Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
PLoS Comput Biol ; 17(9): e1009352, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491986

RESUMEN

Human Papillomaviruses (HPV) are one of the most prevalent sexually transmitted infections (STI) and the most oncogenic viruses known to humans. The vast majority of HPV infections clear in less than 3 years, but the underlying mechanisms, especially the involvement of the immune response, are still poorly known. Building on earlier work stressing the importance of randomness in the type of cell divisions in the clearance of HPV infection, we develop a stochastic mathematical model of HPV dynamics that combines the previous aspect with an explicit description of the intracellular level. We show that the random partitioning of virus episomes upon stem cell division and the occurrence of symmetric divisions dramatically affect viral persistence. These results call for more detailed within-host studies to better understand the relative importance of stochasticity and immunity in HPV infection clearance.


Asunto(s)
Infecciones por Papillomavirus/virología , División Celular/fisiología , Biología Computacional , Simulación por Computador , Interacciones Microbiota-Huesped , Humanos , Modelos Biológicos , Modelos Inmunológicos , Papillomaviridae/inmunología , Papillomaviridae/patogenicidad , Papillomaviridae/fisiología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Plásmidos/fisiología , Procesos Estocásticos , Carga Viral
2.
Nat Rev Microbiol ; 19(6): 347-359, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33469168

RESUMEN

Plasmids have a key role in bacterial ecology and evolution because they mobilize accessory genes by horizontal gene transfer. However, recent studies have revealed that the evolutionary impact of plasmids goes above and beyond their being mere gene delivery platforms. Plasmids are usually kept at multiple copies per cell, producing islands of polyploidy in the bacterial genome. As a consequence, the evolution of plasmid-encoded genes is governed by a set of rules different from those affecting chromosomal genes, and these rules are shaped by unusual concepts in bacterial genetics, such as genetic dominance, heteroplasmy or segregational drift. In this Review, we discuss recent advances that underscore the importance of plasmids in bacterial ecology and evolution beyond horizontal gene transfer. We focus on new evidence that suggests that plasmids might accelerate bacterial evolution, mainly by promoting the evolution of plasmid-encoded genes, but also by enhancing the adaptation of their host chromosome. Finally, we integrate the most relevant theoretical and empirical studies providing a global understanding of the forces that govern plasmid-mediated evolution in bacteria.


Asunto(s)
Bacterias/genética , Evolución Biológica , Transferencia de Gen Horizontal , Genoma Bacteriano , Plásmidos/fisiología , Variación Genética
3.
J Cell Biochem ; 122(3-4): 315-325, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33368623

RESUMEN

It has been demonstrated in previous studies that lncPART1 is dysregulated in non-small cell lung cancer (NSCLC). However, the function of lncPART1 in NSCLC is unclear. Therefore, this experimental design was based on LncPART1 to explore the pathogenesis of NSCLC. Real-time polymerase chain reaction was used to detect the expression of lncPART1 and miR-17-5p in NSCLC. Cell Counting Kit -8, colony formation, and transwell assays were used to examine the effects of lncPART1 and miR-17-5p on NSCLC cell proliferation and migration invasiveness. Target gene prediction, luciferase reporter assays were used to validate downstream target genes for lncPART1 and miR-17-5p. Western blot analysis was used to detect the expression of TGFBETAR2. LncPART1 was highly expressed in NSCLC. LncPART1 significantly promoted cell proliferation of NSCLC cells. miR-17-5p was down-expressed in NSCLC. miR-17-5p overexpression inhibited cell proliferation and migration invasion in NSCLC cells. LncPART1 was able to inhibit miR-17-5p expression and upregulate the expression level of TGFBETAR2. The results of in vivo animal models confirmed that lncPART1 promoted NSCLC progression by miR-17-5p/TGFBETAR2 axis. LncPART1 promoted the progression of NSCLC by miR-17-5p/TGFBETAR2 axis.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Antígeno Ki-67/metabolismo , MicroARNs/metabolismo , Plásmidos/fisiología , ARN no Traducido/metabolismo , Adulto , Anciano , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/genética , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Plásmidos/genética , ARN no Traducido/genética
4.
Trends Microbiol ; 28(2): 150-162, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31624007

RESUMEN

The human DNA tumor viruses Epstein-Barr virus (EBV), Kaposi's sarcoma-associated herpesvirus (KSHV), and human papillomavirus (HPV) share the common property of persisting as multicopy episomes in the nuclei of rapidly dividing host cells. These episomes form the molecular basis for viral latency and are etiologically linked to virus-associated cancers. Episome maintenance requires epigenetic programming to ensure the proper control of viral gene expression, DNA replication, and genome copy number. For these viruses, episome maintenance requires a dedicated virus-encoded episome maintenance protein (EMP), namely LANA (KSHV), EBNA1 (EBV), and E2 (HPV). Here, we review common features of these viral EMPs and discuss recent advances in understanding how they contribute to the epigenetic control of viral episome maintenance during latency.


