ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • Mice  (23)
  • CTM2008-04517; CTM2009-06269-E; CTM2010-11955-E/MAR; DATE/TIME; EMSODEV; EMSO implementation and operation: DEVelopment of instrument module; EMSO-Link; ES5110020; Fixed Point Open Ocean Observatories Network; FixO3; Humidity, relative; Implementation of the Strategy to Ensure the EMSO ERICs Long-term Sustainability; Instalació de sensors en observatorio submari expansible OBSEA; Instalacion de equipos en superfície para la ampliación de las prestaciones del observatorio submarino obsea; Instrumentación y tecnologías aplicadas al estudio, caracterización y explotación sostenible del medio marino; Integración de sensores para monitorización submarina en una red con sincronización temporal; Interoperabilidad e instrumentación de plataformas autónomas marinas para la monitorización sísmica; Interoperabilidad en redes de sensores marinos y ambientales; Interoperabilitat en xarxes de sensors marins i ambientals; INTIMAS; INTMARSIS; JERICO-NEXT; Joint European Research Infrastructure network for Coastal Observatory – Novel European eXpertise for coastal observaTories; MarInTech; MELOA; MeteoMet2; Metrology for essential climate variables; Multi-purpose/Multi-sensor Extra Light Oceanography Apparatua; NeXOS; Next generation, Cost-effective, Compact, Multifunctional Web Enabled Ocean Sensor Systems Empowering Marine, Maritime and Fisheries Management; Nuevas tecnologías para el seguimiento de la acidificación marina en el espacio natural Costa del Garraf; OBSEA:UPC:2009-05; Operatividad laboratorio submarino obsea; OPOBSEA; Precipitation; Pressure, atmospheric; PT2009-0080; Redes de sensores submarinos acústicos aplicados al seguimiento de especies de interes comercial; Redes de sensores submarinos autónomos y cableados aplicados a la monitorización remota de indicadores biológicos; RESBAC; RESBIO; SARTI-MAR; SINEOS; Sismòmetre marí digital amb connexió per cable; Sistemas inalambricos para la extension de observatorios submarinos; Sistemes dAdquisició Remota de dades i Tractament de la Informació en el Medi Marí; Temperature, air; VALTEC09-1-0059; Weather Station, Vantage Pro2, Davis; Wind direction; Wind speed
  • 2010-2014  (23)
Collection
Keywords
Years
Year
  • 1
    Publication Date: 2010-08-06
    Description: Long interspersed element-1 (LINE-1 or L1) retrotransposition continues to affect human genome evolution. L1s can retrotranspose in the germline, during early development and in select somatic cells; however, the host response to L1 retrotransposition remains largely unexplored. Here we show that reporter genes introduced into the genome of various human embryonic carcinoma-derived cell lines (ECs) by L1 retrotransposition are rapidly and efficiently silenced either during or immediately after their integration. Treating ECs with histone deacetylase inhibitors rapidly reverses this silencing, and chromatin immunoprecipitation experiments revealed that reactivation of the reporter gene was correlated with changes in chromatin status at the L1 integration site. Under our assay conditions, rapid silencing was also observed when reporter genes were delivered into ECs by mouse L1s and a zebrafish LINE-2 element, but not when similar reporter genes were delivered into ECs by Moloney murine leukaemia virus or human immunodeficiency virus, suggesting that these integration events are silenced by distinct mechanisms. Finally, we demonstrate that subjecting ECs to culture conditions that promote differentiation attenuates the silencing of reporter genes delivered by L1 retrotransposition, but that differentiation, in itself, is not sufficient to reactivate previously silenced reporter genes. Thus, our data indicate that ECs differ from many differentiated cells in their ability to silence reporter genes delivered by L1 retrotransposition.〈br /〉〈br /〉〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3034402/" target="_blank"〉〈img src="https://static.pubmed.gov/portal/portal3rc.fcgi/4089621/img/3977009" border="0"〉〈/a〉   〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3034402/" target="_blank"〉This paper as free author manuscript - peer-reviewed and accepted for publication〈/a〉〈br /〉〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Garcia-Perez, Jose L -- Morell, Maria -- Scheys, Joshua O -- Kulpa, Deanna A -- Morell, Santiago -- Carter, Christoph C -- Hammer, Gary D -- Collins, Kathleen L -- O'Shea, K Sue -- Menendez, Pablo -- Moran, John V -- 5 P30 CA46592/CA/NCI NIH HHS/ -- GM-069985/GM/NIGMS NIH HHS/ -- GM060518/GM/NIGMS NIH HHS/ -- GM082970/GM/NIGMS NIH HHS/ -- NS-048187/NS/NINDS NIH HHS/ -- R01 DK62027/DK/NIDDK NIH HHS/ -- R01 GM060518/GM/NIGMS NIH HHS/ -- R01 GM060518-12/GM/NIGMS NIH HHS/ -- R01 GM082970/GM/NIGMS NIH HHS/ -- R01 GM082970-04/GM/NIGMS NIH HHS/ -- R01AI051198/AI/NIAID NIH HHS/ -- T32-GM08322/GM/NIGMS NIH HHS/ -- Howard Hughes Medical Institute/ -- England -- Nature. 