GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Research in Microbiology, Elsevier BV, Vol. 154, No. 2 ( 2003-3), p. 105-114
    Type of Medium: Online Resource
    ISSN: 0923-2508
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2003
    detail.hit.zdb_id: 2010710-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 54, No. 11 ( 2013-11), p. 2565-2567
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2013
    detail.hit.zdb_id: 2030637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Molecular Cancer, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2013-12)
    Abstract: Recent epidemiological studies demonstrate that both active and involuntary exposure to tobacco smoke increase the risk of breast cancer. Little is known, however, about the molecular mechanisms by which continuous, long term exposure to tobacco smoke contributes to breast carcinogenesis because most previous studies have focused on short term treatment models. In this work we have set out to investigate the progressive transforming effects of tobacco smoke on non-tumorigenic mammary epithelial cells and breast cancer cells using in vitro and in vivo models of chronic cigarette smoke exposure. Results We show that both non-tumorigenic (MCF 10A, MCF-12A) and tumorigenic (MCF7) breast epithelial cells exposed to cigarette smoke acquire mesenchymal properties such as fibroblastoid morphology, increased anchorage-independent growth, and increased motility and invasiveness. Moreover, transplantation experiments in mice demonstrate that treatment with cigarette smoke extract renders MCF 10A cells more capable to survive and colonize the mammary ducts and MCF7 cells more prone to metastasize from a subcutaneous injection site, independent of cigarette smoke effects on the host and stromal environment. The extent of transformation and the resulting phenotype thus appear to be associated with the differentiation state of the cells at the time of exposure. Analysis by flow cytometry showed that treatment with CSE leads to the emergence of a CD44 hi /CD24 low population in MCF 10A cells and of CD44 + and CD49f + MCF7 cells, indicating that cigarette smoke causes the emergence of cell populations bearing markers of self-renewing stem-like cells. The phenotypical alterations induced by cigarette smoke are accompanied by numerous changes in gene expression that are associated with epithelial to mesenchymal transition and tumorigenesis. Conclusions Our results indicate that exposure to cigarette smoke leads to a more aggressive and transformed phenotype in human mammary epithelial cells and that the differentiation state of the cell at the time of exposure may be an important determinant in the phenotype of the final transformed state.
    Type of Medium: Online Resource
    ISSN: 1476-4598
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2013
    detail.hit.zdb_id: 2091373-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Infection and Immunity, American Society for Microbiology, Vol. 74, No. 10 ( 2006-10), p. 5609-5616
    Abstract: We have previously shown that outer membrane protein A (OmpA) and type 1 fimbriae are the bacterial determinants involved in Escherichia coli K1 binding to human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier. In investigating the role of OmpA in E. coli K1 binding to HBMEC, we showed for the first time that ompA deletion decreased the expression of type 1 fimbriae in E. coli K1. Decreased expression of type 1 fimbriae in the ompA deletion mutant was largely the result of driving the fim promoter toward the type 1 fimbrial phase-OFF orientation. mRNA levels of fimB and fimE were found to be decreased with the OmpA mutant compared to the parent strain. Of interest, the ompA deletion further decreased the abilities of E. coli K1 to bind to and invade HBMEC under the conditions of fixing type 1 fimbria expression in the phase-ON or phase-OFF status. These findings suggest that the decreased ability of the OmpA mutant to interact with HBMEC is not entirely due to its decreased type 1 fimbrial expression and that OmpA and type 1 fimbriae facilitate the interaction of E. coli K1 with HBMEC at least in an additive manner.
    Type of Medium: Online Resource
    ISSN: 0019-9567 , 1098-5522
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2006
    detail.hit.zdb_id: 1483247-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Alcoholism: Clinical and Experimental Research, Wiley, Vol. 31, No. 1 ( 2007-01), p. 130-137
    Type of Medium: Online Resource
    ISSN: 0145-6008 , 1530-0277
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2007
    detail.hit.zdb_id: 2046886-6
    detail.hit.zdb_id: 3167872-5
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: BMC Genomics, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2011-12)
    Abstract: Although the high mobility group A1 ( HMGA1 ) gene is widely overexpressed in diverse cancers and portends a poor prognosis in some tumors, the molecular mechanisms that mediate its role in transformation have remained elusive. HMGA1 functions as a potent oncogene in cultured cells and induces aggressive lymphoid tumors in transgenic mice. Because HMGA1 chromatin remodeling proteins regulate transcription, HMGA1 is thought to drive malignant transformation by modulating expression of specific genes. Genome-wide studies to define HMGA1 transcriptional networks during tumorigenesis, however, are lacking. To define the HMGA1 transcriptome, we analyzed gene expression profiles in lymphoid cells from HMGA1a transgenic mice at different stages in tumorigenesis. Results RNA from lymphoid samples at 2 months (before tumors develop) and 12 months (after tumors are well-established) was screened for differential expression of 〉 20,000 unique genes by microarray analysis (Affymetrix) using a parametric and nonparametric approach. Differential expression was confirmed by quantitative RT-PCR in a subset of genes. Differentially expressed genes were analyzed for cellular pathways and functions using Ingenuity Pathway Analysis. Early in tumorigenesis, HMGA1 induced inflammatory pathways with NFkappaB identified as a major node. In established tumors, HMGA1 induced pathways involved in cell cycle progression, cell-mediated immune response, and cancer. At both stages in tumorigenesis, HMGA1 induced pathways involved in cellular development, hematopoiesis, and hematologic development. Gene set enrichment analysis showed that stem cell and immature T cell genes are enriched in the established tumors. To determine if these results are relevant to human tumors, we knocked-down HMGA1 in human T-cell leukemia cells and identified a subset of genes dysregulated in both the transgenic and human lymphoid tumors. Conclusions We found that HMGA1 induces inflammatory pathways early in lymphoid tumorigenesis and pathways involved in stem cells, cell cycle progression, and cancer in established tumors. HMGA1 also dyregulates genes and pathways involved in stem cells, cellular development and hematopoiesis at both early and late stages of tumorigenesis. These results provide insight into HMGA1 function during tumor development and point to cellular pathways that could serve as therapeutic targets in lymphoid and other human cancers with aberrant HMGA1 expression.
    Type of Medium: Online Resource
    ISSN: 1471-2164
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 2041499-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3969-3969
    Abstract: Abstract 3969 Poster Board III-905 Because T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive lymphoid leukemia with a high rate of relapse in both children and adults, research is urgently needed to discover molecular pathways that could be targeted in therapy. Recent studies have identified NOTCH1 activating mutations, genomic classifiers, and other molecular pathways that are disrupted in T-ALL, although these findings have not yet translated into better therapy or improved long-term survival. Thus, we are investigating molecular pathways that play a causative role in T-ALL and could be modulated by therapy. We previously showed that HMGA1a is overexpressed in human T-ALL. Further, HMGA1a transgenic mice develop aggressive T-ALL with complete penetrance, indicating that HMGA1a induces leukemogenic transformation in vivo. Disruption of the CDKN2A (INK4A/ARF) tumor suppressor locus