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: Cancer Cell, Elsevier BV, Vol. 31, No. 2 ( 2017-02), p. 181-193
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: TAXON, Wiley, Vol. 71, No. 1 ( 2022-02), p. 178-198
    Abstract: The shortage of reliable primary taxonomic data limits the description of biological taxa and the understanding of biodiversity patterns and processes, complicating biogeographical, ecological, and evolutionary studies. This deficit creates a significant taxonomic impediment to biodiversity research and conservation planning. The taxonomic impediment and the biodiversity crisis are widely recognized, highlighting the urgent need for reliable taxonomic data. Over the past decade, numerous countries worldwide have devoted considerable effort to Target 1 of the Global Strategy for Plant Conservation (GSPC), which called for the preparation of a working list of all known plant species by 2010 and an online world Flora by 2020. Brazil is a megadiverse country, home to more of the world's known plant species than any other country. Despite that, Flora Brasiliensis , concluded in 1906, was the last comprehensive treatment of the Brazilian flora. The lack of accurate estimates of the number of species of algae, fungi, and plants occurring in Brazil contributes to the prevailing taxonomic impediment and delays progress towards the GSPC targets. Over the past 12 years, a legion of taxonomists motivated to meet Target 1 of the GSPC, worked together to gather and integrate knowledge on the algal, plant, and fungal diversity of Brazil. Overall, a team of about 980 taxonomists joined efforts in a highly collaborative project that used cybertaxonomy to prepare an updated Flora of Brazil, showing the power of scientific collaboration to reach ambitious goals. This paper presents an overview of the Brazilian Flora 2020 and provides taxonomic and spatial updates on the algae, fungi, and plants found in one of the world's most biodiverse countries. We further identify collection gaps and summarize future goals that extend beyond 2020. Our results show that Brazil is home to 46,975 native species of algae, fungi, and plants, of which 19,669 are endemic to the country. The data compiled to date suggests that the Atlantic Rainforest might be the most diverse Brazilian domain for all plant groups except gymnosperms, which are most diverse in the Amazon. However, scientific knowledge of Brazilian diversity is still unequally distributed, with the Atlantic Rainforest and the Cerrado being the most intensively sampled and studied biomes in the country. In times of “scientific reductionism”, with botanical and mycological sciences suffering pervasive depreciation in recent decades, the first online Flora of Brazil 2020 significantly enhanced the quality and quantity of taxonomic data available for algae, fungi, and plants from Brazil. This project also made all the information freely available online, providing a firm foundation for future research and for the management, conservation, and sustainable use of the Brazilian funga and flora.
    Type of Medium: Online Resource
    ISSN: 0040-0262 , 1996-8175
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 2081189-5
    detail.hit.zdb_id: 204216-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cell Reports, Elsevier BV, Vol. 29, No. 6 ( 2019-11), p. 1660-1674.e7
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 4371-4371
    Abstract: Pheochromocytomas (PCC) and paragangliomas (PGL) are tumors of the autonomic nervous system; 25% are metastatic or locally aggressive. Characterization of the inherited basis of disease has identified a variety of underlying germline mutations; however, understanding of somatic alterations remains limited. As part of The Cancer Genome Atlas, we performed the most comprehensive genomic characterization of PCC/PGL to date, by applying eight genomic profiling assays to 173 patients. Despite having a low overall mutation rate per tumor, we observed remarkable diversity in genomic alterations. 27% of patients had a pathogenic germline mutation among eight known familial PCC/PGL susceptibility genes, thus making PCC/PGL the tumor type with the greatest rate of germline mutations in The Cancer Genome Atlas. 