Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Hepatol ; 75(2): 271-274, 2021 08.
Article in English | MEDLINE | ID: mdl-34062178

ABSTRACT

BACKGROUND & AIMS: Published research promoted on twitter reaches more readers. Tweets with graphics are more engaging than those without. However, data are limited regarding how to optimize multimedia tweets for engagement. METHODS: The "Three facts and a Story" trial is a randomized-controlled trial comparing a tweet featuring a graphical abstract to paired tweets featuring the personal motivations behind the research and a summary of the findings. Fifty-four studies published by the Journal of Hepatology were randomized at the time of online publication. The primary endpoint was assessed at 28-days from online publication with a primary outcome of full-text downloads from the website. Secondary outcomes included page views and twitter engagement including impressions, likes, and retweets. RESULTS: Overall, 31 studies received standard tweets and 23 received story tweets. Five studies were randomized to story tweets but crossed over to standard tweets for lack of author participation. Most papers tweeted were original articles (94% standard, 91% story) and clinical topics (55% standard, 61% story). Story tweets were associated with a significant increase in the number of full text downloads, 51 (34-71) vs. 25 (13-41), p = 0.002. There was also a non-significant increase in the number of page views. Story tweets generated an average of >1,000 more impressions than standard tweets (5,388 vs. 4,280, p = 0.002). Story tweets were associated with a similar number of retweets, and a non-significant increase in the number of likes. CONCLUSION: Tweets featuring the authors and their motivations may increase engagement with published research.


Subject(s)
Information Dissemination/methods , Social Media/standards , Humans , Social Media/instrumentation , Social Media/statistics & numerical data
3.
Mol Cell Biol ; 32(22): 4523-33, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22949508

ABSTRACT

The ability to generate appropriate defense responses is crucial for the survival of an organism exposed to pathogenesis-inducing insults. However, the mechanisms that allow tissues and organs to cope with such stresses are poorly understood. Here we show that caspase-3-knockout mice or caspase inhibitor-treated mice were defective in activating the antiapoptotic Akt kinase in response to various chemical and environmental stresses causing sunburns, cardiomyopathy, or colitis. Defective Akt activation in caspase-3-knockout mice was accompanied by increased cell death and impaired survival in some cases. Mice homozygous for a mutation in RasGAP that prevents its cleavage by caspase-3 exhibited a similar defect in Akt activation, leading to increased apoptosis in stressed organs, marked deterioration of their physiological functions, and stronger disease development. Our results provide evidence for the relevance of caspase-3 as a stress intensity sensor that controls cell fate by either initiating a RasGAP cleavage-dependent cell resistance program or a cell suicide response.


Subject(s)
Cardiomyopathies/enzymology , Caspase 3/genetics , Colitis/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Sunburn/enzymology , p120 GTPase Activating Protein/genetics , Animals , Base Sequence , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Caspase 3/deficiency , Cell Death/drug effects , Cell Death/radiation effects , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate , Doxorubicin , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hemodynamics , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Stress, Physiological , Sunburn/genetics , Ultraviolet Rays , p120 GTPase Activating Protein/antagonists & inhibitors , p120 GTPase Activating Protein/deficiency
4.
PLoS One ; 5(12): e15770, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21209834

ABSTRACT

The molecular chaperone Hsp90 has been found to be essential for viability in all tested eukaryotes, from the budding yeast to Drosophila. In mammals, two genes encode the two highly similar and functionally largely redundant isoforms Hsp90α and Hsp90ß. Although they are co-expressed in most if not all cells, their relative levels vary between tissues and during development. Since mouse embryos lacking Hsp90ß die at implantation, and despite the fact that Hsp90 inhibitors being tested as anti-cancer agents are relatively well tolerated, the organismic functions of Hsp90 in mammals remain largely unknown. We have generated mouse lines carrying gene trap insertions in the Hsp90α gene to investigate the global functions of this isoform. Surprisingly, mice without Hsp90α are apparently normal, with one major exception. Mutant male mice, whose Hsp90ß levels are unchanged, are sterile because of a complete failure to produce sperm. While the development of the male reproductive system appears to be normal, spermatogenesis arrests specifically at the pachytene stage of meiosis I. Over time, the number of spermatocytes and the levels of the meiotic regulators and Hsp90 interactors Hsp70-2, NASP and Cdc2 are reduced. We speculate that Hsp90α may be required to maintain and to activate these regulators and/or to disassemble the synaptonemal complex that holds homologous chromosomes together. The link between fertility and Hsp90 is further supported by our finding that an Hsp90 inhibitor that can cross the blood-testis barrier can partially phenocopy the genetic defects.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Pachytene Stage , Spermatocytes/cytology , Testis/metabolism , Animals , Blood-Testis Barrier , Disease Progression , Female , Male , Meiosis , Mice , Mutation , Phenotype , Spermatogenesis , Spermatozoa/physiology
5.
Diabetes ; 58(11): 2596-606, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19696184