2010 Aug 5;466(7307):769-73. doi: 10.1038/nature09209.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Department of Human Genetics, 1241 East Catherine Street, University of Michigan Medical School, Ann Arbor, Michigan 48109-5618, USA. josel.garcia.perez@juntadeandalucia.es〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/20686575" target="_blank"〉PubMed〈/a〉
    Keywords: Animals ; Cell Differentiation/genetics/physiology ; Cell Line, Tumor ; Chromatin/drug effects/genetics/metabolism ; Chromatin Immunoprecipitation ; Embryonal Carcinoma Stem Cells/*metabolism/pathology ; Epigenesis, Genetic/drug effects/*genetics ; Female ; Gene Expression Regulation, Neoplastic/drug effects ; *Gene Silencing/drug effects ; Genes, Reporter/genetics ; Genetic Engineering ; Genetic Vectors/genetics ; Genome, Human/genetics ; HIV/genetics ; Histone Deacetylase Inhibitors/pharmacology ; Humans ; Long Interspersed Nucleotide Elements/genetics ; Male ; Mice ; Models, Genetic ; Moloney murine leukemia virus/genetics ; Retroelements/*genetics ; Zebrafish/genetics
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 2
    Publication Date: 2010-12-24
    Description: Cancer is a disease consisting of both genetic and epigenetic changes. Although increasing evidence demonstrates that tumour progression entails chromatin-mediated changes such as DNA methylation, the role of histone variants in cancer initiation and progression currently remains unclear. Histone variants replace conventional histones within the nucleosome and confer unique biological functions to chromatin. Here we report that the histone variant macroH2A (mH2A) suppresses tumour progression of malignant melanoma. Loss of mH2A isoforms, histone variants generally associated with condensed chromatin and fine-tuning of developmental gene expression programs, is positively correlated with increasing malignant phenotype of melanoma cells in culture and human tissue samples. Knockdown of mH2A isoforms in melanoma cells of low malignancy results in significantly increased proliferation and migration in vitro and growth and metastasis in vivo. Restored expression of mH2A isoforms rescues these malignant phenotypes in vitro and in vivo. We demonstrate that the tumour-promoting function of mH2A loss is mediated, at least in part, through direct transcriptional upregulation of CDK8. Suppression of CDK8, a colorectal cancer oncogene, inhibits proliferation of melanoma cells, and knockdown of CDK8 in cells depleted of mH2A suppresses the proliferative advantage induced by mH2A loss. Moreover, a significant inverse correlation between mH2A and CDK8 expression levels exists in melanoma patient samples. Taken together, our results demonstrate that mH2A is a critical component of chromatin that suppresses the development of malignant melanoma, a highly intractable cutaneous neoplasm.〈br /〉〈br /〉〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3057940/" target="_blank"〉〈img src="https://static.pubmed.gov/portal/portal3rc.fcgi/4089621/img/3977009" border="0"〉〈/a〉   〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3057940/" target="_blank"〉This paper as free author manuscript - peer-reviewed and accepted for publication〈/a〉〈br /〉〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Kapoor, Avnish -- Goldberg, Matthew S -- Cumberland, Lara K -- Ratnakumar, Kajan -- Segura, Miguel F -- Emanuel, Patrick O -- Menendez, Silvia -- Vardabasso, Chiara -- Leroy, Gary -- Vidal, Claudia I -- Polsky, David -- Osman, Iman -- Garcia, Benjamin A -- Hernando, Eva -- Bernstein, Emily -- 5P30CA016087-27/CA/NCI NIH HHS/ -- CA109388/CA/NCI NIH HHS/ -- R21 CA150117/CA/NCI NIH HHS/ -- R21 CA150117-01/CA/NCI NIH HHS/ -- R21 CA150117-02/CA/NCI NIH HHS/ -- R21CA150117/CA/NCI NIH HHS/ -- England -- Nature. 2010 Dec 23;468(7327):1105-9. doi: 10.1038/nature09590.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Department of Oncological Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, New York 10029, USA.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/21179167" target="_blank"〉PubMed〈/a〉
