1996, ARTHRITIS RHEUM, V39, P146SAHIN U, 1995, P NATL ACAD SCI USA, V92, P11810GE Q, 1994, J BIOL CHEM, V269, P28790

    A proteomic approach based on multiple parallel separation for the unambiguous identification of an antibody cognate antigen

    No full text
    Autoantibodies obtained from cancer patients have been identified as useful tools for cancer diagnostics, prognostics, and as potential targets for immunotherapy. Serological proteome analysis in combination with 2-DE is a classic strategy for identification of tumor-associated antigens in the serum of cancer patients. However, serological proteome analysis cannot always indicate the true antigen out of a complex proteome identified from a single protein spot because the most abundant protein is not always the most antigenic. To address this problem, we utilized multiple parallel separation (MPS) for proteome separation. The common identities present in the fractions obtained using different separation methods were regarded as the true antigens. The merit of our MPS technique was validated using anti-ARPC2 and anti-PTEN antibodies. Next, we applied the MPS technique for the identification of glycyl-tRNA synthetase as the cognate antigen for an autoantibody that was overexpressed in the plasma of breast cancer patients. These results reveal that MPS can unambiguously identify an antibody cognate antigen by reducing false-positives. Therefore, MPS could be used for the characterization of diagnostic antibodies raised in laboratory animals as well as autoantibodies isolated from diseased patients.Desmetz C, 2009, J PROTEOMICS, V72, P982, DOI 10.1016/j.jprot.2009.06.004Achilli F, 2009, DIS MODEL MECH, V2, P359, DOI 10.1242/dmm.002527Mou ZR, 2009, CANCER LETT, V278, P123, DOI 10.1016/j.canlet.2008.09.009Kim HJ, 2009, J PROTEOME RES, V8, P1368, DOI 10.1021/pr8007573Waller LN, 2008, J PROTEOME RES, V7, P4577, DOI 10.1021/pr8001518Caron M, 2007, MOL CELL PROTEOMICS, V6, P1115, DOI 10.1074/mcp.R600016-MCP200Pardo M, 2007, J PROTEOME RES, V6, P2802, DOI 10.1021/pr070021tCanelle L, 2006, ELECTROPHORESIS, V27, P1609, DOI 10.1002/elps.200500712Gires O, 2004, CELL MOL LIFE SCI, V61, P1198, DOI 10.1007/s00018-004-4045-8Imafuku Y, 2004, DIS MARKERS, V20, P149Stulik J, 2003, PROTEOMICS, V3, P951, DOI 10.1002/pmic.200300370Lichtenfels R, 2003, BBA-PROTEINS PROTEOM, V1646, P21, DOI 10.1016/S1570-9639(02)00547-2Wasenius VM, 2003, CLIN CANCER RES, V9, P68Ros A, 2002, PROTEOMICS, V2, P151Brichory FM, 2001, P NATL ACAD SCI USA, V98, P9824Scandurro AB, 2001, INT J ONCOL, V19, P129Klade CS, 2001, PROTEOMICS, V1, P890Chong BE, 2001, RAPID COMMUN MASS SP, V15, P291Soussi T, 2000, CANCER RES, V60, P1777Ward RL, 1999, HUM IMMUNOL, V60, P510Leslie RDG, 1999, DIABETOLOGIA, V42, P3Figeys D, 1998, ELECTROPHORESIS, V19, P1811Yamamoto A, 1997, INTERNAL MED, V36, P724Hirakata M, 1996, ARTHRITIS RHEUM, V39, P146SAHIN U, 1995, P NATL ACAD SCI USA, V92, P11810GE Q, 1994, J BIOL CHEM, V269, P28790

    Cholesterol-modified sphingomyelin chimeric lipid bilayer for improved therapeutic delivery

    No full text
    Abstract Cholesterol (Chol) fortifies packing and reduces fluidity and permeability of the lipid bilayer in vesicles (liposomes)-mediated drug delivery. However, under the physiological environment, Chol is rapidly extracted from the lipid bilayer by biomembranes, which jeopardizes membrane stability and results in premature leakage for delivered payloads, yielding suboptimal clinic efficacy. Herein, we report a Chol-modified sphingomyelin (SM) lipid bilayer via covalently conjugating Chol to SM (SM-Chol), which retains membrane condensing ability of Chol. Systemic structure activity relationship screening demonstrates that SM-Chol with a disulfide bond and longer linker outperforms other counterparts and conventional phospholipids/Chol mixture systems on blocking Chol transfer and payload leakage, increases maximum tolerated dose of vincristine while reducing systemic toxicities, improves pharmacokinetics and tumor delivery efficiency, and enhances antitumor efficacy in SU-DHL-4 diffuse large B-cell lymphoma xenograft model in female mice. Furthermore, SM-Chol improves therapeutic delivery of structurally diversified therapeutic agents (irinotecan, doxorubicin, dexamethasone) or siRNA targeting multi-drug resistant gene (p-glycoprotein) in late-stage metastatic orthotopic KPC-Luc pancreas cancer, 4T1-Luc2 triple negative breast cancer, lung inflammation, and CT26 colorectal cancer animal models in female mice compared to respective FDA-approved nanotherapeutics or lipid compositions. Thus, SM-Chol represents a promising platform for universal and improved drug delivery

    The Impact of HER2-Low Expression on Oncologic Outcomes in Hormone Receptor-Positive Breast Cancer

    No full text
    Breast cancer is a prevalent malignancy with increasing incidence, particularly in Asian countries. Classification based on estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status is pivotal in determining treatment. Recent advances have challenged the traditional dichotomy in HER2 classification, prompting investigation into the HER2-low subtype’s characteristics and outcomes. This retrospective study analyzed 10,186 non-metastatic hormone receptor (HR)-positive, HER2-negative breast cancer cases treated from 2008 to 2020. Data encompassed clinical, pathological, and treatment information. Oncologic outcomes included disease-free survival (DFS), overall survival (OS), and breast cancer-specific survival (BCSS). In total, 56.5% were HER2-low cases. Differences in patient characteristics were noted, with more BRCA1/2 mutations and higher mastectomy rates in the HER2-low group (p = 0.002, p p p = 0.012, p = 0.013, and p = 0.013, respectively). Notably, the prognosis differed between premenopausal and postmenopausal subgroups, with BCSS benefitting premenopausal patients (p = 0.047) and DFS and OS benefitting postmenopausal patients in the HER2-low group (p = 0.004, p = 0.009, respectively). Multivariate analysis confirmed HER2 status as an independent predictor of these outcomes (p = 0.010, p = 0.008, and p = 0.014, respectively). This extensive single-center study elucidates the favorable prognosis associated with HER2-low status in HR-positive breast cancer. However, this effect differs among premenopausal and postmenopausal patients, necessitating further research into the underlying tumor biology

    Berberine Suppresses Cell Motility Through Downregulation of TGF-β1 in Triple Negative Breast Cancer Cells

    No full text
    Background/Aims: Transforming growth factor-beta proteins (TGF-βs) are multifunctional growth factors and powerful modulators of the epithelial-mesenchymal transition (EMT) in a variety of cancer types including breast and lung cancer cells. Here, we demonstrated the inhibitory effect of berberine (BBR) on tumor growth and metastasis of triple negative breast cancer (TNBC) cells via suppression of TGF-β1 expression. Methods: The levels of mRNA expression were analyzed by real-time PCR. The levels of MMP-2, MMP-9 and TGF-β1 protein expression were analyzed by zymography and confocal microscopy, respectively. Cell migration was analyzed by wound healing assay. Tumorigenicity of TNBC cells such as tumor growth and metastasis was analyzed using xenograft models. Results: In a clinical data set, aberrant TGF-β1 expression was associated with poor prognosis of breast cancer patients. Our in vitro results using TNBC cells showed that the expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 and the capacity for cell migration were increased by TGF-β1 treatment. In contrast, basal levels of MMP-2 and MMP-9 were suppressed by a specific TGF-β receptor I inhibitor, SB431542. In addition, TGF-β1–induced MMP-2 and MMP-9 expression and cell migration were decreased by SB431542. Interestingly, we showed for the first time that BBR decreased the level of TGF-β1, but not TGF-β2, in TNBC cells. Furthermore, BBR significantly decreased the level of MMP-2 expression as well as the capacity for cell migration in TNBC cells. Finally, we examined the effect of BBR on in vivo tumor growth and lung metastasis in MDA-MB231 and 4T1 breast cancer xenograft models and showed that both were significantly decreased following BBR treatment. Conclusion: BBR suppresses tumorigenicity of TNBC cells through inhibition of TGF-β1 expression. Therefore, we demonstrate that BBR could be a promising drug for treatment of TNBC

    Autoantibody to Tumor Antigen, Alpha 2-HS Glycoprotein: A Novel Biomarker of Breast Cancer Screening and Diagnosis

    No full text
    We sought to identify a new serum biomarker for breast cancer screening and diagnosis using stepwise proteomic analysis of sera from breast cancer patients to detect the presence of autoantibodies that react with urinary protein. Two-dimensional immunoblotting was done for screening autoimmunogenic tumor antigens in the urine of breast cancer patients. Reactive spots were identified by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Among urinary proteins separated by two-dimensional electrophoresis, 13 spots showed strong reactivity with pooled sera from breast cancer patients or control sera. By mass spectrometry, we identified alpha 2-HS glycoprotein (AHSG) as a tumor antigen. Peripheral blood was obtained from 81 women diagnosed with breast cancer before surgery and 73 female donors without evidence of any malignancy for the individual analysis. In one-dimensional Western blot analysis, AHSG autoantibody was detected in 64 of 81 breast cancer patients (79.1%) and in 7 of 73 controls (9.6%). The sensitivity of this test in breast cancer patients was 79.0%. Our results suggest that AHSG and anti-AHSG autoantibody may be useful serum biomarkers for breast cancer screening and diagnosis.Chang JW, 2008, PROTEOM CLIN APPL, V2, P23, DOI 10.1002/prca.200780049Harris L, 2007, J CLIN ONCOL, V25, P5287, DOI 10.1200/JCO.2007.14.2364Caron M, 2007, MOL CELL PROTEOMICS, V6, P1115, DOI 10.1074/mcp.R600016-MCP200Hu S, 2006, PROTEOMICS, V6, P6326, DOI 10.1002/pmic.200600284Fujita Y, 2006, CLIN CANCER RES, V12, P6415, DOI 10.1158/1078-0432.CCR-06-1315Downes MR, 2006, BIOMARKERS, V11, P406, DOI 10.1080/13547500600799821Chang JW, 2005, FEBS LETT, V579, P2873, DOI 10.1016/j.febslet.2005.04.028Canelle L, 2005, J IMMUNOL METHODS, V299, P77, DOI 10.1016/j.jim.2005.01.015Molina R, 2005, TUMOR BIOL, V26, P281, DOI 10.1159/000089260Swallow CJ, 2004, CANCER RES, V64, P6402Carlsson L, 2004, J ANDROL, V25, P699Hong SH, 2004, CANCER RES, V64, P5504Hutchinson JN, 2004, CANCER RES, V64, P3171Pieper R, 2004, PROTEOMICS, V4, P1159, DOI 10.1002/pmic.200300661Mocellin S, 2003, BBA-REV CANCER, V1653, P61, DOI 10.1016/S0304-419X(03)00032-5Wilson KS, 2002, AM J PATHOL, V161, P1171Szweras M, 2002, J BIOL CHEM, V277, P19991, DOI 10.1074/jbc.M112234200Volkmann M, 2002, ONCOLOGY-BASEL, V63, P297, DOI 10.1159/000065472Sugi T, 2002, J REPROD IMMUNOL, V53, P269Le Naour F, 2001, CLIN CANCER RES, V7, P3328Brichory FM, 2001, P NATL ACAD SCI USA, V98, P9824Bosscher JR, 2001, GYNECOL ONCOL, V81, P138Gion M, 2001, EUR J CANCER, V37, P355Mathews ST, 2000, MOL CELL ENDOCRINOL, V164, P87Chen SH, 2000, FASEB J, V14, P565Menard S, 2000, J CELL PHYSIOL, V182, P150METCALFE S, 2000, BREAST CANCER RES, V2, P438Duffy MJ, 1999, ANN CLIN BIOCHEM, V36, P579Lenner P, 1999, BRIT J CANCER, V79, P927Wang HC, 1998, P NATL ACAD SCI USA, V95, P14429Molina R, 1998, BREAST CANCER RES TR, V51, P109Conroy SE, 1998, EUR J CANCER, V34, P942Disis ML, 1997, J CLIN ONCOL, V15, P3363Disis ML, 1997, ADV CANCER RES, V71, P343Chu KC, 1996, J NATL CANCER I, V88, P1571Bast RC, 1996, J CLIN ONCOL, V14, P2843vonMensdorffPouilly S, 1996, EUR J CANCER, V32A, P1325vanDalen A, 1996, ANTICANCER RES, V16, P2345MADIGAN MP, 1995, J NATL CANCER I, V87, P1681GOUREVITCH MM, 1995, BRIT J CANCER, V72, P934SRINIVAS PR, 1993, MOL ENDOCRINOL, V7, P1445JAMEEL A, 1992, INT J CANCER, V50, P409DNISTRIAN AM, 1991, TUMOR BIOL, V12, P82VANDALEN A, 1990, TUMOR BIOL, V11, P189TABAR L, 1985, LANCET, V1, P829ANDERSON L, 1977, P NATL ACAD SCI USA, V74, P54211