ABSTRACT

OBJECTIVE: Our laboratory has previously established in vitro that a caspase-generated RasGAP NH(2)-terminal moiety, called fragment N, potently protects cells, including insulinomas, from apoptotic stress. We aimed to determine whether fragment N can increase the resistance of pancreatic beta-cells in a physiological setting. RESEARCH DESIGN AND METHODS: A mouse line, called rat insulin promoter (RIP)-N, was generated that bears a transgene containing the rat insulin promoter followed by the cDNA-encoding fragment N. The histology, functionality, and resistance to stress of RIP-N islets were then assessed. RESULTS: Pancreatic beta-cells of RIP-N mice express fragment N, activate Akt, and block nuclear factor kappaB activity without affecting islet cell proliferation or the morphology and cellular composition of islets. Intraperitoneal glucose tolerance tests revealed that RIP-N mice control their glycemia similarly as wild-type mice throughout their lifespan. Moreover, islets isolated from RIP-N mice showed normal glucose-induced insulin secretory capacities. They, however, displayed increased resistance to apoptosis induced by a series of stresses including inflammatory cytokines, fatty acids, and hyperglycemia. RIP-N mice were also protected from multiple low-dose streptozotocin-induced diabetes, and this was associated with reduced in vivo beta-cell apoptosis. CONCLUSIONS: Fragment N efficiently increases the overall resistance of beta-cells to noxious stimuli without interfering with the physiological functions of the cells. Fragment N and the pathway it regulates represent, therefore, a potential target for the development of antidiabetes tools.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Insulin-Secreting Cells/physiology , Peptide Fragments/genetics , ras GTPase-Activating Proteins/genetics , Animals , Apoptosis , Blood Glucose/metabolism , Brain/physiology , Brain/physiopathology , Cell Division/genetics , DNA, Complementary/genetics , Diabetes Mellitus, Experimental/physiopathology , Glucose Tolerance Test , Insulin/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Transgenic , Peptide Fragments/pharmacology , Promoter Regions, Genetic , Rats
6.
Biochem Biophys Res Commun ; 353(2): 463-8, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17188236

ABSTRACT

X-ray structures of two crystal forms of the Src homology 3 domain (SH3) of the Ras GTPase activating protein (RasGAP) were determined at 1.5 and 1.8A resolution. The overall structure comprises a single domain with two tightly packed beta-sheets linked by a short helical segment. An important motif for peptide binding in other SH3 domains is not conserved in RasGAP. The RasGAP SH3 domain forms dimers in the crystal structures, which may provide new functional insight. The dimer interface involves residues also present in a peptide previously identified as an apoptotic sensitizer of tumor cells.


Subject(s)
Models, Chemical , Models, Molecular , p120 GTPase Activating Protein/chemistry , p120 GTPase Activating Protein/ultrastructure , src Homology Domains , Amino Acid Sequence , Binding Sites , Computer Simulation , Crystallography , Dimerization , Molecular Sequence Data , Protein Binding , Protein Conformation
7.
J Biol Chem ; 280(38): 32835-42, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-16046410

ABSTRACT

Apoptosis of pancreatic beta cells is implicated in the onset of type 1 and type 2 diabetes. Consequently, strategies aimed at increasing the resistance of beta cells toward apoptosis could be beneficial in the treatment of diabetes. RasGAP, a regulator of Ras and Rho GTPases, is an atypical caspase substrate, since it inhibits, rather than favors, apoptosis when it is partially cleaved by caspase-3 at position 455. The antiapoptotic signal generated by the partial processing of RasGAP is mediated by the N-terminal fragment (fragment N) in a Ras-phosphatidylinositol 3-kinase-Akt-dependent, but NF-kappaB-independent, manner. Further cleavage of fragment N at position 157 abrogates its antiapoptotic properties. Here we demonstrate that an uncleavable form of fragment N activates Akt, represses NF-kappaB activity, and protects the conditionally immortalized pancreatic insulinoma betaTC-tet cell line against various insults, including exposure to genotoxins, trophic support withdrawal, and incubation with inflammatory cytokines. Fragment N also induced Akt activity and protection against cytokine-induced apoptosis in primary pancreatic islet cells. Fragment N did not alter insulin cell content and insulin secretion in response to glucose. These data indicate that fragment N protects beta cells without affecting their function. The pathways regulated by fragment N are therefore promising targets for antidiabetogenic therapy.


Subject(s)
Apoptosis , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , ras GTPase-Activating Proteins/chemistry , Animals , Caspase 3 , Caspases/metabolism , Cell Line , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , Cytokines/metabolism , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Inflammation , Insulin Secretion , Insulinoma/metabolism , Lentivirus/genetics , Mice , Microscopy, Fluorescence , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Rats, Wistar , Time Factors
8.
Mol Biol Cell ; 16(8): 3511-20, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15901831

ABSTRACT

RasGAP bears two caspase-3 cleavage sites that are used sequentially as caspase activity increases in cells. When caspase-3 is mildly activated, RasGAP is first cleaved at position 455. This leads to the production of an N-terminal fragment, called fragment N, that activates the Ras-PI3K-Akt pathway and that promotes cell survival. At higher caspase activity, RasGAP is further cleaved at position 157 generating two small N-terminal fragments named N1 and N2. We have now determined the contribution of this second cleavage event in the regulation of apoptosis using cells in which the wild-type RasGAP gene has been replaced by a cDNA encoding a RasGAP mutant that cannot be cleaved at position 157. Our results show that cleavage of fragment N at position 157 leads to a marked reduction in Akt activity. This is accompanied by efficient processing of caspase-3 that favors cell death in response to various apoptotic stimuli. In nontumorigenic cells, fragments N1 and N2 do not modulate apoptosis. Therefore, the role of the second caspase-mediated cleavage of RasGAP is to allow the inactivation of the antiapoptotic function of fragment N so that caspases are no longer hampered in their ability to kill cells.


Subject(s)
Apoptosis , Caspases/metabolism , Mutation/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Caspase 3 , Cell Line , Down-Regulation , Enzyme Activation , Fibroblasts , Mice , Mice, Knockout , ras GTPase-Activating Proteins/deficiency
9.
Mol Cell Biol ; 24(23): 10425-36, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542850

ABSTRACT

Tight control of apoptosis is required for proper development and maintenance of homeostasis in multicellular organisms. Cells can protect themselves from potentially lethal stimuli by expressing antiapoptotic factors, such as inhibitors of apoptosis, FLICE (caspase 8)-inhibitory proteins, and members of the Bcl2 family. Here, we describe a mechanism that allows cells to survive once executioner caspases have been activated. This mechanism relies on the partial cleavage of RasGAP by caspase 3 into an amino-terminal fragment called fragment N. Generation of this fragment leads to the activation of the antiapoptotic Akt kinase, preventing further amplification of caspase activity. Partial cleavage of RasGAP is required for cell survival under stress conditions because cells expressing an uncleavable RasGAP mutant cannot activate Akt, cannot prevent amplification of caspase 3 activity, and eventually undergo apoptosis. Executioner caspases therefore control the extent of their own activation by a feedback regulatory mechanism initiated by the partial cleavage of RasGAP that is crucial for cell survival under adverse conditions.


Subject(s)
Caspases/metabolism , ras GTPase-Activating Proteins/chemistry , Animals , Apoptosis , Blotting, Western , Caspase 3 , Caspases/chemistry , Cell Line , Cell Survival , Cells, Cultured , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Electroporation , Enzyme Activation , Fibroblasts/metabolism , Humans , Image Processing, Computer-Assisted , Jurkat Cells , Lentivirus/genetics , Mice , Models, Genetic , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , Stress, Physiological , Time Factors , Transfection , ras GTPase-Activating Proteins/metabolism
10.
Biochem Pharmacol ; 68(6): 1027-31, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15313397

ABSTRACT

Executioner caspases induce the biochemical and cellular changes characteristic of apoptosis. Activation of caspases is therefore regarded as "the kiss of death" resulting in the cell's demise. Recent reports indicate however that in some situations, caspase activation may induce other responses than apoptosis. These findings raise the question of how cells manage to counteract the killing activities of executioner caspases. Experiments performed in our laboratory have unraveled a mechanism that allows cells to survive in the presence of activated executioner caspases. This mechanism is based on the partial cleavage of RasGAP into an N-terminal fragment that activates the Ras-PI3K-Akt survival pathway. This protective pathway may be activated to allow cells to use executioner caspases for other purposes than inducing apoptosis.


Subject(s)
Apoptosis/physiology , Caspases/physiology , ras GTPase-Activating Proteins/physiology , Animals , Caspases/metabolism , Cell Survival/physiology , Enzyme Activation , Humans , Phosphatidylinositol 3-Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/therapeutic use , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...