    Keywords: Animals ; Cell Line, Tumor ; Cell Movement ; Cell Proliferation ; Cyclin-Dependent Kinase 8/*metabolism ; Disease Progression ; Gene Expression Profiling ; *Gene Expression Regulation, Neoplastic ; Gene Knockdown Techniques ; HCT116 Cells ; Histones/deficiency/genetics/*metabolism ; Humans ; Melanoma/*pathology/physiopathology ; Melanoma, Experimental ; Mice ; Mice, Inbred BALB C ; Mice, Inbred C57BL ; Neoplasm Metastasis/*pathology/physiopathology ; Rats ; Up-Regulation
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2010-10-12
    Description: With the capacity for rapid self-renewal and regeneration, the intestinal epithelium is stereotypical of stem cell-supported tissues. Yet the pattern of stem cell turnover remains in question. Applying analytical methods from population dynamics and statistical physics to an inducible genetic labeling system, we showed that clone size distributions conform to a distinctive scaling behavior at short times. This result demonstrates that intestinal stem cells form an equipotent population in which the loss of a stem cell is compensated by the multiplication of a neighbor, leading to neutral drift dynamics in which clones expand and contract at random until they either take over the crypt or they are lost. Combined with long-term clonal fate data, we show that the rate of stem cell replacement is comparable to the cell division rate, implying that neutral drift and symmetrical cell divisions are central to stem cell homeostasis.〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Lopez-Garcia, Carlos -- Klein, Allon M -- Simons, Benjamin D -- Winton, Douglas J -- G0800784/Medical Research Council/United Kingdom -- Cancer Research UK/United Kingdom -- New York, N.Y. -- Science. 2010 Nov 5;330(6005):822-5. doi: 10.1126/science.1196236. Epub 2010 Sep 23.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/20929733" target="_blank"〉PubMed〈/a〉
    Keywords: Animals ; Cell Differentiation ; *Cell Division ; Cell Lineage ; Clone Cells/cytology ; Colon/cytology ; Homeostasis ; Intestinal Mucosa/*cytology ; Intestine, Small/cytology ; Mice ; Stem Cells/*cytology/physiology
    Print ISSN: 0036-8075
    Electronic ISSN: 1095-9203
    Topics: Biology , Chemistry and Pharmacology , Computer Science , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2013-02-22
    Description: Pathogens often infect hosts through collective actions: they secrete growth-promoting compounds or virulence factors, or evoke host reactions that fuel the colonization of the host. Such behaviours are vulnerable to the rise of mutants that benefit from the collective action without contributing to it; how these behaviours can be evolutionarily stable is not well understood. We address this question using the intestinal pathogen Salmonella enterica serovar Typhimurium (hereafter termed S. typhimurium), which manipulates its host to induce inflammation, and thereby outcompetes the commensal microbiota. Notably, the virulence factors needed for host manipulation are expressed in a bistable fashion, leading to a slow-growing subpopulation that expresses virulence genes, and a fast-growing subpopulation that is phenotypically avirulent. Here we show that the expression of the genetically identical but phenotypically avirulent subpopulation is essential for the evolutionary stability of virulence in this pathogen. Using a combination of mathematical modelling, experimental evolution and competition experiments we found that within-host evolution leads to the emergence of mutants that are genetically avirulent and fast-growing. These mutants are defectors that exploit inflammation without contributing to it. In infection experiments initiated with wild-type S. typhimurium, defectors increase only slowly in frequency. In a genetically modified S. typhimurium strain in which the phenotypically avirulent subpopulation is reduced in size, defectors rise more rapidly, inflammation ceases prematurely, and S. typhimurium is quickly cleared from the gut. Our results establish that host manipulation by S. typhimurium is a cooperative trait that is vulnerable to the rise of avirulent defectors; the expression of a phenotypically avirulent subpopulation that grows as fast as defectors slows down this process, and thereby promotes the evolutionary stability of virulence. This points to a key role of bistable virulence gene expression in stabilizing cooperative virulence and may lead the way to new approaches for controlling pathogens.〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Diard, Mederic -- Garcia, Victor -- Maier, Lisa -- Remus-Emsermann, Mitja N P -- Regoes, Roland R -- Ackermann, Martin -- Hardt, Wolf-Dietrich -- England -- Nature. 2013 Feb 21;494(7437):353-6. doi: 10.1038/nature11913.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Institute of Microbiology, ETH Zurich, Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/23426324" target="_blank"〉PubMed〈/a〉
    Keywords: Animals ; *Biological Evolution ; Host-Pathogen Interactions ; Inflammation/microbiology/pathology ; Intestines/microbiology ; Mice ; Mice, Inbred C57BL ; Mutation ; *Phenotype ; Salmonella Infections/microbiology/prevention & control/transmission ; Salmonella typhimurium/genetics/growth & development/*pathogenicity ; Virulence/genetics/physiology ; Virulence Factors/genetics/metabolism
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2014-08-27
    Description: Aberrant activation of oncogenes or loss of tumour suppressor genes opposes malignant transformation by triggering a stable arrest in cell growth, which is termed cellular senescence. This process is finely tuned by both cell-autonomous and non-cell-autonomous mechanisms that regulate the entry of tumour cells to senescence. Whether tumour-infiltrating immune cells can oppose senescence is unknown. Here we show that at the onset of senescence, PTEN null prostate tumours in mice are massively infiltrated by a population of CD11b(+)Gr-1(+) myeloid cells that protect a fraction of proliferating tumour cells from senescence, thus sustaining tumour growth. Mechanistically, we found that Gr-1(+) cells antagonize senescence in a paracrine manner by interfering with the senescence-associated secretory phenotype of the tumour through the secretion of interleukin-1 receptor antagonist (IL-1RA). Strikingly, Pten-loss-induced cellular senescence was enhanced in vivo when Il1ra knockout myeloid cells were adoptively transferred to PTEN null mice. Therapeutically, docetaxel-induced senescence and efficacy were higher in PTEN null tumours when the percentage of tumour-infiltrating CD11b(+)Gr-1(+) myeloid cells was reduced using an antagonist of CXC chemokine receptor 2 (CXCR2). Taken together, our findings identify a novel non-cell-autonomous network, established by innate immunity, that controls senescence evasion and chemoresistance. Targeting this network provides novel opportunities for cancer therapy.〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Di Mitri, Diletta -- Toso, Alberto -- Chen, Jing Jing -- Sarti, Manuela -- Pinton, Sandra -- Jost, Tanja Rezzonico -- D'Antuono, Rocco -- Montani, Erica -- Garcia-Escudero, Ramon -- Guccini, Ilaria -- Da Silva-Alvarez, Sabela -- Collado, Manuel -- Eisenberger, Mario -- Zhang, Zhe -- Catapano, Carlo -- Grassi, Fabio -- Alimonti, Andrea -- England -- Nature. 2014 Nov 6;515(7525):134-7. doi: 10.1038/nature13638. Epub 2014 Aug 24.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉1] Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona CH6500, Switzerland [2]. ; 1] Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona CH6500, Switzerland [2] Faculty of Biology and Medicine, University of Lausanne UNIL, Lausanne CH1011, Switzerland. ; Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona CH6500, Switzerland. ; Institute for Research in Biomedicine (IRB), Bellinzona CH6500, Switzerland. ; 1] Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona CH6500, Switzerland [2] Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain. ; Laboratory of Stem Cells in Cancer and Aging, (stemCHUS) Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), E15706 Santiago de Compostela, Spain. ; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland 21231-1000, USA. ; Divisions of BioStatistics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland 21231-1000, USA. ; 1] Institute for Research in Biomedicine (IRB), Bellinzona CH6500, Switzerland [2] Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan I-20100, Italy.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/25156255" target="_blank"〉PubMed〈/a〉
    Keywords: Animals ; *Cell Aging/drug effects ; *Cell Movement ; Disease Progression ; Drug Resistance, Neoplasm ; Humans ; Immunity, Innate ; Interleukin 1 Receptor Antagonist Protein/deficiency/metabolism/secretion ; Interleukin-1alpha/immunology/metabolism ; Male ; Mice ; Myeloid Cells/*cytology/*metabolism/transplantation ; PTEN Phosphohydrolase/deficiency/genetics/metabolism ; Prostatic Neoplasms/drug therapy/immunology/metabolism/*pathology ; Receptors, Chemokine/*metabolism ; Receptors, Interleukin-8B/antagonists & inhibitors ; Taxoids/pharmacology ; Tumor Escape ; Tumor Microenvironment
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
    Publication Date: 2014-05-23
    Description: Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.〈br /〉〈br /〉〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4122675/" target="_blank"〉〈img src="https://static.pubmed.gov/portal/portal3rc.fcgi/4089621/img/3977009" border="0"〉〈/a〉   〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4122675/" target="_blank"〉This paper as free author manuscript - peer-reviewed and accepted for publication〈/a〉〈br /〉〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Mazur, Pawel K -- Reynoird, Nicolas -- Khatri, Purvesh -- Jansen, Pascal W T C -- Wilkinson, Alex W -- Liu, Shichong -- Barbash, Olena -- Van Aller, Glenn S -- Huddleston, Michael -- Dhanak, Dashyant -- Tummino, Peter J -- Kruger, Ryan G -- Garcia, Benjamin A -- Butte, Atul J -- Vermeulen, Michiel -- Sage, Julien -- Gozani, Or -- DP2 OD007447/OD/NIH HHS/ -- R01 CA172560/CA/NCI NIH HHS/ -- T32 GM007276/GM/NIGMS NIH HHS/ -- U19 AI109662/AI/NIAID NIH HHS/ -- England -- Nature. 2014 Jun 12;510(7504):283-7. doi: 10.1038/nature13320. Epub 2014 May 21.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉1] Department of Pediatrics, Stanford University School of Medicine, California 94305, USA [2] Department of Genetics, Stanford University School of Medicine, California 94305, USA [3]. ; 1] Department of Biology, Stanford University, California 94305, USA [2]. ; Institute for Immunity, Transplantation and Infection, and Department of Medicine, Stanford University School of Medicine, California 94305, USA. ; Department of Molecular Cancer Research and Department of Medical Oncology, University Medical Center Utrecht, 3508 AB Utrecht, The Netherlands. ; Department of Biology, Stanford University, California 94305, USA. ; Epigenetics Program and Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA. ; Cancer Epigenetics DPU, Oncology R&D, GlaxoSmithKline, Collegeville, Pennsylvania 19426 USA. ; 1] Cancer Epigenetics DPU, Oncology R&D, GlaxoSmithKline, Collegeville, Pennsylvania 19426 USA [2] Janssen Research and Development, 1400 McKean Road, Spring House, Pennsylvania 19477, USA (D.D.); Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525GA Nijmegen, The Netherlands (M.V.). ; 1] Department of Pediatrics, Stanford University School of Medicine, California 94305, USA [2] Department of Genetics, Stanford University School of Medicine, California 94305, USA. ; 1] Department of Molecular Cancer Research and Department of Medical Oncology, University Medical Center Utrecht, 3508 AB Utrecht, The Netherlands [2] Janssen Research and Development, 1400 McKean Road, Spring House, Pennsylvania 19477, USA (D.D.); Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, 6525GA Nijmegen, The Netherlands (M.V.).〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/24847881" target="_blank"〉PubMed〈/a〉
    Keywords: Adenocarcinoma/enzymology/genetics/metabolism/pathology ; Animals ; Cell Line, Tumor ; Cell Transformation, Neoplastic/genetics/*metabolism/pathology ; Disease Models, Animal ; Histone-Lysine N-Methyltransferase/*metabolism ; Humans ; Lung Neoplasms/enzymology/genetics/metabolism/pathology ; Lysine/*metabolism ; MAP Kinase Kinase Kinase 2/chemistry/*metabolism ; MAP Kinase Kinase Kinases/chemistry/*metabolism ; Methylation ; Mice ; Mitogen-Activated Protein Kinases/metabolism ; Oncogene Protein p21(ras)/genetics/*metabolism ; Pancreatic Neoplasms/enzymology/genetics/metabolism/pathology ; Protein Phosphatase 2/antagonists & inhibitors/metabolism ; Proto-Oncogene Proteins A-raf/metabolism ; Signal Transduction
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 7
    Publication Date: 2014-02-14
    Description: Regeneration of skeletal muscle depends on a population of adult stem cells (satellite cells) that remain quiescent throughout life. Satellite cell regenerative functions decline with ageing. Here we report that geriatric satellite cells are incapable of maintaining their normal quiescent state in muscle homeostatic conditions, and that this irreversibly affects their intrinsic regenerative and self-renewal capacities. In geriatric mice, resting satellite cells lose reversible quiescence by switching to an irreversible pre-senescence state, caused by derepression of p16(INK4a) (also called Cdkn2a). On injury, these cells fail to activate and expand, undergoing accelerated entry into a full senescence state (geroconversion), even in a youthful environment. p16(INK4a) silencing in geriatric satellite cells restores quiescence and muscle regenerative functions. Our results demonstrate that maintenance of quiescence in adult life depends on the active repression of senescence pathways. As p16(INK4a) is dysregulated in human geriatric satellite cells, these findings provide the basis for stem-cell rejuvenation in sarcopenic muscles.〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Sousa-Victor, Pedro -- Gutarra, Susana -- Garcia-Prat, Laura -- Rodriguez-Ubreva, Javier -- Ortet, Laura -- Ruiz-Bonilla, Vanessa -- Jardi, Merce -- Ballestar, Esteban -- Gonzalez, Susana -- Serrano, Antonio L -- Perdiguero, Eusebio -- Munoz-Canoves, Pura -- England -- Nature. 2014 Feb 20;506(7488):316-21. doi: 10.1038/nature13013. Epub 2014 Feb 12.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉1] Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, CIBER on Neurodegenerative diseases, E-08003 Barcelona, Spain [2] Buck Institute for Research on Aging, Novato, California 94945, USA. ; 1] Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, CIBER on Neurodegenerative diseases, E-08003 Barcelona, Spain [2]. ; Chromatin and Disease Group, Cancer Epigenetics and Biology Programme, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, E-08907 Barcelona, Spain. ; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, CIBER on Neurodegenerative diseases, E-08003 Barcelona, Spain. ; Stem Cell Aging Group, Centro Nacional de Investigaciones Cardiovasculares, E-28029 Madrid, Spain. ; 1] Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University, CIBER on Neurodegenerative diseases, E-08003 Barcelona, Spain [2] Institucio Catalana de Recerca i Estudis Avancats, E-08010 Barcelona, Spain.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/24522534" target="_blank"〉PubMed〈/a〉
    Keywords: Adult ; Aging/*metabolism ; Animals ; Cells, Cultured ; Cyclin-Dependent Kinase Inhibitor p16/deficiency/genetics/*metabolism ; E2F1 Transcription Factor/metabolism ; Humans ; Male ; Mice ; Mice, Inbred C57BL ; Progeria/metabolism/pathology ; Regeneration ; Rejuvenation ; Retinoblastoma Protein/metabolism ; Satellite Cells, Skeletal Muscle/*cytology/*metabolism ; Young Adult
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 8
    Publication Date: 2011-04-05
    Description: Electrical activity has been shown to regulate development in a variety of species and in various structures, including the retina, spinal cord and cortex. Within the mammalian cortex specifically, the development of dendrites and commissural axons in pyramidal cells is activity-dependent. However, little is known about the developmental role of activity in the other major cortical population of neurons, the GABA-producing interneurons. These neurons are morphologically and functionally heterogeneous and efforts over the past decade have focused on determining the mechanisms that contribute to this diversity. It was recently discovered that 30% of all cortical interneurons arise from a relatively novel source within the ventral telencephalon, the caudal ganglionic eminence (CGE). Owing to their late birth date, these interneurons populate the cortex only after the majority of other interneurons and pyramidal cells are already in place and have started to functionally integrate. Here we demonstrate in mice that for CGE-derived reelin (Re)-positive and calretinin (Cr)-positive (but not vasoactive intestinal peptide (VIP)-positive) interneurons, activity is essential before postnatal day 3 for correct migration, and that after postnatal day 3, glutamate-mediated activity controls the development of their axons and dendrites. Furthermore, we show that the engulfment and cell motility 1 gene (Elmo1), a target of the transcription factor distal-less homeobox 1 (Dlx1), is selectively expressed in Re(+) and Cr(+) interneurons and is both necessary and sufficient for activity-dependent interneuron migration. Our findings reveal a selective requirement for activity in shaping the cortical integration of specific neuronal subtypes.〈br /〉〈br /〉〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3641515/" target="_blank"〉〈img src="https://static.pubmed.gov/portal/portal3rc.fcgi/4089621/img/3977009" border="0"〉〈/a〉   〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3641515/" target="_blank"〉This paper as free author manuscript - peer-reviewed and accepted for publication〈/a〉〈br /〉〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉De Marco Garcia, Natalia V -- Karayannis, Theofanis -- Fishell, Gord -- 2R01MH071679-09/MH/NIMH NIH HHS/ -- 5R01MH068469-08/MH/NIMH NIH HHS/ -- 5R01NS039007-1/NS/NINDS NIH HHS/ -- R01 MH071679/MH/NIMH NIH HHS/ -- R01 MH071679-01A1/MH/NIMH NIH HHS/ -- R01 MH071679-07/MH/NIMH NIH HHS/ -- R01 NS032993/NS/NINDS NIH HHS/ -- R01 NS032993-05/NS/NINDS NIH HHS/ -- R01 NS039007/NS/NINDS NIH HHS/ -- R01 NS039007-10/NS/NINDS NIH HHS/ -- R01 NS039007-11/NS/NINDS NIH HHS/ -- England -- Nature. 2011 Apr 21;472(7343):351-5. doi: 10.1038/nature09865. Epub 2011 Apr 3.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Smilow Neuroscience Program, Departments of Cell Biology and Neural Science, New York University Langone Medical Center, New York, New York 10016, USA.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/21460837" target="_blank"〉PubMed〈/a〉
    Keywords: Adaptor Proteins, Signal Transducing/genetics/metabolism ; Animals ; Calbindin 2 ; Cell Adhesion Molecules, Neuronal/metabolism ; *Cell Movement/drug effects ; Cell Shape/drug effects ; Cerebral Cortex/*cytology ; Extracellular Matrix Proteins/metabolism ; Female ; Gene Expression Regulation ; Homeodomain Proteins/genetics/metabolism ; Interneurons/*cytology/drug effects/*metabolism ; Mice ; Nerve Tissue Proteins/metabolism ; Potassium Channels, Inwardly Rectifying/genetics/metabolism ; Pregnancy ; Pyramidal Cells/cytology/metabolism ; Receptors, Ionotropic Glutamate/antagonists & inhibitors/metabolism ; S100 Calcium Binding Protein G/metabolism ; Serine Endopeptidases/metabolism ; Signal Transduction ; Transcription Factors/genetics/metabolism ; Vasoactive Intestinal Peptide/metabolism
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 9
    Publication Date: 2011-03-11
    Description: Activation of microglia and inflammation-mediated neurotoxicity are suggested to play a decisive role in the pathogenesis of several neurodegenerative disorders. Activated microglia release pro-inflammatory factors that may be neurotoxic. Here we show that the orderly activation of caspase-8 and caspase-3/7, known executioners of apoptotic cell death, regulate microglia activation through a protein kinase C (PKC)-delta-dependent pathway. We find that stimulation of microglia with various inflammogens activates caspase-8 and caspase-3/7 in microglia without triggering cell death in vitro and in vivo. Knockdown or chemical inhibition of each of these caspases hindered microglia activation and consequently reduced neurotoxicity. We observe that these caspases are activated in microglia in the ventral mesencephalon of Parkinson's disease (PD) and the frontal cortex of individuals with Alzheimer's disease (AD). Taken together, we show that caspase-8 and caspase-3/7 are involved in regulating microglia activation. We conclude that inhibition of these caspases could be neuroprotective by targeting the microglia rather than the neurons themselves.〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Burguillos, Miguel A -- Deierborg, Tomas -- Kavanagh, Edel -- Persson, Annette -- Hajji, Nabil -- Garcia-Quintanilla, Albert -- Cano, Josefina -- Brundin, Patrik -- Englund, Elisabet -- Venero, Jose L -- Joseph, Bertrand -- England -- Nature. 2011 Apr 21;472(7343):319-24. doi: 10.1038/nature09788. Epub 2011 Mar 9.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet, 171 76, Stockholm, Sweden.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/21389984" target="_blank"〉PubMed〈/a〉
    Keywords: Alzheimer Disease/enzymology/pathology ; Animals ; Caspase 3/deficiency/metabolism ; Caspase 7/deficiency/metabolism ; Caspase 8/genetics/metabolism ; Caspase Inhibitors ; Caspases/deficiency/*metabolism ; Cell Death/drug effects ; Cells, Cultured ; Dopamine/metabolism ; Enzyme Activation ; Frontal Lobe/enzymology/pathology ; Gene Knockdown Techniques ; Humans ; Lipopolysaccharides/pharmacology ; Mice ; Microglia/drug effects/*physiology ; Neostriatum/metabolism ; Neurotoxicity Syndromes/*enzymology/metabolism/*pathology ; Parkinson Disease/enzymology/pathology ; Protein Kinase C-delta/chemistry/metabolism ; Rats ; *Signal Transduction ; Substantia Nigra/enzymology/pathology ; Toll-Like Receptor 4/metabolism
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 10
    Publication Date: 2012-06-23
    Description: Sirtuin proteins regulate diverse cellular pathways that influence genomic stability, metabolism and ageing. SIRT7 is a mammalian sirtuin whose biochemical activity, molecular targets and physiological functions have been unclear. Here we show that SIRT7 is an NAD(+)-dependent H3K18Ac (acetylated lysine 18 of histone H3) deacetylase that stabilizes the transformed state of cancer cells. Genome-wide binding studies reveal that SIRT7 binds to promoters of a specific set of gene targets, where it deacetylates H3K18Ac and promotes transcriptional repression. The spectrum of SIRT7 target genes is defined in part by its interaction with the cancer-associated E26 transformed specific (ETS) transcription factor ELK4, and comprises numerous genes with links to tumour suppression. Notably, selective hypoacetylation of H3K18Ac has been linked to oncogenic transformation, and in patients is associated with aggressive tumour phenotypes and poor prognosis. We find that deacetylation of H3K18Ac by SIRT7 is necessary for maintaining essential features of human cancer cells, including anchorage-independent growth and escape from contact inhibition. Moreover, SIRT7 is necessary for a global hypoacetylation of H3K18Ac associated with cellular transformation by the viral oncoprotein E1A. Finally, SIRT7 depletion markedly reduces the tumorigenicity of human cancer cell xenografts in mice. Together, our work establishes SIRT7 as a highly selective H3K18Ac deacetylase and demonstrates a pivotal role for SIRT7 in chromatin regulation, cellular transformation programs and tumour formation in vivo.〈br /〉〈br /〉〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3412143/" target="_blank"〉〈img src="https://static.pubmed.gov/portal/portal3rc.fcgi/4089621/img/3977009" border="0"〉〈/a〉   〈a href="https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3412143/" target="_blank"〉This paper as free author manuscript - peer-reviewed and accepted for publication〈/a〉〈br /〉〈br /〉〈span class="detail_caption"〉Notes: 〈/span〉Barber, Matthew F -- Michishita-Kioi, Eriko -- Xi, Yuanxin -- Tasselli, Luisa -- Kioi, Mitomu -- Moqtaderi, Zarmik -- Tennen, Ruth I -- Paredes, Silvana -- Young, Nicolas L -- Chen, Kaifu -- Struhl, Kevin -- Garcia, Benjamin A -- Gozani, Or -- Li, Wei -- Chua, Katrin F -- 1018438-142/PHS HHS/ -- 3T32DK007217-36S1/DK/NIDDK NIH HHS/ -- DP2OD007447/OD/NIH HHS/ -- GM 30186/GM/NIGMS NIH HHS/ -- HG 4558/HG/NHGRI NIH HHS/ -- K08 AG028961/AG/NIA NIH HHS/ -- R01 AG028867/AG/NIA NIH HHS/ -- R01 GM030186/GM/NIGMS NIH HHS/ -- R01 GM079641/GM/NIGMS NIH HHS/ -- T32 CA009302/CA/NCI NIH HHS/ -- U01 DA025956/DA/NIDA NIH HHS/ -- U01 DA025956-01/DA/NIDA NIH HHS/ -- U01DA025956/DA/NIDA NIH HHS/ -- England -- Nature. 2012 Jul 5;487(7405):114-8. doi: 10.1038/nature11043.〈br /〉〈span class="detail_caption"〉Author address: 〈/span〉Department of Biology, Stanford University, Stanford, California 94305, USA.〈br /〉〈span class="detail_caption"〉Record origin:〈/span〉 〈a href="http://www.ncbi.nlm.nih.gov/pubmed/22722849" target="_blank"〉PubMed〈/a〉
    Keywords: Acetylation ; Adenovirus E1A Proteins/genetics/metabolism ; Animals ; Base Sequence ; Binding Sites ; Cell Line, Tumor ; Cell Proliferation ; Cell Transformation, Neoplastic/genetics/*metabolism/pathology ; Chromatin/metabolism ; Contact Inhibition ; Disease Progression ; Histone Deacetylases/*metabolism ; Histones/*metabolism ; Humans ; Lysine/*metabolism ; Mice ; Neoplasm Transplantation ; Nucleotide Motifs ; Phenotype ; Promoter Regions, Genetic ; Repressor Proteins/metabolism ; Sirtuins/deficiency/genetics/*metabolism ; Transcription, Genetic ; Transplantation, Heterologous ; ets-Domain Protein Elk-4/metabolism
    Print ISSN: 0028-0836
    Electronic ISSN: 1476-4687
    Topics: Biology , Chemistry and Pharmacology , Medicine , Natural Sciences in General , Physics
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...