38% of patients possessed a somatic driver mutation across 12 genes. RET, NF1 and VHL were affected by both germline and somatic mutation, albeit with different mutation site tendencies. We identified a new somatic driver gene, CSDE1, which had coordinated intron splicing defects, DNA copy number loss, and RNA under-expression, suggesting a loss of function consequence. Most notably, we discovered the first fusion genes in PCC/PGL from RNA and DNA sequencing (7% of patients), demonstrating for the first time that inter-chromosomal translocation and gene fusion is a method of molecular pathogenesis in this disease. Recurrent, novel MAML3 fusion genes spanned three isoforms and were activating based on over-expression of MAML3 and on fusion transcript exonic expression. MAML3 fusion positive tumors had concomitant dual focal DNA amplification of the fusion gene partners and a significantly divergent methylation profile. Another novel driver gene in PCC/PGL, BRAF, was affected by a hotspot somatic mutation and by an activating fusion gene. Through integrated platform analysis, four statistically significant molecular subtypes of PCC/PGL were detected and found to represent divergent molecular etiology – the kinase signaling subtype, the pseudohypoxia subtype, the Wnt-altered subtype, and the cortical admixture subtype. In particular, MAML3 fusions and CSDE1 mutations defined the new Wnt-altered expression subtype of PCC. Adding to the limited set of prognostic markers in PCC/PGL, three molecular markers were positively associated with clinically aggressive disease: germline mutations in SDHB, somatic mutations in ATRX and fusions involving MAML3. Nearly all somatic driver mutations, germline driver mutations and fusion genes were mutually exclusive across the cohort and covered a large portion of the cohort (69%). Our study provides important novel insights into PCC/PGL biology and identifies potential markers for aggressive disease and therapeutic intervention. Citation Format: Lauren Fishbein, Ignaty Leshchiner, Vonn Walter, Ludmila Danilova, A Gordon Robertson, Amy Johnson, Tara Lichtenberg, Bradley A. Murray, Hanse K. Ghayee, Tobias Else, Shiyun Ling, Stuart R. Jefferys, Aguirre A. de Cubas, Brandon Wenz, Esther Korpershoek, Antonio L. Amelio, Liza Makowski, W Kimryn Rathmell, Anne-Paule Gimenez-Roqueplo, Thomas J. Giordano, Sylvia L. Asa, Arthur S. Tischler, The Cancer Genome Atlas Pheochromocytoma and Paraganglioma Analysis Working Group, Karel Pacak, Katherine L. Nathanson, Matthew D. Wilkerson. Integrated molecular characterization of pheochromocytoma and paraganglioma including a novel, recurrent and prognostic fusion gene. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4371.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3784-3784
    Abstract: Metabolic reprogramming is a hallmark of cancer. MYC oncoproteins orchestrate the control of many aspects of metabolism, including mitochondrial biogenesis, glycolysis and glutaminolysis. This response allows the highly proliferative cancer cell to meet the increased demands for macromolecules and energy. Here we report that MYC promotes anabolism by actively suppressing the autophagy-lysosomal catabolic pathway. Notably, disabling this response by reactivating catabolism provokes metabolic catastrophe and senescence in MYC-driven lymphomas. Gene expressing profiling of B220+ B cells from bone marrow of wild type (Wt) and pre-cancerous Eµ-Myc mice, and of Eµ-Myc lymphoma, demonstrated that genes involved in autophagy and lysosomal biogenesis are suppressed in MYC-expressing B cells. This response was due to MYC, as activation of the c-MYC transgene in human P493-6 B cells suppressed the expression of autophagy and lysosomal biogenesis genes. Further, this response was regulated by physiological cues that control endogenous c-Myc expression, as it was evident in naïve mouse splenic B cells treated with mitogen and in cytokine-treated hematopoietic cells. Finally, the suppression of autophagy-lysosomal genes was a hallmark of human malignancies with MYC involvement, including Burkitt lymphoma and colorectal cancer. Importantly, these changes in autophagy-lysosomal gene expression have functional consequences, as there are marked decreases in lysosomes in MYC-expressing versus control mouse embryonic fibroblasts and primary B cells, and in P493-6 B cells following MYC activation. Finally, consistent with an impaired autophagy-lysosomal pathway, there were marked increases in the levels of p62/Sequestrin, a receptor for ubiquitylated cargo on autophagosomes that is normally degraded by the autophagosomal pathway, in premalignant and neoplastic B220+ Eµ-Myc B cells. A master regulator of autophagy and lysosomal biogenesis is TFEB that, like MYC, functions as a basic helix-loop-helix leucine zipper transcription factor. Our studies suggest that MYC can block TFEB function at three levels. First, TFEB expression is suppressed in human malignancies with MYC involvement, suggesting that in some contexts MYC may repress TFEB transcription. Second, MYC can repress TFEB transcription targets by competing with TFEB for binding to autophagy-lysosome gene targets. Specifically, the DNA recognition sequence of TFEB (CLEAR sequence) overlaps with the E-Box sequence of MYC, and chromatin immunoprecipitation analyses showed that MYC bound to these recognition elements in select TFEB targets and repressed their expression. Finally, MYC-expressing B cells display activation of mTORC1, which phosphorylates TFEB and blocks its nuclear localization. Specifically, MYC-expressing B cells have marked increases in the phosphorylation of the mTORC1 substrates S6K and 4EBP1, and show cytoplasmic localization of TFEB. Given these findings, we hypothesized that MYC-transformed lymphoma cells might be sensitive to activating this catabolic pathway. We tested this hypothesis in Eµ-Myc;Rosa26-rtTa lymphoma cells that express the reverse tetracycline transactivator. These lymphoma cells were transduced with a pTight doxycycline- (Dox)-inducible retrovirus that expresses a constitutively active (mTORC1-resistant and nuclear) form of TFEB (TFEB-S211A) and the effects of the induction of TFEB were monitored. Importantly, our analyses established that the induction of TFEB-S211A markedly impaired the proliferation and tumorigenic potential of Eµ-Myc lymphoma. Mechanistically, this proliferative arrest was associated with the induction of the senescence program, and with an increased extracellular acidification rate. Strikingly, this metabolic shift was associated with reductions in oxygen consumption rate and reduced numbers of mitochondria. Therefore, TFEB functions as a tumor suppressor that harnesses MYC-induced tumorigenesis by inducing senescence and disabling mitochondrial anabolic pathways that are needed to sustain the growth of malignant cells. 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1032-1032
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1032-1032
    Abstract: The rate of HPV-induced head and neck squamous cell carcinoma (HNSCC) is steadily increasing and implicated in approximately 60% of all oropharyngeal carcinomas. The advent of whole genome, transcriptome, and proteome analyses have aided in identifying altered signaling pathways in HPV-induced HNSCCs, however, additional tools such as mouse models are needed to study the role of HPV oncogenes in oral tumor initiation and progression. Current inducible models of HPV-driven oral cancer do not accurately recapitulate the levels, stoichiometric ratios, or anatomic location of oncoprotein expression. To address these limitations, we developed a tractable genetically engineered mouse model (GEMM) that enables directed expression of high-risk HPV16 E6 and E7 oncogenes (Rosa26-loxP-STOP-loxP-E7iresE6; “H”) in the oral epithelium. Analysis of mouse embryonic fibroblasts treated with Cre recombinase confirmed E6 and E7 expression and changes in mRNA expression of known E6 and E7 targets. We crossed our H mice to several tissue-specific transgenic Cre-driver mouse strains including the EBV lytic promoter (ED-L2-Cre recombinase; “L”) to drive robust expression in the oropharyngeal squamous epithelia. Analysis of epithelial tissues isolated from LH mice displayed increased oral volumes and cutaneous epithelial thickening associated with hyperplasia, dysplasia, accumulation of neutrophils and recruitment of cytotoxic and regulatory T cells. A second cross to the keratin 14 promoter (KRT14-Cre; “K”) line enabled targeted expression to the basal epithelial layer (KH) and confirmed increased oral volumes and cutaneous epithelial thickening associated with increased suprabasal proliferation and expression of canonical E7 targets. Lastly, a third cross to a tamoxifen-inducible Cre (CreERT2) line driven by the keratin 14 promoter (KRT14-CreERT2; “iK”) enabled conditional and inducible post-natal E6 and E7 expression in basal epithelia (iKH). To prevent systemic oncogene expression in K14+ cells, we validated a method for submucosal delivery of tamoxifen to the tongue. We confirmed that this approach prevents tamoxifen spread and anatomically restricts activation oncogene expression to intra-lingual regions using an optical reporter mouse recently generated by the Amelio lab (Rosa26-loxP-STOP-loxP-LumiFluor; “F”) crossed to the iK line (“iKF”). Notably, direct intra-lingual injection yields higher recombination and reporter activation using 40% less overall tamoxifen than traditional i.p. administration routes. Moreover, immunofluorescent staining revealed mosaic transgene activation in the oropharynx, an important feature for modeling HPV-induced OSCC. In fact, intra-lingual injection of tamoxifen in iKH mice coupled with 4-nitroquniloline 1-oxide (4NQO) administration led to E6 and E7 expression and dysplasia in the tongue epithelia. Ongoing studies are evaluating cooperating mutations in driving HNSCC development. Citation Format: Miranda B. Carper, Scott Troutman, Kevin M. Byrd, Bethany Wagner, Erin C. Henry, Stephanie A. Montgomery, Scott E. Williams, Joseph L. Kissil, Antonio L. Amelio. Novel mouse models of high-risk HPV-related oral cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1032.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 3 ( 2014-02-01), p. 908-920
    Abstract: Myc oncoproteins induce genes driving aerobic glycolysis, including lactate dehydrogenase-A that generates lactate. Here, we report that Myc controls transcription of the lactate transporter SLC16A1/MCT1 and that elevated MCT1 levels are manifest in premalignant and neoplastic Eμ-Myc transgenic B cells and in human malignancies with MYC or MYCN involvement. Notably, disrupting MCT1 function leads to an accumulation of intracellular lactate that rapidly disables tumor cell growth and glycolysis, provoking marked alterations in glycolytic intermediates, reductions in glucose transport, and in levels of ATP, NADPH, and ultimately, glutathione (GSH). Reductions in GSH then lead to increases in hydrogen peroxide, mitochondrial damage, and ultimately, cell death. Finally, forcing glycolysis by metformin treatment augments this response and the efficacy of MCT1 inhibitors, suggesting an attractive combination therapy for MYC/MCT1-expressing malignancies. Cancer Res; 74(3); 908–20. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: ACS Synthetic Biology, American Chemical Society (ACS), Vol. 9, No. 1 ( 2020-01-17), p. 1-9
    Type of Medium: Online Resource
    ISSN: 2161-5063 , 2161-5063
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2020
    detail.hit.zdb_id: 2644383-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 111, No. 32 ( 2014-08-12)
    Abstract: Chimeric oncoproteins created by chromosomal translocations are among the most common genetic mutations associated with tumorigenesis. Malignant mucoepidermoid salivary gland tumors, as well as a growing number of solid epithelial-derived tumors, can arise from a recurrent t (11, 19)(q21;p13.1) translocation that generates an unusual chimeric cAMP response element binding protein (CREB)-regulated transcriptional coactivator 1 (CRTC1)/mastermind-like 2 (MAML2) (C1/M2) oncoprotein comprised of two transcriptional coactivators, the CRTC1 and the NOTCH/RBPJ coactivator MAML2. Accordingly, the C1/M2 oncoprotein induces aberrant expression of CREB and NOTCH target genes. Surprisingly, here we report a gain-of-function activity of the C1/M2 oncoprotein that directs its interactions with myelocytomatosis oncogene (MYC) proteins and the activation of MYC transcription targets, including those involved in cell growth and metabolism, survival, and tumorigenesis. These results were validated in human mucoepidermoid tumor cells that harbor the t (11, 19)(q21;p13.1) translocation and express the C1/M2 oncoprotein. Notably, the C1/M2–MYC interaction is necessary for C1/M2-driven cell transformation, and the C1/M2 transcriptional signature predicts other human malignancies having combined involvement of MYC and CREB . These findings suggest that such gain-of-function properties may also be manifest in other oncoprotein fusions found in human cancer and that agents targeting the C1/M2–MYC interface represent an attractive strategy for the development of effective and safe anticancer therapeutics in tumors harboring the t (11, 19) translocation.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2014
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2450-2450
    Abstract: Metabolic reprogramming is a hallmark of cancer. MYC oncoproteins control many aspects of this response, by inducing the expression of genes involved in mitochondrial biogenesis, glycolysis, glutaminolysis and amino acid transport. This coordinated response allows cancer cells to meet the demands for macromolecules and energy necessary to sustain the anabolic state. Normal cells adapt to nutrient-limiting conditions, such as amino acid (AA) starvation, by activating the autophagy-lysosomal pathway that is necessary for the maintenance of amino acid pools and for providing other building blocks (e.g., ATP) that are needed for cell survival. Surprisingly, our ex vivo and in vivo studies of premalignant and neoplastic MYC-expressing B cells of Eμ-Myc transgenic mice, and of human MYC-driven B cell lymphoma (e.g., Burkitt lymphoma), revealed that MYC suppresses the catabolic autophagy-lysosomal pathway, and that, accordingly, Myc-expressing premalignant and neoplastic B cells are exquisitely sensitive to AA starvation. For example, analyses of the effects of low (6%) versus high (20%) protein diets revealed that limiting AA pools in vivo selectively reduces the numbers of circulating premalignant Eμ-Myc B220+ B cells without affecting B cell numbers in wild type littermate mice. Thus, MYC-driven tumor cells are unable to sufficiently adapt to a state of nutrient deprivation (Figure 1). Expression analyses revealed that this MYC suppresses the autophagy-lysosomal system by transcriptionally repressing genes that encode regulators and components of this pathway, and that this response is a hallmark of human malignancies with MYC involvement. Further, suppressing these genes has functional consequences, where MYC provokes marked reductions in autophagic flux that lead to marked increases in the levels of cargo such as p62/Sequestrin that are normally degraded by this pathway. A master regulator of autophagy and lysosomal biogenesis is TFEB that, like MYC, functions as a basic helix-loop-helix leucine zipper transcription factor and shares a similar DNA recognition sequence. Our studies suggest that MYC blocks TFEB function at three levels. First, MYC can directly repress TFEB transcription. Second, MYC can directly repress TFEB transcription targets by competing with TFEB for binding to the promoter-regulatory regions of autophagy-lysosome gene targets. Third, MYC-expressing B cells have activated mTORC1, which phosphorylates TFEB and blocks its nuclear localization. Notably, forced reactivation of the autophagy-lysosomal pathway via inducible expression of a of a constitutively active (mTORC1-resistant and nuclear) form of TFEB (TFEBS211A) disables the malignant state, where TFEBS211A triggers cell cycle arrest and senescence of both mouse and human MYC-driven lymphomas ex vivo, and compromises tumorigenic potential in vivo. Thus, TFEB acts as tumor suppressor for MYC-driven malignancies. We hypothesized that MYC-driven tumor cells compensate for the reductions in the autophagy pathway and maintain AA homeostasis by activating compensatory mechanisms, including AA transport and the proteasome. In support of this notion, the expression of AA transporters and components of the proteasome, and AA transport and proteasome activity, are markedly augmented in premalignant and neoplastic MYC-expressing B cells. Accordingly, MYC-expressing B cells are exquisitely sensitive to treatment with proteasome inhibitors. Collectively, these findings suggest that MYC drives the anabolic state by suppressing the catabolic autophagy-lysosomal pathway, and that to maintain AA pools MYC-driven cancer cells up-regulate AA transport and the proteasome. This scenario provides attractive opportunities for combination therapies that should disable MYC-driven malignancies, including protein-restricted diets and proteasome and TORC1. Figure 1. Premalignant Eμ-Myc and wild type (WT) littermates were treated for one week with low (6%) and high (20%) protein diets and B220+ cell numbers in peripheral blood were assessed. Figure 1. Premalignant Eμ-Myc and wild type (WT) littermates were treated for one week with low (6%) and high (20%) protein diets and B220+ cell numbers in peripheral blood were assessed. 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: 2015
    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...