Asunto(s)
Epigénesis Genética , Plásmidos/fisiología , Proteínas Virales/fisiología , Latencia del Virus , Replicación Viral , Alphapapillomavirus/fisiología , Genoma Viral , Herpesvirus Humano 4/fisiología , Herpesvirus Humano 8/fisiología , Interacciones Microbiota-Huesped , Humanos , Dominios Proteicos
5.
Nucleic Acids Res ; 45(6): 3158-3171, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28034957

RESUMEN

Genome segregation is a fundamental step in the life cycle of every cell. Most bacteria rely on dedicated DNA partition proteins to actively segregate chromosomes and low copy-number plasmids. Here, by employing super resolution microscopy, we establish that the ParF DNA partition protein of the ParA family assembles into a three-dimensional meshwork that uses the nucleoid as a scaffold and periodically shuttles between its poles. Whereas ParF specifies the territory for plasmid trafficking, the ParG partner protein dictates the tempo of ParF assembly cycles and plasmid segregation events by stimulating ParF adenosine triphosphate hydrolysis. Mutants in which this ParG temporal regulation is ablated show partition deficient phenotypes as a result of either altered ParF structure or dynamics and indicate that ParF nucleoid localization and dynamic relocation, although necessary, are not sufficient per se to ensure plasmid segregation. We propose a Venus flytrap model that merges the concepts of ParA polymerization and gradient formation and speculate that a transient, dynamic network of intersecting polymers that branches into the nucleoid interior is a widespread mechanism to distribute sizeable cargos within prokaryotic cells.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Plásmidos/fisiología , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/química , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , ADN/metabolismo , Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de Escherichia coli/análisis , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Microscopía Fluorescente , Mutación , Plásmidos/genética , Proteínas Represoras/análisis , Proteínas Represoras/genética , Imagen de Lapso de Tiempo
6.
J Biol Chem ; 290(47): 28084-28096, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26420481

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies. KSHV latency-associated nuclear antigen (LANA) mediates persistence of viral episomes in latently infected cells. LANA mediates KSHV DNA replication and segregates episomes to progeny nuclei. The structure of the LANA DNA binding domain was recently solved, revealing a positive electrostatic patch opposite the DNA binding surface, which is the site of BET protein binding. Here we investigate the functional role of the positive patch in LANA-mediated episome persistence. As expected, LANA mutants with alanine or glutamate substitutions in the central, peripheral, or lateral portions of the positive patch maintained the ability to bind DNA by EMSA. However, all of the substitution mutants were deficient for LANA DNA replication and episome maintenance. Mutation of the peripheral region generated the largest deficiencies. Despite these deficiencies, all positive patch mutants concentrated to dots along mitotic chromosomes in cells containing episomes, similar to LANA. The central and peripheral mutants, but not the lateral mutants, were reduced for BET protein interaction as assessed by co-immunoprecipitation. However, defects in BET protein binding were independent of episome maintenance function. Overall, the reductions in episome maintenance closely correlated with DNA replication deficiencies, suggesting that the replication defects account for the reduced episome persistence. Therefore, the electrostatic patch exerts a key role in LANA-mediated DNA replication and episome persistence and may act through a host cell partner(s) other than a BET protein or by inducing specific structures or complexes.


Asunto(s)
Antígenos Virales/metabolismo , Replicación del ADN , Herpesvirus Humano 8/genética , Proteínas Nucleares/metabolismo , Plásmidos/fisiología , Latencia del Virus , Sitios de Unión , Línea Celular Tumoral , Herpesvirus Humano 8/inmunología , Humanos , Electricidad Estática , Secuencias Repetidas Terminales
7.
Stem Cells ; 33(11): 3228-38, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26138940

RESUMEN

To date no authentic embryonic stem cell (ESC) line or germline-competent-induced pluripotent stem cell (iPSC) line has been established for large animals. Despite this fact, there is an impression in the field that large animal ESCs or iPSCs are as good as mouse counterparts. Clarification of this issue is important for a healthy advancement of the stem cell field. Elucidation of the causes of this failure in obtaining high quality iPSCs/ESCs may offer essential clues for eventual establishment of authentic ESCs for large animals including humans. To this end, we first generated porcine iPSCs using nonintegrating replicating episomal plasmids. Although these porcine iPSCs met most pluripotency criteria, they could neither generate cloned piglets through nuclear transfer, nor contribute to later stage chimeras through morula injections or aggregations. We found that the reprogramming genes in iPSCs could not be removed even under negative selection, indicating they are required to maintain self-renewal. The persistent expression of these genes in porcine iPSCs in turn caused differentiation defects in vivo. Therefore, incomplete reprogramming manifested by a reliance on sustained expression of exogenous-reprogramming factors appears to be the main reason for the inability of porcine iPSCs to form iPSC-derived piglets.


Asunto(s)
Vectores Genéticos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Plásmidos/fisiología , Transgenes/fisiología , Quimera por Trasplante/fisiología , Animales , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Femenino , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Ratones Desnudos , Técnicas de Transferencia Nuclear , Porcinos , Porcinos Enanos
8.
J Cardiovasc Pharmacol ; 65(2): 153-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25295466

RESUMEN

Adeno-associated virus (AAV) is a nonpathogenic virus capable of targeting human chromosome 19 for integration at AAVS1 site, and a 16 bp Rep binding element (RBE) sequence of AAV was sufficient for mediating this specific integration in the presence of AAV regulation proteins (Rep). Previously, we cotransduced 2 plasmids, pRBE-CMV-hFIX and pRC, into the AAVS1 transgenic mice by hydrodynamic injection, and a long-term expression of human coagulation Factor IX (hFIX) was observed. The corresponding AAVS1 locus site-specific integrations were verified by nested polymerase chain reaction. In this study, we established a novel hFIX expression plasmid, pRBE-HCR-hAAT-hFIX, driven by a liver-specific promoter by replacing the CMV promoter of pRBE-CMV-hFIX with a humanized promoter consisting of HCR-hAAT. The expression of hFIX in vitro was almost the same in transient transfection of pRBE-CMV-hFIX or pRBE-HCR-hAAT-hFIX. AAVS1-specific integrations were identified both in mice transfected with pRC/pRBE-CMV-hFIX cocktail and pRC/pRBE-HCR-hAAT-hFIX cocktail. However, the expression of hFIX of pRBE-HCR-hAAT-hFIX mice was higher and persisted longer. It achieved more than 1% of normal plasma hFIX concentration and maintained for 240 days. The result suggested that RBE-HCR-hAAT element could improve the expression of hFIX and present potential usage of Rep-RBE site-specific integration in gene therapy for hemophilia B.


Asunto(s)
Cromosomas Humanos Par 19 , Proteínas de Unión al ADN/genética , Terapia Genética/métodos , Hemofilia B/terapia , Proteínas Virales/genética , Animales , Células Cultivadas , Dependovirus/fisiología , Factor IX/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Hígado/metabolismo , Ratones , Ratones Transgénicos , Plásmidos/fisiología , Regiones Promotoras Genéticas
9.
Nat Commun ; 5: 5322, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25358815

RESUMEN

Conjugative plasmids are typically locked in intergenomic and sexual conflicts with co-resident rivals, whose translocation they block using fertility inhibition factors (FINs). We describe here the first crystal structure of an enigmatic FIN Osa deployed by the proteobacterial plasmid pSa. Osa contains a catalytically active version of the ParB/Sulfiredoxin fold with both ATPase and DNase activity, the latter being regulated by an ATP-dependent switch. Using the Agrobacterium tumefaciens VirB/D4 type IV secretion system (T4SS), a relative of the conjugative T4SS, we demonstrate that catalytically active Osa blocks T-DNA transfer into plants. With a partially reconstituted T4SS in vitro, we show that Osa degrades T-DNA in the T-DNA-VirD2 complex before its translocation. Further, we present evidence for conservation and interplay between ATPase and DNase activities throughout the ParB/Sulfiredoxin fold, using other members of the family, namely P1 ParB and RK2 KorB, which have general functional implications across diverse biological contexts.


Asunto(s)
Proteínas Bacterianas/metabolismo , Conjugación Genética , Plásmidos/fisiología , Adenosina Trifosfatasas/metabolismo , ADN Bacteriano/metabolismo , Desoxirribonucleasas/metabolismo , Fertilidad , Multimerización de Proteína , Sistemas de Secreción Tipo IV
10.
J Neurosci ; 33(49): 19341-51, 2013 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-24305829

RESUMEN

(Pro)renin receptor [(P)RR], also known as Atp6ap2, has attracted growing attention as a key molecule for tissue renin-angiotensin system (RAS). In addition to its role in tissue RAS activation, Atp6ap2/(P)RR was originally identified as an accessory subunit for vacuolar H(+)-ATPase (v-ATPase), which is a multisubunit proton pump involved in diverse and fundamental cellular physiology. In this study, to elucidate the physiological function of Atp6ap2/(P)RR during retinal development in mammals, we used Cre-LoxP system to generate photoreceptor-specific conditional knock-out (CKO) mice, and revealed a critical role of Atp6ap2/(P)RR in photoreceptor development. Deletion of photoreceptor Atp6ap2/(P)RR did not affect retinal cell differentiation, but led to laminar disorganization around the outer nuclear layer together with severe dysfunction of photoreceptor cells. In the CKO mice, cell adhesion and polarity molecules, some of which were colocalized with Atp6ap2/(P)RR at the apical edge of the wild-type developing retina, were substantially dispersed together with mislocalization of retinal progenitor cells apart from the apical surface. Among theses molecules, coimmunoprecipitation using retinal homogenates and ATP6AP2/(P)RR-transfected cells showed that Atp6ap2/(P)RR interacted with partitioning defective 3 homolog (PAR3) protein, which is known to function in the Par-atypical protein kinase C (aPKC) system. Furthermore, yeast two-hybrid assays demonstrated direct molecular interaction between ATP6AP2/(P)RR and PAR3. Our present data revealed the novel function of Atp6ap2/(P)RR required for laminar formation during retinal development. We propose that this cellular activity associated with the Par-aPKC system, in addition to the v-ATPase function and tissue RAS activation, is the third biological role of Atp6ap2/(P)RR.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Polaridad Celular/fisiología , ATPasas de Translocación de Protón/metabolismo , Receptores de Superficie Celular/metabolismo , Retina/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Recuento de Células , Proteínas de Ciclo Celular , Electrorretinografía , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mutagénesis/fisiología , Células Fotorreceptoras de Vertebrados/fisiología , Plásmidos/genética , Plásmidos/fisiología , Reacción en Cadena de la Polimerasa , ATPasas de Translocación de Protón/genética , Receptores de Superficie Celular/genética
11.
Am J Physiol Renal Physiol ; 305(3): F255-64, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23720344

RESUMEN

Mitochondria are both a source and target of the actions of reactive oxygen species and possess a complex system of inter-related antioxidants that control redox signaling and protect against oxidative stress. Interestingly, the antioxidant enzyme heme oxygenase-1 (HO-1) is not present in the mitochondria despite the fact that the organelle is the site of heme synthesis and contains multiple heme proteins. Detoxification of heme is an important protective mechanism since the reaction of heme with hydrogen peroxide generates pro-oxidant ferryl species capable of propagating oxidative stress and ultimately cell death. We therefore hypothesized that a mitochondrially localized HO-1 would be cytoprotective. To test this, we generated a mitochondria-targeted HO-1 cell line by transfecting HEK293 cells with a plasmid construct containing the manganese superoxide dismutase mitochondria leader sequence fused to HO-1 cDNA (Mito-HO-1). Nontargeted HO-1-overexpressing cells were generated by transfecting HO-1 cDNA (HO-1) or empty vector (Vector). Mitochondrial localization of HO-1 with increased HO activity in the mitochondrial fraction of Mito-HO-1 cells was observed, but a significant decrease in the expression of heme-containing proteins occurred in these cells. Both cytosolic HO-1- and Mito-HO-1-expressing cells were protected against hypoxia-dependent cell death and loss of mitochondrial membrane potential, but these effects were more pronounced with Mito-HO-1. Furthermore, decrement in production of tricarboxylic acid cycle intermediates following hypoxia was significantly mitigated in Mito-HO-1 cells. These data suggest that specific mitochondrially targeted HO-1 under acute pathological conditions may have beneficial effects, but the selective advantage of long-term expression is constrained by a negative impact on the synthesis of heme-containing mitochondrial proteins.


Asunto(s)
Células Epiteliales/metabolismo , Hemo-Oxigenasa 1/metabolismo , Riñón/metabolismo , Mitocondrias/enzimología , Aerobiosis/fisiología , Western Blotting , Supervivencia Celular/efectos de los fármacos , Citrato (si)-Sintasa/metabolismo , Ciclo del Ácido Cítrico/fisiología , Citocromos c/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Células Epiteliales/enzimología , Células HEK293 , Hemo-Oxigenasa 1/fisiología , Humanos , Inmunohistoquímica , Riñón/citología , Riñón/enzimología , Potencial de la Membrana Mitocondrial/fisiología , Estrés Oxidativo/fisiología , Plásmidos/genética , Plásmidos/fisiología , Canales Aniónicos Dependientes del Voltaje/metabolismo
12.
J Virol ; 87(7): 3699-709, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23325691

RESUMEN

Kaposi's Sarcoma-associated herpesvirus (KSHV) is maintained as a stable episome in latently infected pleural effusion lymphoma (PEL) cells. Episome maintenance is conferred by the binding of the KSHV-encoded LANA protein to the viral terminal repeats (TR). Here, we show that DNA replication in the KSHV TR is coupled with DNA recombination and mediated in part through the cellular replication fork protection factors Timeless (Tim) and Tipin. We show by two-dimensional (2D) agarose gel electrophoresis that replication forks naturally stall and form recombination-like structures at the TR during an unperturbed cell cycle. Chromatin immunoprecipitation (ChIP) assays revealed that Tim and Tipin are selectively enriched at the KSHV TR during S phase and in a LANA-dependent manner. Tim depletion inhibited LANA-dependent TR DNA replication and caused the loss of KSHV episomes from latently infected PEL cells. Tim depletion resulted in the aberrant accumulation of recombination structures and arrested MCM helicase at TR. Tim depletion did not induce the KSHV lytic cycle or apoptotic cell death. We propose that KSHV episome maintenance requires Tim-assisted replication fork protection at the viral terminal repeats and that Tim-dependent recombination-like structures form at TR to promote DNA repeat stability and viral genome maintenance.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN/fisiología , Inestabilidad Genómica/fisiología , Herpesvirus Humano 8/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Plásmidos/fisiología , Recombinación Genética/fisiología , Antígenos Virales/metabolismo , Bromodesoxiuridina , Proteínas Portadoras/metabolismo , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Proteínas de Unión al ADN , Electroforesis en Gel de Campo Pulsado , Electroforesis en Gel Bidimensional , Citometría de Flujo , Inestabilidad Genómica/genética , Humanos , Etiquetado Corte-Fin in Situ , Proteínas Nucleares/metabolismo , Plásmidos/genética , Secuencias Repetidas Terminales/genética , Secuencias Repetidas Terminales/fisiología
13.
Int J Biochem Cell Biol ; 44(12): 2223-32, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22999844

RESUMEN

Ubiquitylation is a widespread post-translational global regulatory system that is essential for the proper functioning of various cellular events. Recent studies have shown that certain types of Escherichia coli can exploit specific aspects of the ubiquitylation system to influence downstream targets. Despite these findings, examination of the effects pathogenic E. coli have on the overall host ubiquitylation system remain unexplored. To study the impact that pathogenic E. coli have on the ubiquitylation levels of host proteins during infections, we analyzed the entire ubiquitylation system during enteropathogenic E. coli infections of cultured cells. We found that these microbes caused a dramatic decrease in ubiquitylated host proteins during these infections. This occurred with a concomitant reduction in the expression of essential E1 activating enzymes in the host, which are integral for the initiation of the ubiquitylation cascade. Control of host E1 enzyme levels was dependent on the E. coli adherence factor plasmid which acted on host aspartyl proteases within enteropathogenic E. coli. Hijacking of the ubiquitylation system did not require the plasmid-encoded regulator or bundle forming pilus expression, as enteropathogenic E. coli mutated in those factors did not revert the ubiquitylation of host proteins or the abundance of E1 enzyme proteins to uninfected levels. Our work shows that E. coli have developed strategies to usurp post-translational systems by targeting crucial enzymes. The ability of enteropathogenic E. coli to inactivate host protein ubiquitylation could enable more efficient effector protein functionality, providing increased bacterial control of host cells during enteropathogenic E. coli pathogenesis.


Asunto(s)
Proteasas de Ácido Aspártico/metabolismo , Escherichia coli Enteropatógena/enzimología , Regulación Enzimológica de la Expresión Génica , Plásmidos/fisiología , Enzimas Activadoras de Ubiquitina/metabolismo , Sistemas de Secreción Bacterianos , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/fisiología , Células Epiteliales/enzimología , Células Epiteliales/microbiología , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Poliubiquitina/metabolismo , Tioléster Hidrolasas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Enzimas Activadoras de Ubiquitina/genética , Proteínas Ubiquitinadas/metabolismo , Ubiquitinación
14.
J Biol Chem ; 287(31): 26146-54, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22674577

RESUMEN

Segregation of the bacterial multidrug resistance plasmid TP228 requires the centromere-binding protein ParG, the parH centromere, and the Walker box ATPase ParF. The cycling of ParF between ADP- and ATP-bound states drives TP228 partition; ATP binding stimulates ParF polymerization, which is essential for segregation, whereas ADP binding antagonizes polymerization and inhibits DNA partition. The molecular mechanism involved in this adenine nucleotide switch is unclear. Moreover, it is unknown how any Walker box protein polymerizes in an ATP-dependent manner. Here, we describe multiple ParF structures in ADP- and phosphomethylphosphonic acid adenylate ester (AMPPCP)-bound states. ParF-ADP is monomeric but dimerizes when complexed with AMPPCP. Strikingly, in ParF-AMPPCP structures, the dimers interact to create dimer-of-dimer "units" that generate a specific linear filament. Mutation of interface residues prevents both polymerization and DNA segregation in vivo. Thus, these data provide insight into a unique mechanism by which a Walker box protein forms polymers that involves the generation of ATP-induced dimer-of-dimer building blocks.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfato/química , Proteínas Bacterianas/química , Proteínas de Unión al ADN/química , Plásmidos/fisiología , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , ADN Bacteriano/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Polarización de Fluorescencia , Datos de Secuencia Molecular , Plásmidos/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína
15.
Braz J Infect Dis ; 16(3): 262-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22729194

RESUMEN

OBJECTIVES: Plasmid pR ST98 is a hybrid resistance-virulence plasmid isolated from Salmonella enterica serovar Typhi (S. typhi). Previous studies demonstrated that pR ST98 could enhance the virulence of its host bacteria. However, the mechanism of pR ST98-increased bacterial virulence is still not fully elucidated. This study was designed to gain further insight into the roles of pR ST98 in host responses. METHODS: Human-derived macrophage-like cell line THP-1 was infected with wild-type (ST8), pR ST98-deletion (ST8-ΔpR ST98), and complemented (ST8-c-pR ST98) S. typhi strains. Macrophage autophagy was performed by extracting the membrane-unbound LC3-I protein from cells, followed by flow cytometric detection of the membrane-associated fraction of LC3-II. Intracellular bacterial growth was determined by colony-forming units (cfu) assay. Macrophage cell death was measured by flow cytometry after propidium iodide (PI) staining. Autophagy activator rapamycin (RAPA) was added to the medium 2 h before infection to investigate the effect of autophagy on intracellular bacterial growth and macrophage cell death after S. typhi infection. RESULTS: Plasmid pR ST98 suppressed autophagy in infected macrophages and enhanced intracellular bacterial growth and S. typhi-induced macrophage cell death. Pretreatment with RAPA effectively restricted intracellular bacterial growth of ST8 and ST8-c-pR ST98, and alleviated ST8 and ST8-c-pR ST98-induced macrophage cell death, but had no significant effect on ST8-ΔpR ST98. CONCLUSIONS: Plasmid pR ST98 enhances intracellular bacterial growth and S. typhi-induced macrophage cell death by suppressing autophagy.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Proteínas Bacterianas/fisiología , Macrófagos/microbiología , Plásmidos/fisiología , Salmonella typhi/fisiología , Células Cultivadas , Citometría de Flujo , Humanos , Salmonella typhi/crecimiento & desarrollo
16.
Braz. j. infect. dis ; Braz. j. infect. dis;16(3): 262-266, May-June 2012. ilus
Artículo en Inglés | LILACS | ID: lil-638560

RESUMEN

OBJECTIVES: Plasmid pR ST98 is a hybrid resistance-virulence plasmid isolated from Salmonella enterica serovar Typhi (S. typhi). Previous studies demonstrated that pR ST98 could enhance the virulence of its host bacteria. However, the mechanism of pR ST98-increased bacterial virulence is still not fully elucidated. This study was designed to gain further insight into the roles of pR ST98 in host responses. METHODS: Human-derived macrophage-like cell line THP-1 was infected with wild-type (ST8), pR ST98-deletion (ST8-ΔpR ST98), and complemented (ST8-c-pR ST98) S. typhi strains. Macrophage autophagy was performed by extracting the membrane-unbound LC3-I protein from cells, followed by flow cytometric detection of the membrane-associated fraction of LC3-II. Intracellular bacterial growth was determined by colony-forming units (cfu) assay. Macrophage cell death was measured by flow cytometry after propidium iodide (PI) staining. Autophagy activator rapamycin (RAPA) was added to the medium 2 h before infection to investigate the effect of autophagy on intracellular bacterial growth and macrophage cell death after S. typhi infection. RESULTS: Plasmid pR ST98 suppressed autophagy in infected macrophages and enhanced intracellular bacterial growth and S. typhi-induced macrophage cell death. Pretreatment with RAPA effectively restricted intracellular bacterial growth of ST8 and ST8-c-pR ST98, and alleviated ST8 and ST8-c-pR ST98-induced macrophage cell death, but had no significant effect on ST8-ΔpR ST98. CONCLUSIONS: Plasmid pR ST98 enhances intracellular bacterial growth and S. typhi-induced macrophage cell death by suppressing autophagy.


Asunto(s)
Humanos , Apoptosis/fisiología , Autofagia/fisiología , Proteínas Bacterianas/fisiología , Macrófagos/microbiología , Plásmidos/fisiología , Salmonella typhi/fisiología , Células Cultivadas , Citometría de Flujo , Salmonella typhi/crecimiento & desarrollo
17.
Proc Natl Acad Sci U S A ; 109(17): 6590-5, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22493268

RESUMEN

Poly(ADP-ribose) polymerase (PARP) inhibitors can generate synthetic lethality in cancer cells defective in homologous recombination. However, the mechanism(s) by which they affect DNA repair has not been established. Here we directly determined the effects of PARP inhibition and PARP1 depletion on the repair of ionizing radiation-induced single- and double-strand breaks (SSBs and DSBs) in human lymphoid cell lines. To do this, we developed an in vivo repair assay based on large endogenous Epstein-Barr virus (EBV) circular episomes. The EBV break assay provides the opportunity to assess quantitatively and simultaneously the induction and repair of SSBs and DSBs in human cells. Repair was efficient in G1 and G2 cells and was not dependent on functional p53. shRNA-mediated knockdown of PARP1 demonstrated that the PARP1 protein was not essential for SSB repair. Among 10 widely used PARP inhibitors, none affected DSB repair, although an inhibitor of DNA-dependent protein kinase was highly effective at reducing DSB repair. Only Olaparib and Iniparib, which are in clinical cancer therapy trials, as well as 4-AN inhibited SSB repair. However, a decrease in PARP1 expression reversed the ability of Iniparib to reduce SSB repair. Because Iniparib disrupts PARP1-DNA binding, the mechanism of inhibition does not appear to involve trapping PARP at SSBs.


Asunto(s)
Daño del ADN , Reparación del ADN , Herpesvirus Humano 4/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Western Blotting , Ciclo Celular , Inhibidores Enzimáticos/farmacología , Humanos , Rayos Infrarrojos , Plásmidos/fisiología , Reacción en Cadena de la Polimerasa , Proteína p53 Supresora de Tumor/fisiología
18.
PLoS One ; 7(1): e30747, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22292031

RESUMEN

Loss of the conserved "cryptic" plasmid from C. trachomatis and C. muridarum is pleiotropic, resulting in reduced innate inflammatory activation via TLR2, glycogen accumulation and infectivity. The more genetically distant C. caviae GPIC is a natural pathogen of guinea pigs and induces upper genital tract pathology when inoculated intravaginally, modeling human disease. To examine the contribution of pCpGP1 to C. caviae pathogenesis, a cured derivative of GPIC, strain CC13, was derived and evaluated in vitro and in vivo. Transcriptional profiling of CC13 revealed only partial conservation of previously identified plasmid-responsive chromosomal loci (PRCL) in C. caviae. However, 2-deoxyglucose (2DG) treatment of GPIC and CC13 resulted in reduced transcription of all identified PRCL, including glgA, indicating the presence of a plasmid-independent glucose response in this species. In contrast to plasmid-cured C. muridarum and C. trachomatis, plasmid-cured C. caviae strain CC13 signaled via TLR2 in vitro and elicited cytokine production in vivo similar to wild-type C. caviae. Furthermore, inflammatory pathology induced by infection of guinea pigs with CC13 was similar to that induced by GPIC, although we observed more rapid resolution of CC13 infection in estrogen-treated guinea pigs. These data indicate that either the plasmid is not involved in expression or regulation of virulence in C. caviae or that redundant effectors prevent these phenotypic changes from being observed in C. caviae plasmid-cured strains.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia/genética , Chlamydia/fisiología , Chlamydia/patogenicidad , Plásmidos/genética , Infecciones del Sistema Genital/microbiología , Receptor Toll-Like 2/fisiología , Virulencia/genética , Animales , Células Cultivadas , Chlamydia/inmunología , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/patología , Modelos Animales de Enfermedad , Evolución Molecular , Femenino , Eliminación de Gen , Cobayas , Células HEK293 , Humanos , Activación de Linfocitos/genética , Plásmidos/fisiología , Infecciones del Sistema Genital/inmunología , Infecciones del Sistema Genital/patología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo
19.
World J Surg ; 33(4): 685-97, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19023615

RESUMEN

We have developed improved liposomal nanoparticles that efficiently condense nucleic acids, proteins, viruses, drugs, and mixtures of these agents on the interior of bilamellar invaginated structures (BIVs) produced by a novel extrusion procedure. The liposomal complexes have extended half-life in the circulation, serum stability, and broad biodistribution; are targetable to specific organs and cell types; can penetrate through tight barriers in several organs; are fusogenic with cell membranes and avoid endosomes; are optimized for nucleic acid:lipid ratio and colloidal suspension in vivo; can be size fractionated to produce totally homogeneous populations of complexes prior to injection; are nontoxic, nonimmunogenic, and can be repeatedly administered; and they are stable in liquid suspensions and freeze-dried formulations. We can add specific ligands either by ionic interactions or by covalent attachments to the surface of these nucleic acid-liposome complexes to accomplish targeted delivery to specific cell surface receptors. Furthermore, the charge on the surface of these complexes can be modified to avoid uptake by nontarget cells using our novel technology called "reversible masking." We have also achieved high-dose systemic delivery of these complexes without toxicity in vivo by further purification of plasmid DNA. At present, these complexes are injected intravenously into patients in clinical trials to treat lung cancer and will be used in upcoming trials to treat breast, pancreatic, and head and neck cancers. Notably, BIV complexes are being injected intravenously into patients with non-small-cell lung carcinoma who have failed to respond to chemotherapy. These patients are living longer and have demonstrated objective responses, including tumor regression.


Asunto(s)
Terapia Genética , Vectores Genéticos , Liposomas/uso terapéutico , Neoplasias/genética , Animales , Biotecnología , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Islas de CpG/fisiología , Sistemas de Liberación de Medicamentos/métodos , Expresión Génica , Terapia Genética/métodos , Semivida , Humanos , Inmunoterapia/métodos , Liposomas/química , Liposomas/farmacología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Nanopartículas , Plásmidos/fisiología , Transfección/métodos
20.
Pflugers Arch ; 456(6): 1025-35, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18425533

RESUMEN

Kidney cysts are characterized by an abnormal tubular geometry that may result from loss of orientation and random cell divisions during renal development. Since cystic kidney disease is caused by mutations of ciliary proteins and cilia act as flow sensors in the kidney, we examined three polarized events in Madin Darby Canine Kidney cells under flow: cell division, cell migration, and centriole movement. We found that the mitotic orientation of dividing cells was not affected by flow and was randomly distributed in relation to the direction of the flow. Flow did not alter the direction and speed of cell migration in a wound-healing assay. However, flow resulted in increased motility of centrioles and biased centrioles to move along the axis of the flow. This effect was lost after flow-induced calcium signaling was abolished by a mutant polycystin 2. Our findings suggest that the cilium may translate fluid flow into altered centriole movements to provide tubular epithelial cells with the spatial orientation required to establish and/or maintain a normal tubular geometry.


Asunto(s)
Centriolos/fisiología , Corriente Citoplasmática/fisiología , Células Epiteliales/fisiología , Túbulos Renales/fisiología , Animales , División Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Centriolos/ultraestructura , Centrosoma/fisiología , Cilios/fisiología , Cilios/ultraestructura , Cámaras de Difusión de Cultivos , Perros , Células Epiteliales/ultraestructura , Colorantes Fluorescentes , Fura-2 , Indicadores y Reactivos , Túbulos Renales/citología , Túbulos Renales/metabolismo , Nefronas/citología , Nefronas/fisiología , Plásmidos/genética , Plásmidos/fisiología , Proteínas/metabolismo , Cicatrización de Heridas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA