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











Base de datos
Intervalo de año de publicación
1.
mBio ; 15(6): e0023024, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38682935

RESUMEN

Strict management of intracellular heme pools, which are both toxic and beneficial, is crucial for bacterial survival during infection. The human pathogen Staphylococcus aureus uses a two-component heme sensing system (HssRS), which counteracts environmental heme toxicity by triggering expression of the efflux transporter HrtBA. The HssS heme sensor is a HisKA-type histidine kinase, characterized as a membrane-bound homodimer containing an extracellular sensor and a cytoplasmic conserved catalytic domain. To elucidate HssS heme-sensing mechanism, a structural simulation of the HssS dimer based on Alphafold2 was docked with heme. In this model, a heme-binding site is present in the HssS dimer between the membrane and extracellular domains. Heme is embedded in the membrane bilayer with its two protruding porphyrin propionates interacting with two conserved Arg94 and Arg163 that are located extracellularly. Single substitutions of these arginines and two highly conserved phenylalanines, Phe25 and Phe128, in the predicted hydrophobic pocket limited the ability of HssS to induce HrtBA synthesis. Combination of the four substitutions abolished HssS activation. Wild-type (WT) HssS copurified with heme from Escherichia coli, whereas heme binding was strongly attenuated in the variants. This study gives evidence that exogenous heme interacts with HssS at the membrane/extracellular interface to initiate HssS activation and induce HrtBA-mediated heme extrusion from the membrane. This "gatekeeper" mechanism could limit intracellular diffusion of exogenous heme in S. aureus and may serve as a paradigm for how efflux transporters control detoxification of exogenous hydrophobic stressors.IMPORTANCEIn the host blood, pathogenic bacteria are exposed to the red pigment heme that concentrates in their lipid membranes, generating cytotoxicity. To overcome heme toxicity, Staphylococcus aureus expresses a membrane sensor protein, HssS. Activation of HssS by heme triggers a phosphotransfer mechanism leading to the expression of a heme efflux system, HrtBA. This detoxification system prevents intracellular accumulation of heme. Our structural and functional data reveal a heme-binding hydrophobic cavity in HssS within the transmembrane domains (TM) helices at the interface with the extracellular domain. This structural pocket is important for the function of HssS as a heme sensor. Our findings provide a new basis for the elucidation of pathogen-sensing mechanisms as a prerequisite to the discovery of inhibitors.


Asunto(s)
Proteínas Bacterianas , Hemo , Transducción de Señal , Staphylococcus aureus , Hemo/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Histidina Quinasa/metabolismo , Histidina Quinasa/genética , Histidina Quinasa/química , Regulación Bacteriana de la Expresión Génica , Sitios de Unión , Membrana Celular/metabolismo
2.
Virulence ; 13(1): 1221-1241, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35880611

RESUMEN

Bacterial pathogens have a critical impact on aquaculture, a sector that accounts for half of the human fish consumption. Flavobacterium psychrophilum (phylum Bacteroidetes) is responsible for bacterial cold-water disease in salmonids worldwide. The molecular factors involved in host invasion, colonization and haemorrhagic septicaemia are mostly unknown. In this study, we identified two new TonB-dependent receptors, HfpR and BfpR, that are required for adaptation to iron conditions encountered during infection and for virulence in rainbow trout. Transcriptional analyses revealed that their expression is tightly controlled and upregulated under specific iron sources and concentrations. Characterization of deletion mutants showed that they act without redundancy: BfpR is required for optimal growth in the presence of high haemoglobin level, while HfpR confers the capacity to acquire nutrient iron from haem or haemoglobin under iron scarcity. The gene hfpY, co-transcribed with hfpR, encodes a protein related to the HmuY family. We demonstrated that HfpY binds haem and contributes significantly to host colonization and disease severity. Overall, these results are consistent with a model in which both BfpR and Hfp systems promote haem uptake and respond to distinct signals to adapt iron acquisition to the different stages of pathogenesis. Our findings give insight into the molecular basis of pathogenicity of a serious pathogen belonging to the understudied family Flavobacteriaceae and point to the newly identified haem receptors as promising targets for antibacterial development.