has been reported in up to 90% of T-ALL, suggesting that this contributes to leukemic transformation in T-cells. To determine if loss of function of the INK4A/ARF tumor suppressor locus could cooperate with HMGA1a in T-ALL, we crossed our HMGA1a transgenics with mice that are null for the INK4A/ARF locus. We found that T-ALL was markedly accelerated in the HMGA1a transgenics that are also null at this tumor suppressor locus. These mice died significantly earlier than the HMGA1 transgenics that are wildtype at the INK4A/ARF locus (4.8 months+/- 1.0 month versus 10.5 +/- 2.5 months, p 〈 0.0001 by the student's t-test). The HMGA1a transgenic, INK4A/ARF null mice recapitulate salient clinical and pathologic features of human T-ALL. All mice appeared ill and exhibit marked organomegaly with extensive leukemic infiltration. Immunophenotyping by fluorescence activated cell sorting showed T-ALL in all cases with identical T-cell markers (CD3, CD8, and αβ-TCR) to those previously reported for the HMGA1a transgenics at 8-10 months. Interestingly, the HMGA1a-INK4A/ARF+/- mice also developed early lymphoid malignancy in 6/17 cases. The accelerated lymphoid malignancy in these mice suggests that there is loss of heterozygosity at the INK4A/ARF locus. Alternatively, haploinsufficiency of INK4A and/or ARF may be sufficient to promote lymphoid tumorigenesis in the setting of HMGA1a overexpression. To determine if HMGA1 overexpression and loss of INK4A/ARF expression occur in human T-ALL, we analyzed gene expression profile analysis in leukemic blasts from children and young adults with T-ALL. We found high levels of HMGA1 expression and low levels of INK4A/ARF expression in most patients. Taken together, our results indicate that overexpression of HMGA1 and loss of function at the INK4A/ARF (CDKN2A) tumor suppressor locus cooperate in T-cell leukemogenesis and these pathways could represent rational therapeutic targets. Further, our mice provide a novel preclinical model for T-ALL. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Physiological Society ; 2004
    In:  American Journal of Physiology-Cell Physiology Vol. 286, No. 1 ( 2004-01), p. C31-C42
    In: American Journal of Physiology-Cell Physiology, American Physiological Society, Vol. 286, No. 1 ( 2004-01), p. C31-C42
    Abstract: Thrombin and related protease-activated receptors 1, 2, 3, and 4 (PAR1–4) play a multifunctional role in many types of cells including endothelial cells. Here, using RT-PCR and immunofluorescence staining, we showed for the first time that PAR1–4 are expressed on primary human brain microvascular endothelial cells (HBMEC). Digital fluorescence microscopy and fura 2 were used to monitor intracellular Ca 2+ concentration ([Ca 2+ ] i ) changes in response to thrombin and PAR1-activating peptide (PAR1-AP) SFFLRN. Both thrombin and PAR1-AP induced a dose-dependent [Ca 2+ ] i rise that was inhibited by pretreatment of HBMEC with the phospholipase C inhibitor U-73122 and the sarco(endo)plasmic reticulum Ca 2+ -ATPase inhibitor thapsigargin. Thrombin induced transient [Ca 2+ ] i increase, whereas PAR1-AP exhibited sustained [Ca 2+ ] i rise. The PAR1-AP-induced sustained [Ca 2+ ] i rise was significantly reduced in the absence of extracellular calcium or in the presence of an inhibitor of store-operated calcium channels, SKF-96365. Restoration of extracellular Ca 2+ to the cells that were initially activated by PAR1-AP in the absence of extracellular Ca 2+ resulted in significant [Ca 2+ ] i rise; however, this effect was not observed after thrombin stimulation. Pretreatment of the cells with a low thrombin concentration (0.1 nM) prevented [Ca 2+ ] i rise in response to high thrombin concentration (10 nM), but pretreatment with PAR1-AP did not prevent subsequent [Ca 2+ ] i rise to high PAR1-AP concentration. Additionally, treatment with thrombin decreased transendothelial electrical resistance in HBMEC, whereas PAR1-AP was without significant effect. These findings suggest that, in contrast to thrombin, stimulation of PAR1 by untethered peptide SFFLRN results in stimulation of store-operated Ca 2+ influx without significantly affecting brain endothelial barrier functions.
    Type of Medium: Online Resource
    ISSN: 0363-6143 , 1522-1563
    Language: English
    Publisher: American Physiological Society
    Publication Date: 2004
    detail.hit.zdb_id: 1477334-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4098-4098
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4098-4098
    Abstract: Pancreatic ductal adenocarcinoma is a highly lethal cancer accounting for over 160,000 deaths worldwide each year. Despite the high prevalence of pancreatic cancer, the molecular events that lead to tumor progression are poorly understood. The high-mobility group A1 gene (HMGA1) is enriched in diverse, high-grade cancers and embryonic stem cells. This gene encodes the HMGA1a and HMGA1b chromatin remodeling proteins, which function in regulating gene expression. We previously showed that HMGA1 induces oncogenic transformation in cultured cells and causes aggressive cancers in transgenic mice. Moreover, we found that high HMGA1 protein levels correlate with poor differentiation status and decreased survival in a study of & gt; 100 primary pancreatic ductal adenocarcinomas. These findings suggest that HMGA1 could drive a poorly differentiated, stem cell-like state and tumor progression in pancreatic ductal adenocarcinoma. In cultured, human pancreatic cells that overexpress K-RAS (HPNE-K-RAS cells), forced overexpression of HMGA1a leads to increased migration, invasion, and a transformed phenotype with anchorage-independent cell growth in soft agar. Because cyclooxygenase-2 (COX-2) is a downstream gene directly activated by HMGA1a in uterine cancers, we investigated the role of the HMGA1-COX-2 pathway in pancreatic adenocarcinoma. Our preliminary data suggest that HMGA1a and COX-2 mRNA levels are positively correlated in a pancreatic cancer cell lines. By chromatin immunoprecipitation, we found that HMGA1a binds to the COX-2 promoter in human pancreatic adenocarcinoma cells. Moreover, COX-2 inhibitors block tumorigenesis in human pancreatic cancer xenografts in nude mice. Taken together, our studies suggest that HMGA1a promotes tumor progression through up-regulation of COX-2 and this pathway could be targeted to treat or prevent human pancreatic adenocarcinoma. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4098.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1371-1371
    Abstract: Abstract 1371 Although the high mobility group AT-hook 1 (HMGA1) gene functions as a potent oncogene in experimental models and high expression of HMGA1 portends a poor prognosis in diverse tumors, its role in leukemogenesis has remained elusive. We showed previously that HMGA1 induces leukemic transformation in cultured cells and causes aggressive lymphoid leukemia in transgenic mice. Inhibiting HMGA1 expression blocks colony formation in human lymphoid leukemia cells in vitro. Moreover, high levels correlate with relapse in childhood acute lymphoblastic leukemia (ALL), suggesting that it plays an important role in ALL. Because HMGA1 functions as a chromatin remodeling protein that modulates gene expression, we hypothesized that it drives leukemogenesis by dysregulating specific genes and pathways. To identify genes and cellular pathways induced by HMGA1 that could be targeted in therapy, we performed global gene expression profile analysis from lymphoid cells from the HMGA1 transgenic mice at different stages in tumorigenesis. All HMGA1 transgenics succumb to lymphoid malignancy with complete penetrance by 8–12 months. Pooled RNA samples at 2 months (before tumors develop) and 12 months (after tumors are well-established) were analyzed for differential expression of 〉 20,000 unique genes by microarray analysis (Affymetrix) using both a parametric and nonparametric approach. A subset of differentially expressed genes was confirmed using quantitative, RT-PCR. Differentially expressed genes were analyzed for cellular pathways and functions using Ingenuity Pathway Analysis (IPA; www.ingenuity.com) and Gene Set Enrichment Analysis. To determine if these genes and pathways were relevant in human ALL, we knocked down HMGA1 expression in human ALL cells and assessed expression of a subset of the differentially expressed genes. Early in leukemogenesis (at 2 months), 113 genes were differentially expressed in the HMGA1 transgenics compared to controls. In established leukemia (12 months), 715 genes were differentially expressed. In established tumors, the dysregulated genes are involved in cancer, cell cycle regulation, and cell-mediated immune response by Ingenuity Pathway Analysis. Geneset enrichment showed that embryonic stem cell genes are enriched in the established leukemic cells. At both early and late stages in leukemogenesis, differentially regulated genes are involved in cellular development, hematopoiesis, and hematologic development. Early in leukemogenesis, most of the significantly dysregulated genes are involved in the inflammatory response and included NF-kappaB as a major node. In human ALL cells, knock-down of HMGA1 also resulted in knock-down of genes identified in our transgenic model, suggesting that these HMGA1 regulated genes are also relevant to human ALL. In summary, we found that HMGA1 induces inflammatory pathways early in leukemogenesis and pathways involved in embryonic stem cells, cell cycle progression, and cancer in established tumors. HMGA1 also dysregulates genes involved in cellular development and hematopoiesis at both early and late stages of tumorigenesis. Some of these HMGA1 pathways were also present in human ALL cells. Moreover, these results provide mechanistic insight into HMGA1 function at different stages in leukemogenesis and point to cellular pathways that could serve as therapeutic targets in ALL. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...