    Uneven recovery patterns of compromised health-related quality of life (EQ-5D-3 L) domains for breast Cancer survivors: a comparative study

    No full text
    Abstract Background Although several studies have evaluated health-related quality of life (HRQoL) in breast cancer survivors, few have compared HRQoL between breast cancer survivors and an age-matched general population in terms of improvement patterns according to time after surgery. Thus, we compared the postoperative changes in HRQoL in breast cancer survivors with those of age-matched general population groups using the EuroQoL five-dimension three-level questionnaire (EQ-5D-3 L). Methods EQ-5D-3 L questionnaires were obtained from 686 breast cancer survivors during follow-up visits. They were divided into five groups according to time after surgery: 0–5 months, 6–11 months, 12–35 months, 36–59 months, and ≥ 60 months. Their EQ-5D-3 L data, covering five dimensions (mobility, self-care, usual activities, pain/discomfort, and anxiety/depression), were compared with those of age-matched general population groups. Results The mean EQ-5D-3 L index of breast cancer survivors was high in group with longer time after surgery and the mean EQ-5D-3 L index of breast cancer group over 5 years after surgery was similar to that of an age-matched general population (0.919 vs 0.928, p = 0.305). Although there were deficits in all dimensions of breast cancer survivors, motility eventually exceeded that of general population groups and self-care and usual activities of groups over 3 years after surgery matched those of general population however, pain/discomfort and anxiety/depression of survivors over 5 years after surgery remained worse than those of the general population (p = 0.028, p < 0.001). Conclusions Motility, self-care, and usual activities decreased in the early postoperative period for breast cancer survivors but showed recovery after 3 years. However, pain/discomfort and anxiety/depression remained poorer in these patients than in the general population for many years
    corecore