Asunto(s)
Enfermedades de los Peces , Infecciones por Flavobacteriaceae , Oncorhynchus mykiss , Animales , Enfermedades de los Peces/microbiología , Infecciones por Flavobacteriaceae/microbiología , Infecciones por Flavobacteriaceae/veterinaria , Flavobacterium , Hemo/metabolismo , Humanos , Hierro/metabolismo , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/microbiología , Virulencia , Factores de Virulencia/genética
3.
mBio ; 12(1)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531389

RESUMEN

Enterococcus faecalis is a commensal Gram-positive pathogen found in the intestines of mammals and is also a leading cause of severe infections occurring mainly among antibiotic-treated dysbiotic hospitalized patients. Like most intestinal bacteria, E. faecalis does not synthesize heme (in this report, heme refers to iron protoporphyrin IX regardless of the iron redox state). Nevertheless, environmental heme can improve E. faecalis fitness by activating respiration metabolism and a catalase that limits hydrogen peroxide stress. Since free heme also generates toxicity, its intracellular levels need to be strictly controlled. Here, we describe a unique transcriptional regulator, FhtR (named FhtR for faecalis heme transport regulator), which manages heme homeostasis by controlling an HrtBA-like efflux pump (named HrtBA Ef for the HrtBA from E. faecalis). We show that FhtR, by managing intracellular heme concentration, regulates the functional expression of the heme-dependent catalase A (KatA), thus participating in heme detoxification. The biochemical features of FhtR binding to DNA, and its interaction with heme that induces efflux, are characterized. The FhtR-HrtBA Ef system is shown to be relevant in a mouse intestinal model. We further show that FhtR senses heme from blood and hemoglobin but also from crossfeeding by Escherichia coli These findings bring to light the central role of heme sensing by FhtR in response to heme fluctuations within the gastrointestinal tract, which allow this pathogen to limit heme toxicity while ensuring expression of an oxidative defense system.IMPORTANCEEnterococcus faecalis, a normal and harmless colonizer of the human intestinal flora can cause severe infectious diseases in immunocompromised patients, particularly those that have been heavily treated with antibiotics. Therefore, it is important to understand the factors that promote its resistance and its virulence. E. faecalis, which cannot synthesize heme, an essential but toxic metabolite, needs to scavenge this molecule from the host to respire and fight stress generated by oxidants. Here, we report a new mechanism used by E. faecalis to sense heme and trigger the synthesis of a heme efflux pump that balances the amount of heme inside the bacteria. We show in a mouse model that E. faecalis uses this mechanisms within the gastrointestinal tract.


Asunto(s)
Proteínas Bacterianas/fisiología , Enterococcus faecalis/metabolismo , Hemo/metabolismo , Animales , Femenino , Tracto Gastrointestinal/microbiología , Homeostasis , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas
4.
Microbiol Spectr ; 7(4)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31298208

RESUMEN

Lactococcus lactis is the best characterized species among the lactococci, and among the most consumed food-fermenting bacteria worldwide. Thanks to their importance in industrialized food production, lactococci are among the lead bacteria understood for fundamental metabolic pathways that dictate growth and survival properties. Interestingly, lactococci belong to the Streptococcaceae family, which includes food, commensal and virulent species. As basic metabolic pathways (e.g., respiration, metal homeostasis, nucleotide metabolism) are now understood to underlie virulence, processes elucidated in lactococci could be important for understanding pathogen fitness and synergy between bacteria. This chapter highlights major findings in lactococci and related bacteria, and covers five themes: distinguishing features of lactococci, metabolic capacities including the less known respiration metabolism in Streptococcaceae, factors and pathways modulating stress response and fitness, interbacterial dialogue via metabolites, and novel applications in health and biotechnology.


Asunto(s)
Lactococcus lactis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fermentación , Lactococcus lactis/clasificación , Lactococcus lactis/metabolismo , Redes y Vías Metabólicas
5.
Sci Rep ; 7: 40435, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28091535

RESUMEN

Heme is essential for several cellular key functions but is also toxic. Whereas most bacterial pathogens utilize heme as a metabolic cofactor and iron source, the impact of host heme during bacterial infection remains elusive. The opportunist pathogen Streptococcus agalactiae does not synthesize heme but still uses it to activate a respiration metabolism. Concomitantly, heme toxicity is mainly controlled by the HrtBA efflux transporter. Here we investigate how S. agalactiae manages heme toxicity versus benefits in the living host. Using bioluminescent bacteria and heme-responsive reporters for in vivo imaging, we show that the capacity of S. agalactiae to overcome heme toxicity is required for successful infection, particularly in blood-rich organs. Host heme is simultaneously required, as visualized by a generalized infection defect of a respiration-negative mutant. In S. agalactiae, HrtBA expression responds to an intracellular heme signal via activation of the two-component system HssRS. A hssRS promoter-driven intracellular luminescent heme sensor was designed to identify host compartments that supply S. agalactiae with heme. S. agalactiae acquires heme in heart, kidneys, and liver, but not in the brain. We conclude that S. agalactiae response to heme is organ-dependent, and its efflux may be particularly relevant in late stages of infection.


Asunto(s)
Hemo/metabolismo , Streptococcus agalactiae/patogenicidad , Aerobiosis/efectos de los fármacos , Animales , Genes Bacterianos , Hemo/toxicidad , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Ratones , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/metabolismo , Virulencia/efectos de los fármacos
6.
Mol Microbiol ; 93(4): 823-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25040434

RESUMEN

Lactococcus lactis is a fermenting Gram-positive bacterium widely used for production of dairy products. Lacking haem biosynthesis genes, L. lactis can still shift to an energetically favourable respiratory metabolism by activating a terminal cytochrome bd oxidase when haem is added to an aerated culture. Haem intracellular homeostasis is mediated by the hrtRBA operon encoding the conserved membrane HrtBA haem efflux permease and the unique intracellular haem sensor and regulator, HrtR. Here we report that membrane-associated menaquinones (MK) favour the accumulation of reduced haem in membranes. An oxidative environment, provided by oxygen, prevents and reverses haemin reduction by MK and thus limits haem accumulation in membranes. HrtBA counteracts MK-dependent membrane retention of excess haem in membrane, suggesting direct efflux from this compartment. Moreover, both HrtBA and MK-mediated reduction have a strong impact on haem intracellular pools, as determined via HrtR haem sensor induction, suggesting that intracellular haem acquisition is controlled at the membrane level without the need for dedicated import systems. Our conclusions lead to a new hypothesis of haem acquisition and regulation in which HrtBA and the bacterial membrane have central roles in L. lactis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemo/metabolismo , Lactococcus lactis/metabolismo , Vitamina K 2/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Citosol/química , Homeostasis , Oxidación-Reducción
7.
Adv Microb Physiol ; 61: 69-124, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23046952

RESUMEN

Heme, an iron-containing porphyrin, is the prosthetic group for numerous key cellular enzymatic and regulatory processes. Many bacteria encode the biosynthetic enzymes needed for autonomous heme production. Remarkably, however, numerous other bacteria lack a complete heme biosynthesis pathway, yet encode heme-requiring functions. For such heme-auxotrophic bacteria (HAB), heme or porphyrins must be captured from the environment. Functional studies, aided by genomic analyses, provide insight into the HAB lifestyle, how they acquire and manage heme, and the uses of heme that make it worthwhile, and sometimes necessary, to capture this bioactive molecule.


Asunto(s)
Bacterias/metabolismo , Hemo/metabolismo , Animales , Procesos Autotróficos , Bacterias/enzimología , Bacterias/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Hemo/biosíntesis , Humanos , Porfirinas/metabolismo
8.
Annu Rev Food Sci Technol ; 3: 37-58, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22385163

RESUMEN

The lactic acid bacteria (LAB) are essential for food fermentations and their impact on gut physiology and health is under active exploration. In addition to their well-studied fermentation metabolism, many species belonging to this heterogeneous group are genetically equipped for respiration metabolism. In LAB, respiration is activated by exogenous heme, and for some species, heme and menaquinone. Respiration metabolism increases growth yield and improves fitness. In this review, we aim to present the basics of respiration metabolism in LAB, its genetic requirements, and the dramatic physiological changes it engenders. We address the question of how LAB acquired the genetic equipment for respiration. We present at length how respiration can be used advantageously in an industrial setting, both in the context of food-related technologies and in novel potential applications.


Asunto(s)
Microbiología de Alimentos , Tecnología de Alimentos , Lactobacillaceae/metabolismo , Biotecnología/tendencias , Fermentación , Microbiología de Alimentos/tendencias , Tecnología de Alimentos/tendencias , Fenómenos Genéticos , Genética Microbiana/métodos , Lactobacillaceae/genética , Lactobacillaceae/crecimiento & desarrollo , Oxígeno/metabolismo , Filogenia , Probióticos/metabolismo , Especificidad de la Especie
9.
J Biol Chem ; 287(7): 4752-8, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22084241

RESUMEN

Most commensal and food bacteria lack heme biosynthesis genes. For several of these, the capture of environmental heme is a means of activating aerobic respiration metabolism. Our previous studies in the Gram-positive bacterium Lactococcus lactis showed that heme exposure strongly induced expression of a single operon, called here hrtRBA, encoding an ortholog of the conserved membrane hrt (heme-regulated transporter) and a unique transcriptional regulator that we named HrtR. We show that HrtR expressed as a fusion protein is a heme-binding protein. Heme iron interaction with HrtR is non-covalent, hexacoordinated, and involves two histidines, His-72 and His-149. HrtR specifically binds a 15-nt palindromic sequence in the hrtRBA promoter region, which is needed for hrtRBA repression. HrtR-DNA binding is abolished by heme addition, which activates expression of the HrtB-HrtA (HrtBA) transporter in vitro and in vivo. The use of HrtR as an intracellular heme sensor appears to be conserved among numerous commensal bacteria, in contrast with numerous Gram-positive pathogens that use an extracellular heme-sensing system, HssRS, to regulate hrt. Finally, we show for the first time that HrtBA permease controls heme toxicity by its direct and specific efflux. The use of an intracellular heme sensor to control heme efflux constitutes a novel paradigm for bacterial heme homeostasis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Hemo/metabolismo , Hemoproteínas/metabolismo , Lactococcus lactis/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/genética , Transporte Biológico Activo/fisiología , Proteínas Portadoras/genética , Hemo/genética , Proteínas de Unión al Hemo , Hemoproteínas/genética , Lactococcus lactis/genética , Proteínas de Transporte de Membrana/genética , Operón/fisiología
10.
Curr Opin Biotechnol ; 22(2): 143-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21211959

RESUMEN

Lactic acid bacteria (LAB) are a phylogenetically diverse group named for their main attribute in food fermentations, that is, production of lactic acid. However, several LAB are genetically equipped for aerobic respiration metabolism when provided with exogenous sources of heme (and menaquinones for some species). Respiration metabolism is energetically favorable and leads to less oxidative and acid stress during growth. As a consequence, the growth and survival of several LAB can be dramatically improved under respiration-permissive conditions. Respiration metabolism already has industrial applications for the production of dairy starter cultures. In view of the growth and survival advantages conferred by respiration, and the availability of heme and menaquinones in natural environments, we recommend that respiration be accepted as a part of the natural lifestyle of numerous LAB.


Asunto(s)
Bacterias/metabolismo , Hemo/metabolismo , Ácido Láctico/biosíntesis , Lactobacillaceae/metabolismo
11.
PLoS Pathog ; 6(4): e1000860, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20421944

RESUMEN

Streptococcus agalactiae is a major neonatal pathogen whose infectious route involves septicemia. This pathogen does not synthesize heme, but scavenges it from blood to activate a respiration metabolism, which increases bacterial cell density and is required for full virulence. Factors that regulate heme pools in S. agalactiae are unknown. Here we report that one main strategy of heme and protoporphyrin IX (PPIX) homeostasis in S. agalactiae is based on a regulated system of efflux using two newly characterized operons, gbs1753 gbs1752 (called pefA pefB), and gbs1402 gbs1401 gbs1400 (called pefR pefC pefD), where pef stands for 'porphyrin-regulated efflux'. In vitro and in vivo data show that PefR, a MarR-superfamily protein, is a repressor of both operons. Heme or PPIX both alleviate PefR-mediated repression. We show that bacteria inactivated for both Pef efflux systems display accrued sensitivity to these porphyrins, and give evidence that they accumulate intracellularly. The DeltapefR mutant, in which both pef operons are up-regulated, is defective for heme-dependent respiration, and attenuated for virulence. We conclude that this new efflux regulon controls intracellular heme and PPIX availability in S. agalactiae, and is needed for its capacity to undergo respiration metabolism, and to infect the host.


Asunto(s)
Hemo/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Protoporfirinas/metabolismo , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Northern Blotting , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Proteínas de Transporte de Membrana/genética , Ratones , Operón , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Streptococcus agalactiae/genética
12.
J Biol Chem ; 285(21): 16032-41, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20332091

RESUMEN

Heme is a redox-reactive molecule with vital and complex roles in bacterial metabolism, survival, and virulence. However, few intracellular heme partners were identified to date and are not well conserved in bacteria. The opportunistic pathogen Streptococcus agalactiae (group B Streptococcus) is a heme auxotroph, which acquires exogenous heme to activate an aerobic respiratory chain. We identified the alkyl hydroperoxide reductase AhpC, a member of the highly conserved thiol-dependent 2-Cys peroxiredoxins, as a heme-binding protein. AhpC binds hemin with a K(d) of 0.5 microm and a 1:1 stoichiometry. Mutagenesis of cysteines revealed that hemin binding is dissociable from catalytic activity and multimerization. AhpC reductase activity was unchanged upon interaction with heme in vitro and in vivo. A group B Streptococcus ahpC mutant displayed attenuation of two heme-dependent functions, respiration and activity of a heterologous catalase, suggesting a role for AhpC in heme intracellular fate. In support of this hypothesis, AhpC-bound hemin was protected from chemical degradation in vitro. Our results reveal for the first time a role for AhpC as a heme-binding protein.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemo/metabolismo , Peroxirredoxinas/metabolismo , Streptococcus agalactiae/enzimología , Proteínas Bacterianas/genética , Hemo/genética , Mutagénesis/fisiología , Mutación , Peroxirredoxinas/genética , Unión Proteica , Streptococcus agalactiae/genética
13.
Hum Gene Ther ; 17(9): 882-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16972756

RESUMEN

Nonviral vectors represent a promising approach for the safe delivery of therapeutic DNA in genetic and acquired human diseases. Before synthetic vector systems can be used for clinical applications, their limited efficacy must be addressed. At the cellular level, successful gene transfer is dependent on several additional factors including DNA uptake, release from the DNA-vector complex, and nucleocytoplasmic transport. This paper reviews the major metabolic and physical impediments that plasmid DNA vectorized by synthetic vectors encounters between the cytosol and the nucleus. Plasmid DNA that escapes the endolysosomal compartment encounters the diffusional and metabolic barriers of the cytoplasm, reducing the number of intact plasmids that reach the nuclear envelope. Nuclear translocation of DNA requires either the disassembly of the nuclear envelope during cell division or active nuclear transport via the nuclear pore complex. In the nucleus, plasmid DNA is relatively stable, but its transcription and its fate during cell division are still debated. A better understanding of the cellular and molecular basis of nonviral gene transfer during nucleocytoplasmic trafficking may provide strategies to overcome those obstacles that limit the efficiency of nonviral gene delivery. We review some of the current methods of gene transfer mediated by synthetic vectors, highlighting systems that exploit our actual knowledge of the nucleocytoplasmic transport of plasmid DNA.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , ADN/metabolismo , Plásmidos/metabolismo , Transporte Biológico
14.
Mol Biol Cell ; 17(6): 2513-23, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16554368

RESUMEN

The endosomal sorting complexes required for transport, ESCRT-I, -II, and -III, are thought to mediate the biogenesis of multivesicular endosomes (MVEs) and endosomal sorting of ubiquitinated membrane proteins. Here, we have compared the importance of the ESCRT-I subunit tumor susceptibility gene 101 (Tsg101) and the ESCRT-III subunit hVps24/CHMP3 for endosomal functions and receptor signaling. Like Tsg101, endogenous hVps24 localized mainly to late endosomes. Depletion of hVps24 by siRNA showed that this ESCRT subunit, like Tsg101, is important for degradation of the epidermal growth factor (EGF) receptor (EGFR) and for transport of the receptor from early endosomes to lysosomes. Surprisingly, however, whereas depletion of Tsg101 caused sustained EGF activation of the mitogen-activated protein kinase pathway, depletion of hVps24 had no such effect. Moreover, depletion of Tsg101 but not of hVps24 caused a major fraction of internalized EGF to accumulate in nonacidified endosomes. Electron microscopy of hVps24-depleted cells showed an accumulation of EGFRs in MVEs that were significantly smaller than those in control cells, probably because of an impaired fusion with lyso-bisphosphatidic acid-positive late endosomes/lysosomes. Together, our results reveal functional differences between ESCRT-I and ESCRT-III in degradative protein trafficking and indicate that degradation of the EGFR is not required for termination of its signaling.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Regulación hacia Abajo , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte , Endosomas/metabolismo , Endosomas/ultraestructura , Receptores ErbB/genética , Silenciador del Gen , Células HeLa , Humanos , Lisosomas/metabolismo , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética
15.
Traffic ; 7(3): 282-97, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16497223

RESUMEN

Ubiquitination induced down-regulation of cell surface proteins by internalization and lysosomal targeting plays a fundamental role in cell physiology and pathogenesis of diseases. The molecular basis of a single ubiquitin (Ub) as an autonomous endocytic signal, the widely accepted mechanism, however, remains elusive in higher eukaryotes. Using Ub containing reporter proteins without signalling abilities, we present evidence that only multiple Ub moieties, linked either covalently or assembled as oligomers with an intact interface for recognition by Ub-interacting motifs (UIMs), are recognized by the endocytic machinery in vivo and associate with a subset of Ub-binding clathrin adaptors in vitro. Genetic and pharmacological approaches show that internalization of plasma membrane proteins harbouring multiple Ub moieties is clathrin-dependent, but caveolin-independent. Functional assays demonstrate the cargo-dependent involvement of eps15/15R and epsin, UIM containing clathrin adaptors, in the endocytosis of model proteins, CD4 and the activated beta(2)-adrenergic receptor complex, containing polymeric or oligomeric Ub. These results provide a paradigm for the clathrin-mediated uptake of ubiquitinated membrane proteins in mammalian cells, requiring the assembly of multiple UIM-Ub interactions to overcome the low affinity binding of mono-Ub to UIM.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Proteínas de Unión al Calcio/fisiología , Clatrina/fisiología , Endocitosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/fisiología , Poliubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Western Blotting , Antígenos CD4/metabolismo , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Clatrina/genética , Cricetinae , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Modelos Biológicos , Poliubiquitina/química , Pruebas de Precipitina , Proteínas Recombinantes de Fusión/metabolismo
16.
J Biol Chem ; 280(48): 40216-25, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16204257

RESUMEN

The caspase-activated DNase (CAD) is the primary nuclease responsible for oligonucleosomal DNA fragmentation during apoptosis. The DNA fragmentation factor (DFF) is composed of the 40-kDa CAD (DFF40) in complex with its cognate 45-kDa inhibitor (inhibitor of CAD: ICAD or DFF45). The association of ICAD with CAD not only inhibits the DNase activity but is also essential for the co-translational folding of CAD. Activation of CAD requires caspase-3-dependent proteolysis of ICAD. The tertiary structures of neither the inactive nor the activated DFF have been conclusively established. Whereas the inactive DFF is thought to consist of the CAD/ICAD heterodimer, activated CAD has been isolated as a large (>MDa) multimer, as well as a monomer. To establish the subunit stoichiometry of DFF and some of its structural determinants in normal and apoptotic cells, we utilized size-exclusion chromatography in combination with co-immunoprecipitation and mutagenesis techniques. Both endogenous and heterologously expressed DFF have an apparent molecular mass of 160-190 kDa and contain 2 CAD and 2 ICAD molecules (CAD/ICAD)2 in HeLa cells. Although the N-terminal (CIDE-N) domain of CAD is not required for ICAD binding, it is necessary but not sufficient for ICAD homodimerization in the DFF. In contrast, the CIDE-N domain of ICAD is required for CAD/ICAD association. Using bioluminescence resonance energy transfer (BRET), dimerization of ICAD in DFF was confirmed in live cells. In apoptotic cells, endogenous and exogenous CAD forms limited oligomers, representing the active nuclease. A model is proposed for the rearrangement of the DFF subunit stoichiometry in cells undergoing programmed cell death.


Asunto(s)
Apoptosis , Desoxirribonucleasas/química , Animales , Células COS , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Chlorocebus aethiops , Cromatografía , Cromatografía en Gel , Fragmentación del ADN , Desoxirribonucleasas/metabolismo , Dimerización , Activación Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Mutagénesis , Plásmidos/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Biosíntesis de Proteínas , Pliegue de Proteína , Estructura Terciaria de Proteína , Transfección
17.
Adv Drug Deliv Rev ; 57(5): 755-67, 2005 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-15757759

RESUMEN

Gene transfer using non-viral vectors is a promising approach for the safe delivery of therapeutic DNA in genetic and acquired human diseases. Whereas the lack of specific immune response favors the use of plasmid-cationic polymer complexes, the limited efficacy and short duration of transgene expression impose major hurdles in the application of non-viral gene delivery techniques. Here, we review the major cellular, metabolic and physico-chemical impediments that non-viral vectors encounter before plasmid DNA enters the nucleus. Following endocytosis of DNA-polycation complexes, a large fraction of the DNA is targeted to the lysosomes. Since the cytosolic release of heterologous DNA is a prerequisite for nuclear translocation, entrapment and degradation of plasmid DNA in endo-lysosomes constitute one of the major impediments to efficient gene transfer. Plasmid DNA that escapes the endo-lysosomal compartment encounters the diffusional and metabolic barriers of the cytoplasm, reducing greatly the number of intact plasmids that reach the nucleosol. Nuclear translocation of DNA requires either the disassembly of the nuclear envelope or active nuclear transport via the nuclear pore complex. A better understanding of the cellular and molecular basis of non-viral vector trafficking from the extracellular compartment into the nucleus may provide strategies to overcome those obstacles that limit the efficiency of gene delivery.


Asunto(s)
ADN Circular/farmacocinética , Técnicas de Transferencia de Gen , Líquido Intracelular/metabolismo , Plásmidos , Animales , Humanos
18.
J Cell Biol ; 167(5): 851-62, 2004 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-15569712

RESUMEN

Although compelling evidence supports the central role of caspase-activated DNase (CAD) in oligonucleosomal DNA fragmentation in apoptotic nuclei, the regulation of CAD activity remains elusive in vivo. We used fluorescence photobleaching and biochemical techniques to investigate the molecular dynamics of CAD. The CAD-GFP fusion protein complexed with its inhibitor (ICAD) was as mobile as nuclear GFP in the nucleosol of dividing cells. Upon induction of caspase-3-dependent apoptosis, activated CAD underwent progressive immobilization, paralleled by its attenuated extractability from the nucleus. CAD immobilization was mediated by its NH2 terminus independently of its DNA-binding activity and correlated with its association to the interchromosomal space. Preventing the nuclear attachment of CAD provoked its extracellular release from apoptotic cells. We propose a novel paradigm for the regulation of CAD in the nucleus, involving unrestricted accessibility of chromosomal DNA at the initial phase of apoptosis, followed by its nuclear immobilization that may prevent the release of the active nuclease into the extracellular environment.


Asunto(s)
Apoptosis/genética , División Celular/genética , Núcleo Celular/enzimología , Desoxirribonucleasas/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Células COS , Caspasa 3 , Caspasas/genética , Caspasas/metabolismo , Núcleo Celular/genética , Cricetinae , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Células HeLa , Humanos , Ratones , Estructura Terciaria de Proteína/genética , Transporte de Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
Curr Gene Ther ; 2(2): 183-94, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12109215

RESUMEN

Non-viral vector mediated gene transfer, compared to viral vector mediated one, is a promising tool for the safe delivery of therapeutic DNA in genetic and acquired human diseases. Although the lack of specific immune response favor the clinical application of non-viral vectors, comprising of an expression cassette complexed to cationic liposome or cationic polymer, the limited efficacy and short duration of transgene expression impose major hurdles in the widespread application of non-viral gene therapy. The trafficking of transgene, complexed with chemical vectors, has been the subject of intensive investigations to improve our understanding of cellular and extracellular barriers impeding gene delivery. Here, we review those physical and metabolic impediments that account, at least in part, for the inefficient translocation of transgene into the nucleus of target cells. Following the internalization of the DNA-polycation complex by endocytosis, a large fraction is targeted to the lysosomal compartment by default. Since the cytosolic release of heterelogous DNA is a prerequisite for nuclear translocation, entrapment and degradation of plasmid DNA in endo-lysosomes constitute a major impediment to efficient gene transfer. Only a small fraction of internalized plasmid DNA penetrates the cytoplasm. Plasmid DNA encounters the diffusional and metabolic barriers of the cytoplasm, further decreasing the number of intact plasmid molecules reaching the nuclear pore complex (NPC), the gateway of nucleosol. Nuclear translocation of DNA requires either the disassembly of the nuclear envelope or active nuclear transport via the NPC. Comparison of viral and plasmid DNA cellular trafficking should reveal strategies that viruses have developed to overcome those cellular barriers that impede non-viral DNA delivery in gene therapy attempts.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/metabolismo , ADN/metabolismo , ADN Viral/metabolismo , Endocitosis , Humanos , Microscopía Fluorescente , Modelos Biológicos , Modelos Genéticos , Plásmidos/genética , Plásmidos/metabolismo , Virus/genética
20.
Mol Ther ; 5(2): 161-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11829523

RESUMEN

In this study we have compared the process of lipid-mediated transfection in primary and established myoblasts, in an attempt to elucidate the mechanisms responsible for the scarce transfectability of the former. We determined the metabolic stability of cytoplasmically injected and lipofected DNA in primary and established myoblasts and carried out a comparative time course analysis of luciferase reporter-gene expression and DNA stability. The efficiency of the transcription-translation machinery of the two cell types was compared by intranuclear injection of naked plasmid DNA encoding luciferase. Subcellular colocalization of fluorescein-labeled lipopolyplexes with specific endosomal and lysosomal markers was performed by confocal microscopy to monitor the intracellular trafficking of plasmid DNA during transfection. The metabolic stability of plasmid DNA was similar in primary and established myoblasts after both lipofection and cytoplasmic injection. In both cell types, lipofection had no detectable effect on the rate of cell proliferation. Confocal analysis showed that nuclear translocation of transfected DNA coincided with localization in a compartment devoid of endosome- or lysosome-specific marker proteins. The residency time of plasmid DNA in this compartment differed for primary and established myoblasts. Our findings suggest that the lower transfectability of primary myoblasts is mostly due to a difference in the intracellular delivery pathway that correlates with more rapid delivery of internalized complex to the lysosomal compartment.


Asunto(s)
Liposomas , Músculo Esquelético/citología , Transfección , División Celular , Células Cultivadas , ADN , Feto , Expresión Génica , Humanos , Luciferasas/genética , Músculo Esquelético/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA