Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
Nature ; 561(7723): 338-342, 2018 09.
Article in English | MEDLINE | ID: mdl-30185906

ABSTRACT

Meiotic recombination differs between males and females; however, when and how these differences are established is unknown. Here we identify extensive sex differences at the initiation of recombination by mapping hotspots of meiotic DNA double-strand breaks in male and female mice. Contrary to past findings in humans, few hotspots are used uniquely in either sex. Instead, grossly different recombination landscapes result from up to fifteen-fold differences in hotspot usage between males and females. Indeed, most recombination occurs at sex-biased hotspots. Sex-biased hotspots seem to be partly determined by chromosome structure, and DNA methylation, which is absent in females at the onset of meiosis, has a substantial role. Sex differences are also evident later in meiosis as the rate at which meiotic breaks are repaired as crossovers differs between males and females in distal regions. The suppression of distal crossovers may help to minimize age-related aneuploidy that arises owing to cohesion loss during dictyate arrest in females.


Subject(s)
Crossing Over, Genetic/genetics , Meiosis/genetics , Sex Characteristics , Animals , DNA Breaks, Double-Stranded , DNA Methylation/genetics , Female , Male , Mice
3.
Genes Dev ; 30(3): 266-80, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26833728

ABSTRACT

Meiotic recombination is required for the segregation of homologous chromosomes and is essential for fertility. In most mammals, the DNA double-strand breaks (DSBs) that initiate meiotic recombination are directed to a subset of genomic loci (hot spots) by sequence-specific binding of the PRDM9 protein. Rapid evolution of the DNA-binding specificity of PRDM9 and gradual erosion of PRDM9-binding sites by gene conversion will alter the recombination landscape over time. To better understand the evolutionary turnover of recombination hot spots and its consequences, we mapped DSB hot spots in four major subspecies of Mus musculus with different Prdm9 alleles and in their F1 hybrids. We found that hot spot erosion governs the preferential usage of some Prdm9 alleles over others in hybrid mice and increases sequence diversity specifically at hot spots that become active in the hybrids. As crossovers are disfavored at such hot spots, we propose that sequence divergence generated by hot spot turnover may create an impediment for recombination in hybrids, potentially leading to reduced fertility and, eventually, speciation.


Subject(s)
Biological Evolution , Genetic Speciation , Histone-Lysine N-Methyltransferase/metabolism , Mice/classification , Mice/genetics , Recombination, Genetic/genetics , Alleles , Animals , DNA Breaks, Double-Stranded , Histone-Lysine N-Methyltransferase/genetics , Hybridization, Genetic , Protein Binding
4.
Genes Dev ; 30(7): 871, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27036969

ABSTRACT

Due to a technical error in processing the figures in the above-mentioned article, Figures 3, A and B; 4B; 5B; and 6, A and C contained errors or missing elements. The errors have been corrected in both the PDF and full-text HTML files online.

5.
BMC Genomics ; 14: 493, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-23870400

ABSTRACT

BACKGROUND: Homologous recombination is the key process that generates genetic diversity and drives evolution. SPO11 protein triggers recombination by introducing DNA double stranded breaks at discreet areas of the genome called recombination hotspots. The hotspot locations are largely determined by the DNA binding specificity of the PRDM9 protein in human, mice and most other mammals. In budding yeast Saccharomyces cerevisae, which lacks a Prdm9 gene, meiotic breaks are formed opportunistically in the regions of accessible chromatin, primarily at gene promoters. The genome-wide distribution of hotspots in this organism can be altered by tethering Spo11 protein to Gal4 recognition sequences in the strain expressing Spo11 attached to the DNA binding domain of the Gal4 transcription factor. To establish whether similar re-targeting of meiotic breaks can be achieved in PRDM9-containing organisms we have generated a Gal4BD-Spo11 mouse that expresses SPO11 protein joined to the DNA binding domain of yeast Gal4. RESULTS: We have mapped the genome-wide distribution of the recombination initiation sites in the Gal4BD-Spo11 mice. More than two hundred of the hotspots in these mice were novel and were likely defined by Gal4BD, as the Gal4 consensus motif was clustered around the centers in these hotspots. Surprisingly, meiotic DNA breaks in the Gal4BD-Spo11 mice were significantly depleted near the ends of chromosomes. The effect is particularly striking at the pseudoautosomal region of the X and Y chromosomes - normally the hottest region in the genome. CONCLUSIONS: Our data suggest that specific, yet-unidentified factors influence the initiation of meiotic recombination at subtelomeric chromosomal regions.


Subject(s)
Alleles , Endodeoxyribonucleases/genetics , Recombination, Genetic/genetics , Telomere/genetics , Animals , Binding Sites , Chromosome Pairing/genetics , Cluster Analysis , DNA Breaks, Double-Stranded , Gene Knock-In Techniques , Genomics , Mice
6.
J Med Chem ; 50(15): 3465-81, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17591763

ABSTRACT

Diacylglycerol (DAG) lactones have provided a powerful platform for structural exploration of the interactions between ligands and the C1 domains of protein kinase C (PKC). In this study, we report that DAG-dioxolanones, novel derivatives of DAG-lactones, exploit an additional point of contact (glutamine 27) in their binding with the C1b domain of PKC delta. Mutation of this point of contact to glutamate selectively impairs binding of the DAG-dioxolanones compared to that of the corresponding DAG-lactones (1200- to 3000-fold versus 35- to 55-fold, respectively). The differential response of this mutated C1b domain to the DAG-dioxolanones relative to the DAG-lactones provides a unique tool to probe the role of the C1b domain in PKC delta function, where the response to the DAG-lactones affords a positive control for retained function. Using this approach, we show that the C1b domain of PKC delta plays the predominant role in the translocation of PKC delta to the membrane in the presence of DAG.


Subject(s)
Diglycerides/chemistry , Dioxolanes/chemistry , Lactones/chemistry , Models, Molecular , Protein Kinase C-delta/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cricetulus , Diglycerides/pharmacology , Dioxolanes/pharmacology , Glutamine/chemistry , Green Fluorescent Proteins/genetics , Molecular Conformation , Molecular Sequence Data , Mutation , Protein Binding , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stereoisomerism
7.
J Med Chem ; 49(11): 3185-203, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16722637

ABSTRACT

Diacylglycerol lactones (DAG-lactones) are known to operate as effective agonists of protein kinase C (PKC), surpassing in potency the activity of natural diacylglycerol (DAG). Localization of activated PKC isozymes in the cell is determined in part by the different cellular scaffolds, the lipid composition of the specific membranes, and the targeting information intrinsic to the individual isoforms bound to DAG. This multifaceted control of diversity suggests that, to develop effective DAG-lactones capable of honing in on a specific cellular target, we need to gain a better understanding of the chemical space surrounding its binding site. Seeking to augment the chemical repertoire of DAG-lactone side chains that could steer the translocation of PKC to specific cellular domains, we report herein the effects of incorporating simple or substituted phenyl residues. A combined series of n-alkyl and phenyl substitutions were used to explore the optimal location of the phenyl group on the side chains. The substantial differences in binding affinity between DAG-lactones with identical functionalized phenyl groups at either the sn-1 or sn-2 position are consistent with the proposed binding model in which the DAG-lactone binds to the C1 domain of PKC with the acyl chain oriented toward the interior of the membrane and the alpha-alkylidene or alpha-arylalkylidene chains directed to the surface of the C1 domain adjacent to the lipid interface. We conclude that DAG-lactones containing alpha-phenylalkylidene side chains at the sn-2 position represent excellent scaffolds upon which to explore further chemical diversity.


Subject(s)
Diglycerides/chemistry , Lactones/chemistry , Models, Molecular , Protein Kinase C/chemistry , Diglycerides/chemical synthesis , Furans/chemical synthesis , Furans/chemistry , Lactones/chemical synthesis , Ligands , Molecular Conformation , Protein Binding , Protein Structure, Tertiary , Stereoisomerism
8.
J Med Chem ; 48(18): 5738-48, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16134942

ABSTRACT

Diacylglycerol (DAG) lactones with altered functionality (C=O --> CH(2) or C=O --> C=S) at the sn-1 and sn-2 carbonyl pharmacophores were synthesized and used as probes to dissect the individual role of each carbonyl in the binding to protein kinase C (PKC). The results suggest that the hydrated sn-1 carbonyl is engaged in very strong hydrogen-bonding interactions with the charged lipid headgroups and organized water molecules at the lipid interface. Conversely, the sn-2 carbonyl has a more modest contribution to the binding process as a result of its involvement with the receptor (C1 domain) via conventional hydrogen bonding to the protein. The parent DAG-lactones, E-6 and Z-7, were designed to bind exclusively in the sn-2 binding mode to ensure the correct orientation and disposition of pharmacophores at the binding site.


Subject(s)
Diglycerides/chemistry , Lactones/chemistry , Protein Kinase C/chemistry , Binding Sites , Diglycerides/chemical synthesis , Lactones/chemical synthesis , Molecular Conformation , Protein Binding , Quantitative Structure-Activity Relationship , Radioligand Assay , Stereoisomerism , Thermodynamics
9.
Org Lett ; 6(14): 2413-6, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15228292

ABSTRACT

[structure: see text] Commercially available 2-methylenepropane-1,3-diol was converted to chiral epoxide (R)-2 via Sharpless asymmetric epoxidation in >96% ee. Regiospecific epoxide ring opening and reduction of the intermediate alkyne set the stage for a one-pot lactonization to give (R)-6, a convenient precursor for all functionalized chiral DAG-lactones used as potent PK-C ligands. The synthesis of the most potent DAG-lactones known to date, (Z)-10 and (E)-10, served to confirm PK-C's exclusive preference for the (R)-stereochemistry in this class of compounds.


Subject(s)
Diglycerides/chemistry , Lactones/chemical synthesis , Protein Kinase C/metabolism , Binding Sites , Catalysis , Epoxy Compounds/chemical synthesis , Epoxy Compounds/chemistry , Ligands , Molecular Conformation , Molecular Structure , Stereoisomerism
10.
J Med Chem ; 57(9): 3835-44, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24684293

ABSTRACT

To explore the feasibility of developing ligands targeted to the atypical C1 domains of protein kinase C ƎĀ¶ and ƎĀ¹, we have prepared diacylglycerol lactones substituted with hydrophilic groups on their side chains, which potentially could interact with the arginine residues that distinguish the atypical C1 domains of PKCƎĀ¶ and PKCƎĀ¹ from typical C1 domains, and we have measured their binding to mutated versions of the C1b domain of PKCƎĀ“ that incorporate one or more of these arginine residues. The most selective of the diacylglycerol lactones showed only a 10-fold reduction in binding affinity with the triple arginine mutant (N7R/S10R/L20R) compared to the wild-type, whereas phorbol 12,13-dibutyrate showed a 6000-fold loss of affinity. Molecular modeling confirms that these ligands are indeed able to interact with the arginine residues. Our results show that dramatic changes in selectivity can be obtained through appropriate substitution of diacylglycerol lactones.


Subject(s)
Diglycerides/pharmacology , Lactones/pharmacology , Protein Kinase C/chemistry , Amino Acid Sequence , Diglycerides/chemistry , Lactones/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Sequence Homology, Amino Acid , Spectrometry, Mass, Fast Atom Bombardment
11.
J Biol Chem ; 284(2): 1302-12, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19001377

ABSTRACT

Classic and novel protein kinase C (PKC) isozymes contain two zinc finger motifs, designated "C1a" and "C1b" domains, which constitute the recognition modules for the second messenger diacylglycerol (DAG) or the phorbol esters. However, the individual contributions of these tandem C1 domains to PKC function and, reciprocally, the influence of protein context on their function remain uncertain. In the present study, we prepared PKCdelta constructs in which the individual C1a and C1b domains were deleted, swapped, or substituted for one another to explore these issues. As isolated fragments, both the deltaC1a and deltaC1b domains potently bound phorbol esters, but the binding of [(3)H]phorbol 12,13-dibutyrate ([(3)H]PDBu) by the deltaC1a domain depended much more on the presence of phosphatidylserine than did that of the deltaC1b domain. In intact PKCdelta, the deltaC1b domain played the dominant role in [(3)H]PDBu binding, membrane translocation, and down-regulation. A contribution from the deltaC1a domain was nonetheless evident, as shown by retention of [(3)H]PDBu binding at reduced affinity, by increased [(3)H]PDBu affinity upon expression of a second deltaC1a domain substituting for the deltaC1b domain, and by loss of persistent plasma membrane translocation for PKCdelta expressing only the deltaC1b domain, but its contribution was less than predicted from the activity of the isolated domain. Switching the position of the deltaC1b domain to the normal position of the deltaC1a domain (or vice versa) had no apparent effect on the response to phorbol esters, suggesting that the specific position of the C1 domain within PKCdelta was not the primary determinant of its activity.


Subject(s)
Protein Kinase C-delta/chemistry , Protein Kinase C-delta/metabolism , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Down-Regulation , Humans , Ligands , Mice , Molecular Sequence Data , Mutation/genetics , Phorbol Esters/metabolism , Protein Binding , Protein Kinase C-delta/genetics , Protein Structure, Tertiary , Protein Transport , Substrate Specificity , Zinc Fingers
12.
J Biol Chem ; 283(16): 10543-9, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18263588

ABSTRACT

C1 domains mediate the recognition and subsequent signaling response to diacylglycerol and phorbol esters by protein kinase C (PKC) and by several other families of signal-transducing proteins such as the chimerins or RasGRP. MRCK (myotonic dystrophy kinase-related Cdc42 binding kinase), a member of the dystrophia myotonica protein kinase family that functions downstream of Cdc42, contains a C1 domain with substantial homology to that of the diacylglycerol/phorbol ester-responsive C1 domains and has been reported to bind phorbol ester. We have characterized here the interaction of the C1 domains of the two MRCK isoforms alpha and beta with phorbol ester. The MRCK C1 domains bind [20-(3)H]phorbol 12,13-dibutyrate with K(d) values of 10 and 17 nm, respectively, reflecting 60-90-fold weaker affinity compared with the protein kinase C delta C1b domain. In contrast to binding by the C1b domain of PKCdelta, the binding by the C1 domains of MRCK alpha and beta was fully dependent on the presence of phosphatidylserine. Comparison of ligand binding selectivity showed resemblance to that by the C1b domain of PKCalpha and marked contrast to that of the C1b domain of PKCdelta. In intact cells, as in the binding assays, the MRCK C1 domains required 50-100-fold higher concentrations of phorbol ester for induction of membrane translocation. We conclude that additional structural elements within the MRCK structure are necessary if the C1 domains of MRCK are to respond to phorbol ester at concentrations comparable with those that modulate PKC.


Subject(s)
Phorbol Esters/chemistry , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Amino Acid Sequence , Biological Transport , Cell Line, Tumor , Escherichia coli/metabolism , Humans , Kinetics , Molecular Sequence Data , Myotonin-Protein Kinase , Phosphatidylserines/chemistry , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Protein Transport , Protein-Tyrosine Kinases/chemistry
13.
J Biol Chem ; 281(44): 33773-88, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16950780

ABSTRACT

The C1 domain zinc finger structure is highly conserved among the protein kinase C (PKC) superfamily members. As the interaction site for the second messenger sn-1,2-diacylglycerol (DAG) and for the phorbol esters, the C1 domain has been an important target for developing selective ligands for different PKC isoforms. However, the C1 domains of the atypical PKC members are DAG/phorbol ester-insensitive. Compared with the DAG/phorbol ester-sensitive C1 domains, the rim of the binding cleft of the atypical PKC C1 domains possesses four additional positively charged arginine residues (at positions 7, 10, 11, and 20). In this study, we showed that mutation to arginines of the four corresponding sites in the C1b domain of PKCdelta abolished its high potency for phorbol 12,13-dibutyrate in vitro, with only marginal remaining activity for phorbol 12-myristate 13-acetate in vivo. We also demonstrated both in vitro and in vivo that the loss of potency to ligands was cumulative with the introduction of the arginine residues along the rim of the binding cavity rather than the consequence of loss of a single, specific residue. Computer modeling reveals that these arginine residues reduce access of ligands to the binding cleft and change the electrostatic profile of the C1 domain surface, whereas the basic structure of the binding cleft is still maintained. Finally, mutation of the four arginine residues of the atypical PKC C1 domains to the corresponding residues in the deltaC1b domain conferred response to phorbol ester. We speculate that the arginine residues of the C1 domain of atypical PKCs may provide an opportunity for the design of ligands selective for the atypical PKCs.


Subject(s)
Arginine/chemistry , Arginine/metabolism , Cell Membrane/enzymology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Arginine/genetics , Cell Line, Tumor , Computational Biology , Cricetinae , Humans , Ions/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Kinase C/genetics , Protein Structure, Tertiary , Protein Transport , Sequence Alignment , Static Electricity
14.
J Biol Chem ; 280(29): 27329-38, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15923197

ABSTRACT

Although multiple natural products are potent ligands for the diacylglycerol binding C1 domain of protein kinase C (PKC), RasGRP, and related targets, the high conservation of C1 domains has impeded the development of selective ligands. We characterized here a diacylglycerol-lactone, 130C037, emerging from a combinatorial chemical synthetic strategy, which showed substantial selectivity. 130C037 gave shallow binding curves for PKC isoforms alpha, beta, gamma, delta, and epsilon, with apparent Ki values ranging from 340 nm for PKCalpha to 29 nm for PKCepsilon. When binding to isolated C1 domains of PKCalpha and -delta, 130C037 showed good affinity (Ki= 1.78 nm) only for deltaC1b, whereas phorbol 12,13-dibutyrate showed affinities within 10-fold for all. In LNCaP cells, 130C037 likewise selectively induced membrane translocation of deltaC1b. 130C037 bound intact RasGRP1 and RasGRP3 with Ki values of 3.5 and 3.8 nm, respectively, reflecting 8- and 90-fold selectivity relative to PKCepsilon and PKCalpha. By Western blot of Chinese hamster ovary cells, 130C037 selectively induced loss from the cytosol of RasGRP3 (ED50 = 286 nm), partial reduction of PKCepsilon (ED50 > 10 microm), and no effect on PKCalpha. As determined by confocal microscopy in LNCaP cells, 130C037 caused rapid translocation of RasGRP3, limited slow translocation of PKCepsilon, and no translocation of PKCalpha. Finally, 130C037 induced Erk phosphorylation in HEK-293 cells ectopically expressing RasGRP3 but not in control cells, whereas phorbol ester induced phosphorylation in both. The properties of 130C037 provide strong proof of principle for the feasibility of developing ligands with selectivity among C1 domain-containing therapeutic targets.


Subject(s)
Diglycerides/pharmacology , Lactones/pharmacology , Protein Kinase C/metabolism , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Isoenzymes , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation/drug effects , Protein Binding , Protein Kinase C/chemistry , Protein Kinase C-alpha , Protein Kinase C-delta , Protein Transport , ras Guanine Nucleotide Exchange Factors
15.
J Biol Chem ; 280(18): 18179-88, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15710623

ABSTRACT

The neuroprotective properties of bis(7)-tacrine, a novel dimeric acetylcholinesterase (AChE) inhibitor, on glutamate-induced excitotoxicity were investigated in primary cultured cerebellar granule neurons (CGNs). Exposure of CGNs to 75 mum glutamate resulted in neuronal apoptosis as demonstrated by Hoechst staining, TUNEL, and DNA fragmentation assays. The bis(7)-tacrine treatment (0.01-1 mum) on CGNs markedly reduced glutamate-induced apoptosis in dose- and time-dependent manners. However, donepezil and other AChE inhibitors, even at concentrations of inhibiting AChE to the similar extents as 1 mum bis(7)-tacrine, failed to prevent glutamate-induced excitotoxicity in CGNs; moreover, both atropine and dihydro-beta-erythroidine, the cholinoreceptor antagonists, did not affect the anti-apoptotic properties of bis(7)-tacrine, suggesting that the neuroprotection of bis(7)-tacrine appears to be independent of inhibiting AChE and cholinergic transmission. In addition, ERK1/2 and p38 pathways, downstream signals of N-methyl-d-aspartate (NMDA) receptors, were rapidly activated after the exposure of glutamate to CGNs. Bis(7)-tacrine inhibited the apoptosis and the activation of these two signals with the same efficacy as the coapplication of PD98059 and SB203580. Furthermore, using fluorescence Ca(2+) imaging, patch clamp, and receptor-ligand binding techniques, bis(7)-tacrine was found effectively to buffer the intracellular Ca(2+) increase triggered by glutamate, to reduce NMDA-activated currents and to compete with [(3)H]MK-801 with an IC(50) value of 0.763 mum in rat cerebellar cortex membranes. These findings strongly suggest that bis(7)-tacrine prevents glutamate-induced neuronal apoptosis through directly blocking NMDA receptors at the MK-801-binding site, which offers a new and clinically significant modality as to how the agent exerts neuroprotective effects.


Subject(s)
Apoptosis/drug effects , Cholinesterase Inhibitors/pharmacology , Glutamic Acid/pharmacology , Indans/pharmacology , Neurons/cytology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Tacrine/analogs & derivatives , Animals , Apoptosis/physiology , Cells, Cultured , Cholinesterase Inhibitors/chemistry , Dimerization , Donepezil , Dose-Response Relationship, Drug , Neurons/drug effects , Neurons/enzymology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/physiology , Tacrine/chemistry , Tacrine/pharmacology
16.
Biochem Biophys Res Commun ; 299(5): 762-9, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12470644

ABSTRACT

We showed previously that a cytosolic Ca(2+) signal is involved in regulating UV-induced apoptosis in HeLa cells. In this study, we found evidence that this Ca(2+) signal occurs upstream of the release of cytochrome c from mitochondria. First, when we abolished [Ca(2+)](i) increases by injecting BAPTA or heparin into UV-treated HeLa cells, cytochrome c release was either blocked or severely delayed. Second, using a living cell imaging technique, we observed a series of transient [Ca(2+)](i) increases (typically lasting about 40-60s) in many apoptotic cells induced by either UV- or TNFalpha-treatment. Third, using GFP-tagged cytochrome c, we found that the Ca(2+) spikes appear in a time window before cytochrome c was released. Finally, by fixing the TNFalpha-treated cell at the time when it started to display Ca(2+) spikes, we examined the distribution of its endogenous cytochrome c using immunostaining. We found that cytochrome c was not yet released from mitochondria. These findings suggest the existence of certain apoptotic pathways, in which an early Ca(2+) signal is activated upstream of cytochrome c release.


Subject(s)
Apoptosis , Calcium Signaling , Cytochrome c Group/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , HeLa Cells , Heparin/pharmacology , Humans , Kinetics , Microscopy, Confocal , Mitochondria/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays
17.
Biochem Biophys Res Commun ; 304(2): 217-22, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12711301

ABSTRACT

In this study, we reported the first measurement of the dynamics of activation of caspase-8 in a single living cell. This measurement was conducted using a specially developed molecular sensor based on the FRET (fluorescence resonance energy transfer) technique. This sensor was constructed by fusing a CFP (cyan fluorescent protein) and a YFP (yellow fluorescent protein) with a linker containing a tandem caspase-8-specific cleavage site. The change of the FRET ratio upon cleavage was larger than 4-fold. Using this sensor, we found that during TNFalpha-induced apoptosis, the activation of caspase-8 was a slower process than that of caspase-3, and it was initiated much earlier than the caspase-3 activation. Inhibition of caspase-9 delayed the full activation of caspase-3 but did not affect the dynamics of caspase-8. Results of these single-cell measurements suggested that caspase-3 was activated by caspase-8 through two parallel pathways during TNFalpha-induced apoptosis in HeLa cells.


Subject(s)
Apoptosis , Caspases/metabolism , Fluorescence Resonance Energy Transfer , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Caspase 3 , Caspase 8 , Caspase 9 , Enzyme Activation , Fluorescent Dyes/chemistry , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Microscopy, Fluorescence , Models, Biological , Molecular Sequence Data , Recombinant Fusion Proteins/analysis , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
18.
J Neurochem ; 91(5): 1219-30, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15569265

ABSTRACT

Minocycline has been shown to have remarkably neuroprotective qualities, but underlying mechanisms remain elusive. We reported here the robust neuroprotection by minocycline against glutamate-induced apoptosis through regulations of p38 and Akt pathways. Pre-treatment of cerebellar granule neurons (CGNs) with minocycline (10-100 microm) elicited a dose-dependent reduction of glutamate excitotoxicity and blocked glutamate-induced nuclear condensation and DNA fragmentations. Using patch-clamping and fluorescence Ca2+ imaging techniques, it was found that minocycline neither blocked NMDA receptors, nor reduced glutamate-caused rises in intracellular Ca2+. Instead, confirmed by immunoblots, minocycline in vivo and in vitro was shown to directly inhibit the activation of p38 caused by glutamate. A p38-specific inhibitor, SB203580, also attenuated glutamate excitotoxicity. Furthermore, the neuroprotective effects of minocycline were blocked by phosphatidylinositol 3-kinase (PI3-K) inhibitors LY294002 and wortmannin, while pharmacologic inhibition of glycogen synthase kinase 3beta (GSK3beta) attenuated glutamate-induced apoptosis. In addition, immunoblots revealed that minocycline reversed the suppression of phosphorylated Akt and GSK3beta caused by glutamate, as were abolished by PI3-K inhibitors. These results demonstrate that minocycline prevents glutamate-induced apoptosis in CGNs by directly inhibiting p38 activity and maintaining the activation of PI3-K/Akt pathway, which offers a novel modality as to how the drug exerts protective effects.


Subject(s)
Apoptosis/drug effects , Cerebellum/cytology , Glutamic Acid/pharmacology , Minocycline/pharmacology , Neurons/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 2 , Animals , Animals, Newborn , Blotting, Western/methods , Calcium/metabolism , Cell Count/methods , Cell Survival/drug effects , Cells, Cultured , Chromatin/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Fragmentation/drug effects , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Humans , Membrane Potentials/drug effects , Microscopy, Confocal/methods , N-Methylaspartate/pharmacology , Neurons/pathology , Neurons/physiology , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques/methods , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Serine/metabolism , Signal Transduction/drug effects , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Time Factors , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL