Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
EMBO Rep ; 19(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29945933

RESUMEN

Reducing insulin/IGF-1 signaling (IIS) extends lifespan, promotes protein homeostasis (proteostasis), and elevates stress resistance of worms, flies, and mammals. How these functions are orchestrated across the organism is only partially understood. Here, we report that in the nematode Caenorhabditis elegans, the IIS positively regulates the expression of caveolin-1 (cav-1), a gene which is primarily expressed in neurons of the adult worm and underlies the formation of caveolae, a subtype of lipid microdomains that serve as platforms for signaling complexes. Accordingly, IIS reduction lowers cav-1 expression and lessens the quantity of neuronal caveolae. Reduced cav-1 expression extends lifespan and mitigates toxic protein aggregation by modulating the expression of aging-regulating and signaling-promoting genes. Our findings define caveolae as aging-governing signaling centers and underscore the potential for cav-1 as a novel therapeutic target for the promotion of healthy aging.


Asunto(s)
Envejecimiento/metabolismo , Caenorhabditis elegans/metabolismo , Caveolas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Transducción de Señal , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/ultraestructura , Proteínas de Caenorhabditis elegans/metabolismo , Caveolas/ultraestructura , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Respuesta al Choque Térmico , Longevidad , Modelos Biológicos , Proteostasis , Interferencia de ARN , Factores de Transcripción/metabolismo , Rayos Ultravioleta
2.
EMBO J ; 34(22): 2820-39, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26438723

RESUMEN

Do different neurodegenerative maladies emanate from the failure of a mutual protein folding mechanism? We have addressed this question by comparing mutational patterns that are linked to the manifestation of distinct neurodegenerative disorders and identified similar neurodegeneration-linked proline substitutions in the prion protein and in presenilin 1 that underlie the development of a prion disorder and of familial Alzheimer's disease (fAD), respectively. These substitutions were found to prevent the endoplasmic reticulum (ER)-resident chaperone, cyclophilin B, from assisting presenilin 1 to fold properly, leading to its aggregation, deposition in the ER, reduction of γ-secretase activity, and impaired mitochondrial distribution and function. Similarly, reduced quantities of the processed, active presenilin 1 were observed in brains of cyclophilin B knockout mice. These discoveries imply that reduced cyclophilin activity contributes to the development of distinct neurodegenerative disorders, propose a novel mechanism for the development of certain fAD cases, and support the emerging theme that this disorder can stem from aberrant presenilin 1 function. This study also points at ER chaperones as targets for the development of counter-neurodegeneration therapies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Encéfalo/metabolismo , Presenilina-1/metabolismo , Agregación Patológica de Proteínas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Línea Celular , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Presenilina-1/genética , Prolina/genética , Prolina/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Pliegue de Proteína
3.
J Neurosci ; 33(14): 6102-11, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23554491

RESUMEN

In the nematode Caenorhabditis elegans, the heat shock response (HSR) is regulated at the organismal level by a network of thermosensory neurons that senses elevated temperatures and activates the HSR in remote tissues. Which neuronal receptors are required for this signaling mechanism and in which neurons they function are largely unanswered questions. Here we used worms that were engineered to exhibit RNA interference hypersensitivity in neurons to screen for neuronal receptors that are required for the activation of the HSR and identified a putative G-protein coupled receptor (GPCR) as a novel key component of this mechanism. This gene, which we termed GPCR thermal receptor 1 (gtr-1), is expressed in chemosensory neurons and has no role in heat sensing but is critically required for the induction of genes that encode heat shock proteins in non-neural tissues upon exposure to heat. Surprisingly, the knock-down of gtr-1 by RNA interference protected worms expressing the Alzheimer's-disease-linked aggregative peptide Aß3-42 from proteotoxicity but had no effect on lifespan. This study provides several novel insights: (1) it shows that chemosensory neurons play important roles in the nematode's HSR-regulating mechanism, (2) it shows that lifespan and heat stress resistance are separable, and (3) it strengthens the emerging notion that the ability to respond to heat comes at the expense of protein homeostasis (proteostasis).


Asunto(s)
Regulación de la Expresión Génica/fisiología , Respuesta al Choque Térmico/fisiología , Calor , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sensación Térmica/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Animales Modificados Genéticamente , Infecciones Bacterianas/prevención & control , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Huevos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/genética , Músculos/metabolismo , Mutación/genética , Parálisis/genética , Fragmentos de Péptidos/metabolismo , Lectinas de Plantas/genética , Lectinas de Plantas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Conducta Sexual Animal , Transducción de Señal/genética , Transducción de Señal/fisiología , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Sensación Térmica/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Mol Microbiol ; 84(1): 93-104, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22384939

RESUMEN

Autoinducer-2 (AI-2) a signal produced by a range of phylogenetically distant microorganisms, enables inter-species cell-cell communication and regulates many bacterial phenotypes. Certain bacteria can interfere with AI-2-regulated behaviours of neighbouring species by internalizing AI-2 using the Lsr transport system (encoded by the lsr operon). AI-2 imported by the Lsr is phosphorylated by the LsrK kinase and AI-2-phosphate is the inducer of the lsr operon. Here we show that in Escherichia coli the phosphoenolpyruvate phosphotransferase system (PTS) is required for Lsr activation and is essential for AI-2 internalization. Although the phosphorylation state of Enzyme I of PTS is important for this regulation, LsrK is necessary for the phosphorylation of AI-2, indicating that AI-2 is not phosphorylated by PTS. Our results suggest that AI-2 internalization is initiated by a PTS-dependent mechanism, which provides sufficient intracellular AI-2 to relieve repression of the lsr operon and, thus induce depletion of AI-2 from the extracellular environment. The fact that AI-2 internalization is not only controlled by the community-dependent accumulation of AI-2, but also depends on the phosphorylation state of PTS suggests that E. coli can integrate information on the availability of substrates with external communal information to control quorum sensing and its interference.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Homoserina/análogos & derivados , Lactonas/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Percepción de Quorum , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Homoserina/metabolismo , Mutación , Operón , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Activación Transcripcional
5.
J Cell Sci ; 124(Pt 11): 1891-902, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558416

RESUMEN

Despite the activity of cellular quality-control mechanisms, subsets of mature and newly synthesized polypeptides fail to fold properly and form insoluble aggregates. In some cases, protein aggregation leads to the development of human neurodegenerative maladies, including Alzheimer's and prion diseases. Aggregates of misfolded prion protein (PrP), which appear in cells after exposure to the drug cyclosporin A (CsA), and disease-linked PrP mutants have been found to accumulate in juxtanuclear deposition sites termed 'aggresomes'. Recently, it was shown that cells can contain at least two types of deposition sites for misfolded proteins: a dynamic quality-control compartment, which was termed 'JUNQ', and a site for terminally aggregated proteins called 'IPOD'. Here, we show that CsA-induced PrP aggresomes are dynamic structures that form despite intact proteasome activity, recruit chaperones and dynamically exchange PrP molecules with the cytosol. These findings define the CsA-PrP aggresome as a JUNQ-like dynamic quality-control compartment that mediates the refolding or degradation of misfolded proteins. Together, our data suggest that the formation of PrP aggresomes protects cells from proteotoxic stress.


Asunto(s)
Ciclosporina/farmacología , Cuerpos de Inclusión/metabolismo , Priones/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Cristalinas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Complejos Multiproteicos/metabolismo , Enfermedades por Prión/metabolismo , Priones/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Ubiquitina/metabolismo , Proteínas Ubiquitinadas/metabolismo
6.
Oncogene ; 39(8): 1830, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31576012

RESUMEN

The original version of this Article contained an error in the author affiliations. Vladislav V. Verkhusha was incorrectly associated with the School of Mathematics, Statistics & Applied Mathematics, National University of Ireland Galway, Galway, Ireland. The correct affiliation is Anatomy and Structural Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA.

7.
BMC Med Genomics ; 13(1): 179, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33256706

RESUMEN

BACKGROUND: The Cancer Genome Atlas identified four molecular subgroups of endometrial cancer with survival differences based on whole genome, transcriptomic, and proteomic characterization. Clinically accessible algorithms that reproduce this data are needed. Our aim was to determine if targeted sequencing alone allowed for molecular classification of endometrial cancer. METHODS: Using a custom-designed 156 gene panel, we analyzed 47 endometrial cancers and matching non-tumor tissue. Variants were annotated for pathogenicity and medical records were reviewed for the clinicopathologic variables. Using molecular characteristics, tumors were classified into four subgroups. Group 1 included patients with > 570 unfiltered somatic variants, > 9 cytosine to adenine nucleotide substitutions per sample, and < 1 cytosine to guanine nucleotide substitution per sample. Group 2 included patients with any somatic mutation in MSH2, MSH6, MLH1, PMS2. Group 3 included patients with TP53 mutations without mutation in mismatch repair genes. Remaining patients were classified as group 4. Analyses were performed using SAS 9.4 (SAS Institute Inc., Cary, North Carolina, USA). RESULTS: Endometrioid endometrial cancers had more candidate variants of potential pathogenic interest (median 6 IQR 4.13 vs. 2 IQR 2.3; p < 0.01) than uterine serous cancers. PTEN (82% vs. 15%, p < 0.01) and PIK3CA (74% vs. 23%, p < 0.01) mutations were more frequent in endometrioid than serous carcinomas. TP53 (18% vs. 77%, p < 0.01) mutations were more frequent in serous carcinomas. Visual inspection of the number of unfiltered somatic variants per sample identified six grade 3 endometrioid samples with high tumor mutational burden, all of which demonstrated POLE mutations, most commonly P286R and V411L. Of the grade 3 endometrioid carcinomas, those with POLE mutations were less likely to have risk factors necessitating adjuvant treatment than those with low tumor mutational burden. Targeted sequencing was unable to assign samples to microsatellite unstable, copy number low, and copy number high subgroups. CONCLUSIONS: Targeted sequencing can predict the presence of POLE mutations based on the tumor mutational burden. However, targeted sequencing alone is inadequate to classify endometrial cancers into molecular subgroups identified by The Cancer Genome Atlas.


Asunto(s)
ADN de Neoplasias/genética , Neoplasias Endometriales/clasificación , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Proteínas de Neoplasias/genética , Anciano , Carcinoma Endometrioide/genética , Variaciones en el Número de Copia de ADN , Reparación de la Incompatibilidad de ADN/genética , ADN Polimerasa II/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/mortalidad , Neoplasias Endometriales/terapia , Femenino , Humanos , Mutación INDEL , Persona de Mediana Edad , Anotación de Secuencia Molecular , Neoplasias Primarias Secundarias/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Polimorfismo de Nucleótido Simple
8.
Curr Opin Microbiol ; 10(2): 189-98, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17387037

RESUMEN

Quorum sensing is a form of cell-cell signaling in bacteria that provides information regarding population density, species composition, and environmental and metabolic signals. It enables community-wide coordination of gene expression, and presumably benefits group behaviors. Multiple regulatory small RNAs (sRNAs) act centrally in quorum sensing, integrating signals with other environmental stimuli, to produce an appropriate output.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Percepción de Quorum/genética , ARN Bacteriano/metabolismo , ARN no Traducido/metabolismo , Bacterias/genética , Regulación Bacteriana de la Expresión Génica , Transducción de Señal
9.
Oncogene ; 38(30): 5839-5859, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31285548

RESUMEN

The cytoskeletal interacting protein Septin 9 (SEPT9), a member of the septin gene family, has been proposed to have oncogenic functions. It is a known hot spot of retroviral tagging insertion and a fusion partner of both de novo and therapy-induced mixed lineage leukemia (MLL). Of all septins, SEPT9 holds the strongest link to cancer, especially breast cancer. Murine models of breast cancer frequently exhibit SEPT9 amplification in the form of double minute chromosomes, and about 20% of human breast cancer display genomic amplification and protein over expression at the SEPT9 locus. Yet, a clear mechanism by which SEPT9 elicits tumor-promoting functions is lacking. To obtain unbiased insights on molecular signatures of SEPT9 upregulation in breast tumors, we overexpressed several of its isoforms in breast cancer cell lines. Global transcriptomic profiling supports a role of SEPT9 in invasion. Functional studies reveal that SEPT9 upregulation is sufficient to increase degradation of the extracellular matrix, while SEPT9 downregulation inhibits this process. The degradation pattern is peripheral and associated with focal adhesions (FAs), where it is coupled with increased expression of matrix metalloproteinases (MMPs). SEPT9 overexpression induces MMP upregulation in human tumors and in culture models and promotes MMP3 secretion to the media at FAs. Downregulation of SEPT9 or chemical inhibition of septin filament assembly impairs recruitment of MMP3 to FAs. Our results indicate that SEPT9 promotes upregulation and both trafficking and secretion of MMPs near FAs, thus enhancing migration and invasion of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis , Movimiento Celular , Matriz Extracelular/metabolismo , Adhesiones Focales , Glándulas Mamarias Humanas/patología , Metaloproteinasas de la Matriz/metabolismo , Isoformas de Proteínas/fisiología , Septinas/fisiología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Matriz Extracelular/enzimología , Humanos , Células MCF-7 , Glándulas Mamarias Humanas/metabolismo , Invasividad Neoplásica , Septinas/genética , Microambiente Tumoral , Regulación hacia Arriba
10.
Elife ; 72018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30403374

RESUMEN

Although aging-regulating pathways were discovered a few decades ago, it is not entirely clear how their activities are orchestrated, to govern lifespan and proteostasis at the organismal level. Here, we utilized the nematode Caenorhabditis elegans to examine whether the alteration of aging, by reducing the activity of the Insulin/IGF signaling (IIS) cascade, affects protein SUMOylation. We found that IIS activity promotes the SUMOylation of the germline protein, CAR-1, thereby shortening lifespan and impairing proteostasis. In contrast, the expression of mutated CAR-1, that cannot be SUMOylated at residue 185, extends lifespan and enhances proteostasis. A mechanistic analysis indicated that CAR-1 mediates its aging-altering functions, at least partially, through the notch-like receptor glp-1. Our findings unveil a novel regulatory axis in which SUMOylation is utilized to integrate the aging-controlling functions of the IIS and of the germline and provide new insights into the roles of SUMOylation in the regulation of organismal aging.


Asunto(s)
Envejecimiento/metabolismo , Caenorhabditis elegans/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/metabolismo , Proteostasis , Transducción de Señal , Sumoilación , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Técnicas de Silenciamiento del Gen , Células Germinativas/metabolismo , Gónadas/metabolismo , Longevidad , Modelos Biológicos , Estrés Fisiológico , Transcripción Genética
11.
Oncotarget ; 8(60): 102033-102045, 2017 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-29254223

RESUMEN

Next generation sequencing (NGS) technologies have revolutionized our approach to genomic research. The use of whole genome sequencing (WGS), whole exome sequencing (WES), transcriptome profiling, and targeted DNA sequencing has exponentially improved our understanding of the human genome and the genetic complexities underlying malignancy. Yet, WGS and WES clinical applications remain limited due to high costs and the large volume of data generated. When utilized to address biological questions in basic science studies, targeted sequencing panels have proven extremely valuable due to reduced costs and higher sequencing depth. However, the routine application of targeted sequencing to the clinical setting is limited to a few cancer subtypes. Some highly aggressive tumor types, like type 2 endometrial cancer (EC), could greatly benefit from routine genomic analysis using targeted sequencing. To explore the potential utility of a mid size panel (~150 genes) in the clinical setting, we developed and validated a custom panel against WGS, WES, and another commercially available targeted panel. Our results indicate that a mid size custom designed panel is as efficient as WGS and WES in mapping variants of biological and clinical relevance, rendering higher coverage, at a lower cost, with fewer variants of uncertain significance. Because of the much higher sequencing depth that could be achieved, our results demonstrate that targeted sequencing outperformed WGS and WES in the mapping of pathogenic variants in a breast cancer case, as well as a case of mixed serous and high-grade endometrioid EC, the most aggressive EC subtype.

12.
Cell Rep ; 9(6): 2192-205, 2014 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-25497098

RESUMEN

In the nematode Caenorhabditis elegans, insulin/insulin-like growth factor 1 (IGF-1) signaling (IIS) reduction hyperactivates the transcription factors DAF-16 and heat shock factor 1 (HSF-1), creating long-lived, stress-resistant worms that are protected from proteotoxicity. How DAF-16 executes its distinct functions in response to IIS reduction is largely obscure. Here, we report that NHL-1, a member of the TRIM-NHL protein family, acts in chemosensory neurons to promote stress resistance in distal tissues by DAF-16 activation but is dispensable for the activation of HSF-1. The expression of nhl-1 is regulated by the IIS, defining a neuronal regulatory circuit that controls the organismal stress response. The knockdown of nhl-1 protects nematodes that express the Alzheimer-disease-associated Aß peptide from proteotoxicity but has no effect on lifespan. Our findings indicate that DAF-16- and HSF-1-regulated heat-responsive mechanisms are differentially controlled by neurons and show that one neuronal protein can be involved in the activation of different stress responses in remote tissues.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Células Quimiorreceptoras/metabolismo , Factores de Transcripción Forkhead/genética , Proteínas del Tejido Nervioso/genética , Transducción de Señal , Somatomedinas/metabolismo , Estrés Fisiológico , Factores de Transcripción/genética
13.
Aging Cell ; 11(3): 491-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22360389

RESUMEN

Reducing the activity of the insulin/IGF-1 signaling pathway (IIS) modifies development, elevates stress resistance, protects from toxic protein aggregation (proteotoxicity), and extends lifespan (LS) of worms, flies, and mice. In the nematode Caenorhabditis elegans, LS extension by IIS reduction is entirely dependent upon the activity of the transcription factors DAF-16 and the heat shock factor-1 (HSF-1). While DAF-16 determines LS exclusively during early adulthood, it is required for proteotoxicity protection also during late adulthood. In contrast, HSF-1 protects from proteotoxicity during larval development. Despite the critical requirement for HSF-1 for LS extension, the temporal requirements for this transcription factor as a LS determinant are unknown. To establish the temporal requirements of HSF-1 for longevity assurance, we conditionally knocked down hsf-1 during larval development and adulthood of C. elegans and found that unlike daf-16, hsf-1 is foremost required for LS determination during early larval development, required for a lesser extent during early adulthood and has small effect on longevity also during late adulthood. Our findings indicate that early developmental events affect LS and suggest that HSF-1 sets during development of the conditions that enable DAF-16 to promote longevity during reproductive adulthood. This study proposes a novel link between HSF-1 and the longevity functions of the IIS.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Longevidad/fisiología , Factores de Transcripción/fisiología , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/biosíntesis , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Senescencia Celular/fisiología , Factores de Transcripción Forkhead , Técnicas de Silenciamiento del Gen , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Cell ; 125(4): 679-90, 2006 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-16713562

RESUMEN

In response to DNA damage, cells activate checkpoint signaling cascades to control cell-cycle progression and elicit DNA repair in order to maintain genomic integrity. The sensing and repair of lesions is critical for Bacillus subtilis cells entering the developmental process of sporulation as damaged DNA may prevent the cells from completing spore morphogenesis. We report the identification of the protein DisA (DNA integrity scanning protein, annotated YacK), which is required to delay the initiation of sporulation in response to chromosomal damage. DisA is a nonspecific DNA binding protein that forms a single focus, which moves rapidly within the bacterial cell, pausing at sites of DNA damage. We propose that the DisA focus scans along the chromosomes searching for lesions. Upon encountering a lesion, DisA delays entry into sporulation until the damage is repaired.


Asunto(s)
Bacillus subtilis/genética , Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Cromosomas Bacterianos/metabolismo , Daño del ADN , Genes cdc , Esporas Fúngicas , Bacillus subtilis/citología , Proteínas Bacterianas/genética , Ciclo Celular/fisiología , Cromosomas Bacterianos/genética , Proteínas de Escherichia coli , Hibridación Fluorescente in Situ , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda