Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Zoo Wildl Med ; 55(2): 301-312, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38875187

RESUMEN

The wild rhinoceros populations have declined drastically in the past decades because the rhinoceros are heavily hunted for their horns. Zoological institutions aim to conserve rhinoceros populations in captivity, but one of the challenges of ex situ conservation is to provide food sources that resemble those available in the wild. Considering that the mammalian gut microbiota is a pivotal player in their host's health, the gut microbiota of rhinoceros may also play a role in the bioavailability of nutrients. Therefore, this study aims to characterize the fecal microbiome composition of grazing white rhinoceros (WR; Ceratotherium simum) and greater one-horned rhinoceros (GOHR; Rhinoceros unicornis) as well as the browsing black rhinoceros (BR; Diceros bicornis) kept in European zoos. Over the course of 1 yr, 166 fecal samples in total were collected from 9 BR (n = 39), 10 GOHR (n = 56), and 14 WR (n = 71) from 23 zoological institutions. The bacterial composition in the samples was determined using 16S rRNA gene Illumina sequencing. The fecal microbiomes of rhinoceros clustered by species, with BR clustering more distantly from GOHR and WR. Furthermore, the data report clustering of rhinoceros microbiota according to individual rhinoceros and institutional origin, showing that zoological institutions play a significant role in shaping the gut microbiome of rhinoceros species. In addition, BR exhibit a relatively higher microbial diversity than GOHR and WR. BR seem more susceptible to microbial gut changes and appear to have a more diverse microbiome composition among individuals than GOHR and WR. These data expand on the role of gut microbes and can provide baseline data for continued efforts in rhinoceros conservation and health status.


Asunto(s)
Animales de Zoológico , Microbioma Gastrointestinal , Perisodáctilos , Animales , Perisodáctilos/microbiología , Animales de Zoológico/microbiología , Europa (Continente) , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Especificidad de la Especie , Heces/microbiología , ARN Ribosómico 16S/genética , ARN Bacteriano/genética
2.
Br J Clin Pharmacol ; 89(3): 1105-1114, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36214216

RESUMEN

AIM: To assess viral clearance, pharmacokinetics, tolerability and symptom evolution following ensovibep administration in symptomatic COVID-19 outpatients. METHODS: In this open-label, first-in-patient study a single dose of either 225 mg (n = 6) or 600 mg (n = 6) of ensovibep was administered intravenously in outpatients with mild-to-moderate COVID-19 symptoms. Pharmacokinetic profiles were determined (90-day period). Pharmacodynamic assessments consisted of viral load (qPCR and cultures) and symptom questionnaires. Immunogenicity against ensovibep and SARS-CoV-2-neutralizing activity were determined. Safety and tolerability were assessed throughout a 13-week follow-up. RESULTS: Both doses showed similar pharmacokinetics (first-order) with mean half-lives of 14 (SD 5.0) and 13 days (SD 5.7) for the 225- and 600-mg groups, respectively. Pharmacologically relevant serum concentrations were maintained in all subjects for at least 2 weeks postdose, regardless of possible immunogenicity against ensovibep. Viral load changes from baseline at day 15 were 5.1 (SD 0.86) and 5.3 (SD 2.2) log10 copies/mL for the 225- and 600-mg doses, respectively. COVID-19 symptom scores decreased from 10.0 (SD 4.1) and 11.3 (SD 4.0) to 1.6 (SD 3.1) and 3.3 (SD 2.4) in the first week for the 225- and 600-mg groups, respectively. No anti-SARS-CoV-2 neutralizing activity was present predose and all patients had SARS-CoV-2 antibodies at day 91. Adverse events were of mild-to-moderate severity, transient and self-limiting. CONCLUSION: Single-dose intravenous administration of 225 or 600 mg of ensovibep appeared safe and well tolerated in patients with mild-to-moderate COVID-19. Ensovibep showed favourable pharmacokinetics in patients and the pharmacodynamic results warrant further research in a larger phase 2/3 randomized-controlled trail.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Proteínas Recombinantes de Fusión , Anticuerpos Antivirales , Método Doble Ciego
3.
Mol Biol Evol ; 39(2)2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35143659

RESUMEN

Supergenes are nonrecombining genomic regions ensuring the coinheritance of multiple, coadapted genes. Despite the importance of supergenes in adaptation, little is known on how they originate. A classic example of supergene is the S locus controlling heterostyly, a floral heteromorphism occurring in 28 angiosperm families. In Primula, heterostyly is characterized by the cooccurrence of two complementary, self-incompatible floral morphs and is controlled by five genes clustered in the hemizygous, ca. 300-kb S locus. Here, we present the first chromosome-scale genome assembly of any heterostylous species, that of Primula veris (cowslip). By leveraging the high contiguity of the P. veris assembly and comparative genomic analyses, we demonstrated that the S-locus evolved via multiple, asynchronous gene duplications and independent gene translocations. Furthermore, we discovered a new whole-genome duplication in Ericales that is specific to the Primula lineage. We also propose a mechanism for the origin of S-locus hemizygosity via nonhomologous recombination involving the newly discovered two pairs of CFB genes flanking the S locus. Finally, we detected only weak signatures of degeneration in the S locus, as predicted for hemizygous supergenes. The present study provides a useful resource for future research addressing key questions on the evolution of supergenes in general and the S locus in particular: How do supergenes arise? What is the role of genome architecture in the evolution of complex adaptations? Is the molecular architecture of heterostyly supergenes across angiosperms similar to that of Primula?


Asunto(s)
Flores , Primula , Cromosomas , Flores/genética , Duplicación de Gen , Genómica , Humanos , Primula/genética
4.
Plant J ; 101(6): 1378-1396, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31692190

RESUMEN

Marchantia polymorpha has recently become a prime model for cellular, evo-devo, synthetic biological, and evolutionary investigations. We present a pseudomolecule-scale assembly of the M. polymorpha genome, making comparative genome structure analysis and classical genetic mapping approaches feasible. We anchored 88% of the M. polymorpha draft genome to a high-density linkage map resulting in eight pseudomolecules. We found that the overall genome structure of M. polymorpha is in some respects different from that of the model moss Physcomitrella patens. Specifically, genome collinearity between the two bryophyte genomes and vascular plants is limited, suggesting extensive rearrangements since divergence. Furthermore, recombination rates are greatest in the middle of the chromosome arms in M. polymorpha like in most vascular plant genomes, which is in contrast with P. patens where recombination rates are evenly distributed along the chromosomes. Nevertheless, some other properties of the genome are shared with P. patens. As in P. patens, DNA methylation in M. polymorpha is spread evenly along the chromosomes, which is in stark contrast with the angiosperm model Arabidopsis thaliana, where DNA methylation is strongly enriched at the centromeres. Nevertheless, DNA methylation and recombination rate are anticorrelated in all three species. Finally, M. polymorpha and P. patens centromeres are of similar structure and marked by high abundance of retroelements unlike in vascular plants. Taken together, the highly contiguous genome assembly we present opens unexplored avenues for M. polymorpha research by linking the physical and genetic maps, making novel genomic and genetic analyses, including map-based cloning, feasible.


Asunto(s)
Genoma de Planta/genética , Marchantia/genética , Centrómero/genética , Mapeo Cromosómico/métodos , Cromosomas de las Plantas/genética , Genes de Plantas/genética , Ligamiento Genético , Modelos Genéticos , Recombinación Genética/genética , Secuencias Repetidas en Tándem/genética
5.
J Virol ; 83(22): 11940-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19740986

RESUMEN

The replication of enteroviruses is sensitive to brefeldin A (BFA), an inhibitor of endoplasmic reticulum-to-Golgi network transport that blocks activation of guanine exchange factors (GEFs) of the Arf GTPases. Mammalian cells contain three BFA-sensitive Arf GEFs: GBF1, BIG1, and BIG2. Here, we show that coxsackievirus B3 (CVB3) RNA replication is insensitive to BFA in MDCK cells, which contain a BFA-resistant GBF1 due to mutation M832L. Further evidence for a critical role of GBF1 stems from the observations that viral RNA replication is inhibited upon knockdown of GBF1 by RNA interference and that replication in the presence of BFA is rescued upon overexpression of active, but not inactive, GBF1. Overexpression of Arf proteins or Rab1B, a GTPase that induces GBF1 recruitment to membranes, failed to rescue RNA replication in the presence of BFA. Additionally, the importance of the interaction between enterovirus protein 3A and GBF1 for viral RNA replication was investigated. For this, the rescue from BFA inhibition of wild-type (wt) replicons and that of mutant replicons of both CVB3 and poliovirus (PV) carrying a 3A protein that is impaired in binding GBF1 were compared. The BFA-resistant GBF1-M832L protein efficiently rescued RNA replication of both wt and mutant CVB3 and PV replicons in the presence of BFA. However, another BFA-resistant GBF1 protein, GBF1-A795E, also efficiently rescued RNA replication of the wt replicons, but not that of mutant replicons, in the presence of BFA. In conclusion, this study identifies a critical role for GBF1 in CVB3 RNA replication, but the importance of the 3A-GBF1 interaction requires further study.


Asunto(s)
Enterovirus Humano B/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , ARN Viral/biosíntesis , Replicación Viral/fisiología , Factor 1 de Ribosilacion-ADP/fisiología , Animales , Brefeldino A/farmacología , Línea Celular , Enterovirus Humano B/efectos de los fármacos , GTP Fosfohidrolasas/fisiología , Células HeLa , Humanos , Replicación Viral/efectos de los fármacos
6.
Traffic ; 10(3): 316-23, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19170981

RESUMEN

ADP-ribosylation factor (Arf) proteins are small guanosine triphosphatases (GTPases) that act as major regulators of intracellular vesicular trafficking and secretory organelle pathway integrity. Like all small monomeric GTPases, Arf proteins cycle between a GDP-bound and a GTP-bound state, and this cycling is catalysed by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins. While the class I Arfs, especially Arf1, have been studied extensively, little is known as yet about the function and regulation of class II Arfs, Arf4 and Arf5. In this study, we show that Arf proteins show class-specific dynamic behaviour. Moreover, unlike class I Arfs, membrane association of class II Arfs is resistant to inhibition of large Arf GEFs by Brefeldin A. Through the construction of Arf chimeric proteins, evidence is provided that the N-terminal amphipathic helix and a class-specific residue in the conserved interswitch domain determine the membrane-binding properties of class I and class II Arf proteins. Our results show that fundamental differences exist in behaviour and regulation of these small GTPases.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Membrana Celular/metabolismo , Factores de Ribosilacion-ADP/química , Factores de Ribosilacion-ADP/genética , Secuencia de Aminoácidos , Animales , Brefeldino A/farmacología , Línea Celular , Membrana Celular/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Haplorrinos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Especificidad por Sustrato
7.
J Gen Virol ; 88(Pt 11): 3027-3030, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17947526

RESUMEN

During enterovirus infection, host cell membranes are rigorously rearranged and modified. One ubiquitously expressed lipid-modifying enzyme that might contribute to these alterations is phospholipase D (PLD). Here, we investigated PLD activity in coxsackievirus-infected cells. We show that PLD activity is not required for efficient coxsackievirus RNA replication. Instead, PLD activity rapidly decreased upon infection and upon ectopic expression of the viral 3A protein, which inhibits the PLD activator ADP-ribosylation factor 1. However, similar decreases were observed during infection with coxsackieviruses carrying defective mutant 3A proteins. Possible causes for the reduction of PLD activity and the biological consequences are discussed.


Asunto(s)
Enterovirus/enzimología , Fosfolipasa D/metabolismo , Proteínas Virales/metabolismo , Factor 1 de Ribosilacion-ADP/antagonistas & inhibidores , Animales , Línea Celular , Chlorocebus aethiops , Fosfolipasa D/antagonistas & inhibidores , ARN Viral/biosíntesis , Proteínas Virales/antagonistas & inhibidores , Replicación Viral
8.
J Virol ; 81(10): 5238-45, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17329336

RESUMEN

The 3A protein of coxsackievirus B3 (CVB3), a small membrane protein that forms homodimers, inhibits endoplasmic reticulum-to-Golgi complex transport. Recently, we described the underlying mechanism by showing that the CVB3 3A protein binds to and inhibits the function of GBF1, a guanine nucleotide exchange factor for ADP-ribosylation factor 1 (Arf1), thereby interfering with Arf1-mediated COP-I recruitment. This study was undertaken to gain more insight into the molecular determinants underlying the interaction between 3A and GBF1. Here we show that 3A mutants that have lost the ability to dimerize are no longer able to bind to GBF1 and trap it on membranes. Moreover, we identify a conserved region in the N terminus of 3A that is crucial for GBF1 binding but not for 3A dimerization. Analysis of the binding domain in GBF1 showed that the extreme N terminus, the dimerization/cyclophilin binding domain, and the homology upstream of Sec7 domain are required for the interaction with 3A. In contrast to that of full-length GBF1, overexpression of a GBF1 mutant lacking its extreme N terminus failed to rescue the effects of 3A. Together, these data provide insight into the molecular requirements of the interaction between 3A and GBF1.


Asunto(s)
Enterovirus Humano B/metabolismo , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia Conservada/genética , Dimerización , Enterovirus Humano B/genética , Factores de Intercambio de Guanina Nucleótido/genética , Microscopía Confocal , Microscopía Fluorescente , Modelos Moleculares , Mutación , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Virales/genética
9.
J Virol ; 80(23): 11852-60, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17005635

RESUMEN

The 3A protein of the coxsackievirus B3 (CVB3), an enterovirus that belongs to the family of the picornaviruses, inhibits endoplasmic reticulum-to-Golgi transport. Recently, we elucidated the underlying mechanism by showing that CVB3 3A interferes with ADP-ribosylation factor 1 (Arf1)-dependent COP-I recruitment to membranes by binding and inhibiting the function of GBF1, a guanine nucleotide exchange factor that is required for the activation of Arf1 (E. Wessels et al., Dev. Cell 11:191-201, 2006). Here, we show that the 3A protein of poliovirus, another enterovirus, is also able to interfere with COP-I recruitment through the same mechanism. No interference with protein transport or COP-I recruitment was observed for the 3A proteins of any of the other picornaviruses tested here (human rhinovirus [HRV], encephalomyocarditis virus, foot-and-mouth disease virus, and hepatitis A virus). We show that the 3A proteins of HRV, which are the most closely related to the enteroviruses, are unable to inhibit COP-I recruitment, due to a reduced ability to bind GBF1. When the N-terminal residues of the HRV 3A proteins are replaced by those of CVB3 3A, chimeric proteins are produced that have gained the ability to bind GBF1 and, by consequence, to inhibit protein transport. These results show that the N terminus of the CVB3 3A protein is important for binding of GBF1 and its transport-inhibiting function. Taken together, our data demonstrate that the activity of the enterovirus 3A protein to inhibit GBF1-dependent COP-I recruitment is unique among the picornaviruses.


Asunto(s)
Proteína Coat de Complejo I/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Picornaviridae/química , Transporte de Proteínas/efectos de los fármacos , Proteínas Virales/farmacología , Animales , Línea Celular , Proteína Coat de Complejo I/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/fisiología , Picornaviridae/genética , Transporte de Proteínas/fisiología , Proteínas Virales/metabolismo
10.
Dev Cell ; 11(2): 191-201, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890159

RESUMEN

Many viruses modify cellular processes for their own benefit. The enterovirus 3A protein inhibits endoplasmic reticulum (ER)-to-Golgi transport, a function previously suggested to be important for viral suppression of immune responses. Here, we show that a virus carrying a 3A protein defective in inhibiting ER-to-Golgi transport is indeed less virulent in mice, and we unravel the mechanism by which 3A inhibits this trafficking step. Evidence is provided that 3A inhibits the activation of the GTPase ADP-ribosylation factor 1 (Arf1), which regulates the recruitment of the COP-I coat complex to membranes. 3A specifically inhibits the function of GBF1, a guanine nucleotide exchange factor for Arf1, by interacting with its N terminus. By specifically interfering with GBF1-mediated Arf1 activation, 3A may prove a valuable tool in dissecting the early steps of the secretory pathway.


Asunto(s)
Factor 1 de Ribosilacion-ADP/antagonistas & inhibidores , Proteína Coat de Complejo I/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Proteínas Virales/farmacología , Factor 1 de Ribosilacion-ADP/metabolismo , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Chlorocebus aethiops , Proteína Coat de Complejo I/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/fisiología , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/fisiología , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/fisiología , Ratones , Modelos Animales , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología
11.
J Biol Chem ; 281(38): 28232-43, 2006 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-16867984

RESUMEN

The coxsackievirus B3 3A protein forms homodimers and plays important roles in both viral RNA (vRNA) replication and the viral inhibition of intracellular protein transport. The molecular determinants that are required for each of these functions are yet poorly understood. Based on the NMR structure of the closely related poliovirus 3A protein, a molecular model of the coxsackievirus B3 3A protein was constructed. Using this structural model, specific mutants were designed to study the structure-function relationship of 3A. The mutants were tested for their capacity to dimerize, support vRNA replication, and block protein transport. A hydrophobic interaction between the monomers and an intermolecular salt bridge were identified as major determinants required for dimerization. We show that dimerization is important for both efficient vRNA replication and inhibition of protein transport. In addition, determinants were identified that were not required for dimerization but that were essential for either one of the biological functions of 3A. The combination of the in silico and in vivo results obtained in this study provides important insights in both the structural and functional aspects of 3A.


Asunto(s)
ARN Viral/biosíntesis , Proteínas Virales/química , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Dimerización , Modelos Moleculares , Datos de Secuencia Molecular , Transporte de Proteínas , Relación Estructura-Actividad
12.
J Virol ; 79(8): 5163-73, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15795300

RESUMEN

The ability of the 3A protein of coxsackievirus B (CVB) to inhibit protein secretion was investigated for this study. Here we show that the ectopic expression of CVB 3A blocked the transport of both the glycoprotein of vesicular stomatitis virus, a membrane-bound secretory marker, and the alpha-1 protease inhibitor, a luminal secretory protein, at a step between the endoplasmic reticulum (ER) and the Golgi complex. CVB 3A contains a conserved proline-rich region in its N terminus. The importance of this proline-rich region was investigated by introducing Pro-to-Ala substitutions. The mutation of Pro19 completely abolished the ability of 3A to inhibit ER-to-Golgi transport. The mutation of Pro14, Pro17, or Pro20 also impaired this ability, but to a lesser extent. The mutation of Pro18 had no effect. We also investigated the possible importance of this proline-rich region for the function of 3A in viral RNA replication. To this end, we introduced the Pro-to-Ala mutations into an infectious cDNA clone of CVB3. The transfection of cells with in vitro-transcribed RNAs of these clones gave rise to mutant viruses that replicated with wild-type characteristics. We concluded that the proline-rich region in CVB 3A is required for its ability to inhibit ER-to-Golgi transport, but not for its function in viral RNA replication. The functional relevance of the proline-rich region is discussed in light of the proposed structural model of 3A.


Asunto(s)
Retículo Endoplásmico/fisiología , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Aparato de Golgi/fisiología , Prolina , Proteínas Virales/metabolismo , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Cartilla de ADN , Dimerización , Enterovirus Humano B/crecimiento & desarrollo , Riñón , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Plásmidos , Conformación Proteica , Transporte de Proteínas , ARN Viral/genética , Transcripción Genética , Proteínas Virales/química , Proteínas Virales/genética
13.
Eur J Pharmacol ; 502(3): 185-93, 2004 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-15476744

RESUMEN

Hexadecylphosphocholine (HePC, Miltefosine) is an antitumour phospholipid and known inducer of apoptosis in human breast cancer cells. The mechanism underlying the induction of cell death by HePC, however, is not clear yet. In this study, we have investigated the cytotoxic effects of HePC on canine mammary tumour cells (CMTs) in vitro. Upon addition of HePC, CMTs rapidly exhibited several features that resembled apoptotic cell death. Cells showed externalization of phosphatidylserine, a hallmark of apoptosis, within 5 min after addition of HePC at concentrations as low as 10 microM. Furthermore, rapid swelling of mitochondria was observed. Rounding and detachment of cells followed within 30 min. However, fragmentation of nuclear DNA could not be observed. Overall, HePC was shown to induce a type of cell death in CMTs that in some aspects resembles apoptosis, though the process proceeds much more rapidly than reported for other tumour cell lines.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Crecimiento/farmacología , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Animales , Antineoplásicos/uso terapéutico , Células CHO , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Cricetinae , Perros , Relación Dosis-Respuesta a Droga , Inhibidores de Crecimiento/uso terapéutico , Fosforilcolina/uso terapéutico , Factores de Tiempo
14.
Biochim Biophys Acta ; 1636(2-3): 99-107, 2004 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15164757

RESUMEN

The anticancer drug hexadecylphosphocholine (HePC), an alkyl-lysophospholipid analog (ALP), has been shown to induce apoptosis and inhibit the synthesis of phosphatidylcholine (PC) in a number of cell lines. We investigated whether inhibition of PC synthesis plays a major causative role in the induction of apoptosis by HePC. We therefore directly compared the apoptosis caused by HePC in CHO cells to the apoptotic process in CHO-MT58 cells, which contain a genetic defect in PC synthesis. HePC-provoked apoptosis was found to differ substantially from the apoptosis observed in MT58 cells, since it was (i) not accompanied by a large decrease in the amount of PC and diacylglycerol (DAG), (ii) not preceded by induction of the pro-apoptotic protein GADD153/CHOP, and (iii) not dependent on the synthesis of new proteins. Furthermore, lysoPC as well as lysophosphatidylethanolamine (lysoPE) could antagonize the apoptosis induced by HePC, whereas only lysoPC was able to rescue MT58 cells. HePC also induced a rapid externalisation of phosphatidylserine (PS). These observations suggest that inhibition of PC synthesis is not the primary pathway in HePC-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Fosfatidilcolinas/biosíntesis , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Animales , Células CHO , Cricetinae , Fosfatidilcolinas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA