Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38464067

RESUMEN

Chemical proteomics enables the global assessment of small molecule-protein interactions in native biological systems and has emerged as a versatile approach for ligand discovery. The range of small molecules explored by chemical proteomics has, however, been limited. Here, we describe a diversity-oriented synthesis (DOS)-inspired library of stereochemically-defined compounds bearing diazirine and alkyne units for UV light-induced covalent modification and click chemistry enrichment of interacting proteins, respectively. We find that these 'photo-stereoprobes' interact in a stereoselective manner with hundreds of proteins from various structural and functional classes in human cells and demonstrate that these interactions can form the basis for high-throughput screening-compatible nanoBRET assays. Integrated phenotypic analysis and chemical proteomics identified photo-stereoprobes that modulate autophagy by engaging the mitochondrial serine protease CLPP. Our findings show the utility of photo-stereoprobes for expanding the ligandable proteome, furnishing target engagement assays, and discovering and characterizing bioactive small molecules by cell-based screening.

2.
Cell Rep ; 43(3): 113944, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38489265

RESUMEN

Population genetics continues to identify genetic variants associated with diseases of the immune system and offers a unique opportunity to discover mechanisms of immune regulation. Multiple genetic variants linked to severe fungal infections and autoimmunity are associated with caspase recruitment domain-containing protein 9 (CARD9). We leverage the CARD9 R101C missense variant to uncover a biochemical mechanism of CARD9 activation essential for antifungal responses. We demonstrate that R101C disrupts a critical signaling switch whereby phosphorylation of S104 releases CARD9 from an autoinhibited state to promote inflammatory responses in myeloid cells. Furthermore, we show that CARD9 R101C exerts dynamic effects on the skin cellular contexture during fungal infection, corrupting inflammatory signaling and cell-cell communication circuits. Card9 R101C mice fail to control dermatophyte infection in the skin, resulting in high fungal burden, yet show minimal signs of inflammation. Together, we demonstrate how translational genetics reveals molecular and cellular mechanisms of innate immune regulation.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Micosis , Animales , Ratones , Fosforilación , Proteínas Adaptadoras de Señalización CARD/metabolismo , Transducción de Señal , Inflamación , Antifúngicos
3.
Cell Rep ; 42(7): 112708, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37392388

RESUMEN

Autophagy is an essential cellular process that is deeply integrated with innate immune signaling; however, studies that examine the impact of autophagic modulation in the context of inflammatory conditions are lacking. Here, using mice with a constitutively active variant of the autophagy gene Beclin1, we show that increased autophagy dampens cytokine production during a model of macrophage activation syndrome and in adherent-invasive Escherichia coli (AIEC) infection. Moreover, loss of functional autophagy through conditional deletion of Beclin1 in myeloid cells significantly enhances innate immunity in these contexts. We further analyzed primary macrophages from these animals with a combination of transcriptomics and proteomics to identify mechanistic targets downstream of autophagy. Our study reveals glutamine/glutathione metabolism and the RNF128/TBK1 axis as independent regulators of inflammation. Altogether, our work highlights increased autophagic flux as a potential approach to reduce inflammation and defines independent mechanistic cascades involved in this control.


Asunto(s)
Enfermedad de Crohn , Infecciones por Escherichia coli , Animales , Ratones , Enfermedad de Crohn/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Autofagia/genética , Macrófagos/metabolismo , Inflamación/metabolismo , Citocinas/metabolismo
4.
Nat Immunol ; 23(7): 1063-1075, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35668320

RESUMEN

Extracellular acidification occurs in inflamed tissue and the tumor microenvironment; however, a systematic study on how pH sensing contributes to tissue homeostasis is lacking. In the present study, we examine cell type-specific roles of the pH sensor G protein-coupled receptor 65 (GPR65) and its inflammatory disease-associated Ile231Leu-coding variant in inflammation control. GPR65 Ile231Leu knock-in mice are highly susceptible to both bacterial infection-induced and T cell-driven colitis. Mechanistically, GPR65 Ile231Leu elicits a cytokine imbalance through impaired helper type 17 T cell (TH17 cell) and TH22 cell differentiation and interleukin (IL)-22 production in association with altered cellular metabolism controlled through the cAMP-CREB-DGAT1 axis. In dendritic cells, GPR65 Ile231Leu elevates IL-12 and IL-23 release at acidic pH and alters endo-lysosomal fusion and degradation capacity, resulting in enhanced antigen presentation. The present study highlights GPR65 Ile231Leu as a multistep risk factor in intestinal inflammation and illuminates a mechanism by which pH sensing controls inflammatory circuits and tissue homeostasis.


Asunto(s)
Colitis , Receptores Acoplados a Proteínas G , Animales , Colitis/metabolismo , Concentración de Iones de Hidrógeno , Inflamación/metabolismo , Lisosomas/metabolismo , Ratones , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Th17/metabolismo
5.
Autophagy ; 18(2): 452-454, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34812102

RESUMEN

Human genetics and loss-of-function studies revealed a critical role for macroautophagy/autophagy in host defense. The autophagic delivery of intracellular pathogens to lysosomes is a central mechanism of innate immunity; thus, augmentation of host xenophagy represents a promising and powerful approach to combat infections. The precise mechanisms required for autophagosome biogenesis and maturation, however, remain unclear. Using a targeted genetic screen against phosphoinositide kinases and phosphatases, our recent work identified an essential role for the phosphoinositide phosphatase SACM1L/SAC1 in xenophagy. Re-expression of wild-type or catalytically-dead SACM1L in CRISPR knockout cells confirmed that SACM1L enzymatic activity is required to suppress replication of intracellular Salmonella. Time-dependent, quantitative and live confocal imaging demonstrated that SACM1L-deficient cells accumulate phosphatidylinositol-4-phosphate (PtdIns4P) on bacteria-containing autophagosomes, resulting in delayed fusion with degradative lysosomes and reduced bacterial killing. We further discovered that the secreted Salmonella effector protein SteA, which specifically binds PtdIns4P, exacerbates the SACM1L-dependent delay in autophagosomal maturation. These findings reveal a relationship in which the balance between host defense and bacterial survival depends upon autophagosomal membrane composition.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa , Monoéster Fosfórico Hidrolasas , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Autofagosomas/metabolismo , Autofagia/genética , Bacterias/metabolismo , Mecanismos de Defensa , Humanos , Lisosomas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Salmonella/metabolismo
6.
Cell Rep ; 36(4): 109434, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320354

RESUMEN

Phosphoinositides are important molecules in lipid signaling, membrane identity, and trafficking that are spatiotemporally controlled by factors from both mammalian cells and intracellular pathogens. Here, using small interfering RNA (siRNA) directed against phosphoinositide kinases and phosphatases, we screen for regulators of the host innate defense response to intracellular bacterial replication. We identify SAC1, a transmembrane phosphoinositide phosphatase, as an essential regulator of xenophagy. Depletion or inactivation of SAC1 compromises fusion between Salmonella-containing autophagosomes and lysosomes, leading to increased bacterial replication. Mechanistically, the loss of SAC1 results in aberrant accumulation of phosphatidylinositol-4-phosphate [PI(4)P] on Salmonella-containing autophagosomes, thus facilitating recruitment of SteA, a PI(4)P-binding Salmonella effector protein, which impedes lysosomal fusion. Replication of Salmonella lacking SteA is suppressed by SAC-1-deficient cells, however, demonstrating bacterial adaptation to xenophagy. Our findings uncover a paradigm in which a host protein regulates the level of its substrate and impairs the function of a bacterial effector during xenophagy.


Asunto(s)
Autofagosomas , Macroautofagia , Fosfatos de Fosfatidilinositol , Fosfoinosítido Fosfatasas , Salmonella , Humanos , Autofagosomas/metabolismo , Proteínas Bacterianas/metabolismo , Citosol/microbiología , Células HEK293 , Células HeLa , Lípidos/química , Lisosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoinosítido Fosfatasas/metabolismo , Salmonella/crecimiento & desarrollo , Salmonella/metabolismo
7.
Cell Rep ; 33(6): 108371, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33176151

RESUMEN

Transcription factor EB (TFEB) activates lysosomal biogenesis genes in response to environmental cues. Given implications of impaired TFEB signaling and lysosomal dysfunction in metabolic, neurological, and infectious diseases, we aim to systematically identify TFEB-directed circuits by examining transcriptional responses to TFEB subcellular localization and stimulation. We reveal that steady-state nuclear TFEB is sufficient to activate transcription of lysosomal, autophagy, and innate immunity genes, whereas other targets require higher thresholds of stimulation. Furthermore, we identify shared and distinct transcriptional signatures between mTOR inhibition and bacterial autophagy. Using a genome-wide CRISPR library, we find TFEB targets that protect cells from or sensitize cells to lysosomal cell death. BHLHE40 and BHLHE41, genes responsive to high, sustained levels of nuclear TFEB, act in opposition to TFEB upon lysosomal cell death induction. Further investigation identifies genes counter-regulated by TFEB and BHLHE40/41, adding this negative feedback to the current understanding of TFEB regulatory mechanisms.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Técnicas de Inactivación de Genes , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Lisosomas/metabolismo , Mitocondrias/metabolismo , Transcripción Genética
8.
ACS Infect Dis ; 4(10): 1499-1507, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30058798

RESUMEN

Toxoplasma gondii is an obligate intracellular parasite capable of causing severe disease due to congenital infection and in patients with compromised immune systems. Control of infection is dependent on a robust Th1 type immune response including production of interferon gamma (IFN-γ), which is essential for control. IFN-γ activates a variety of antimicrobial mechanisms in host cells, which are then able to control intracellular parasites such as T. gondii. Despite the effectiveness of these pathways in controlling acute infection, the immune system is unable to eradicate chronic infections that can persist for life. Similarly, while antibiotic treatment can control acute infection, it is unable to eliminate chronic infection. To identify compounds that would act synergistically with IFN-γ, we performed a high-throughput screen of diverse small molecule libraries to identify inhibitors of T. gondii. We identified a number of compounds that inhibited parasite growth in vitro at low µM concentrations and that demonstrated enhanced potency in the presence of a low level of IFN-γ. A subset of these compounds act by enhancing the recruitment of light chain 3 (LC3) to the parasite-containing vacuole, suggesting they work by an autophagy-related process, while others were independent of this pathway. The pattern of IFN-γ dependence was shared among the majority of analogs from 6 priority scaffolds, and analysis of structure activity relationships for one such class revealed specific stereochemistry associated with this feature. Identification of these IFN-γ-dependent leads may lead to development of improved therapeutics due to their synergistic interactions with immune responses.


Asunto(s)
Inhibidores de Crecimiento/análisis , Inhibidores de Crecimiento/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Interferón gamma/metabolismo , Toxoplasma/crecimiento & desarrollo , Autofagia/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Inhibidores de Crecimiento/química , Células HeLa , Humanos , Inmunidad Innata , Modelos Lineales , Luciferasas/análisis , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Células TH1/inmunología , Vacuolas/metabolismo
9.
Biochemistry ; 56(51): 6639-6651, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29185708

RESUMEN

Beclin-1 (BECN1) is an essential component of macroautophagy. This process is a highly conserved survival mechanism that recycles damaged cellular components or pathogens by encasing them in a bilayer vesicle that fuses with a lysosome to allow degradation of the vesicular contents. Mutations or altered expression profiles of BECN1 have been linked to various cancers and neurodegenerative diseases. Viruses, including HIV and herpes simplex virus 1 (HSV-1), are also known to specifically target BECN1 as a means of evading host defense mechanisms. Autophagy is regulated by the interaction between BECN1 and Bcl-2, a pro-survival protein in the apoptotic pathway that stabilizes the BECN1 homodimer. Disruption of the homodimer by phosphorylation or competitive binding promotes autophagy through an unknown mechanism. We report here the first recombinant synthesis (3-5 mg/L in an Escherichia coli culture) and characterization of full-length, human BECN1. Our analysis reveals that full-length BECN1 exists as a soluble homodimer (KD ∼ 0.45 µM) that interacts with Bcl-2 (KD = 4.3 ± 1.2 µM) and binds to lipid membranes. Dimerization is proposed to be mediated by a coiled-coil region of BECN1. A construct lacking the C-terminal BARA domain but including the coiled-coil region exhibits a homodimer KD 3.5-fold weaker than that of full-length BECN1, indicating that both the BARA domain and the coiled-coil region of BECN1 contribute to dimer formation. Using site-directed mutagenesis, we show that residues at the C-terminus of the coiled-coil region previously shown to interact with the BARA domain play a key role in dimerization and mutations weaken the interface by ∼5-fold.


Asunto(s)
Autofagia , Beclina-1/química , Multimerización de Proteína , Secuencia de Aminoácidos , Beclina-1/biosíntesis , Beclina-1/genética , Escherichia coli , Humanos , Mutagénesis Sitio-Dirigida , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
10.
MAbs ; 6(4): 957-67, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24758837

RESUMEN

Antibody combination therapeutics (ACTs) are polyvalent biopharmaceuticals that are uniquely suited for the control of complex diseases, including antibiotic resistant infectious diseases, autoimmune disorders and cancers. However, ACTs also represent a distinct manufacturing challenge because the independent manufacture and subsequent mixing of monoclonal antibodies quickly becomes cost prohibitive as more complex mixtures are envisioned. We have developed a virus-free recombinant protein expression platform based on adeno-associated viral (AAV) elements that is capable of rapid and consistent production of complex antibody mixtures in a single batch format. Using both multiplexed immunoassays and cation exchange (CIEX) chromatography, cell culture supernatants generated using our system were assessed for stability of expression and ratios of the component antibodies over time. Cultures expressing combinations of three to ten antibodies maintained consistent expression levels and stable ratios of component antibodies for at least 60 days. Cultures showed remarkable reproducibility following cell banking, and AAV-based cultures showed higher stability and productivity than non-AAV based cultures. Therefore, this non-viral AAV-based expression platform represents a predictable, reproducible, quick and cost effective method to manufacture or quickly produce for preclinical testing recombinant antibody combination therapies and other recombinant protein mixtures.


Asunto(s)
Dependovirus , Expresión Génica , Anticuerpos de Cadena Única/biosíntesis , Línea Celular , Cromatografía por Intercambio Iónico/métodos , Técnicas de Cocultivo , Quimioterapia Combinada , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/aislamiento & purificación
11.
PLoS Pathog ; 7(5): e1002056, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21637816

RESUMEN

Coxiella burnetii, the causative agent of human Q fever, is an intracellular pathogen that replicates in an acidified vacuole derived from the host lysosomal network. This pathogen encodes a Dot/Icm type IV secretion system that delivers bacterial proteins called effectors to the host cytosol. To identify new effector proteins, the functionally analogous Legionella pneumophila Dot/Icm system was used in a genetic screen to identify fragments of C. burnetii genomic DNA that when fused to an adenylate cyclase reporter were capable of directing Dot/Icm-dependent translocation of the fusion protein into mammalian host cells. This screen identified Dot/Icm effectors that were proteins unique to C. burnetii, having no overall sequence homology with L. pneumophila Dot/Icm effectors. A comparison of C. burnetii genome sequences from different isolates revealed diversity in the size and distribution of the genes encoding many of these effectors. Studies examining the localization and function of effectors in eukaryotic cells provided evidence that several of these proteins have an affinity for specific host organelles and can disrupt cellular functions. The identification of a transposon insertion mutation that disrupts the dot/icm locus was used to validate that this apparatus was essential for translocation of effectors. Importantly, this C. burnetii Dot/Icm-deficient mutant was found to be defective for intracellular replication. Thus, these data indicate that C. burnetii encodes a unique subset of bacterial effector proteins translocated into host cells by the Dot/Icm apparatus, and that the cumulative activities exerted by these effectors enables C. burnetii to successfully establish a niche inside mammalian cells that supports intracellular replication.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Viabilidad Microbiana , Animales , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , ADN Bacteriano/genética , Femenino , Pruebas Genéticas , Células HEK293 , Células HeLa , Humanos , Riñón/citología , Riñón/microbiología , Ovario/citología , Ovario/microbiología , Fenotipo , Neoplasias del Cuello Uterino/microbiología , Neoplasias del Cuello Uterino/patología , Células Vero
12.
Proc Natl Acad Sci U S A ; 107(44): 18997-9001, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20944063

RESUMEN

Coxiella burnetii and Legionella pneumophila are evolutionarily related pathogens with different intracellular infection strategies. C. burnetii persists within and is transmitted by mammalian hosts, whereas, L. pneumophila is found primarily in the environment associated with protozoan hosts. Although a type IV secretion system encoded by the defect in organelle trafficking (dot) and intracellular multiplication (icm) genes is a virulence determinant that remains highly conserved in both bacteria, the two pathogens encode a different array of effector proteins that are delivered into host cells by the Dot/Icm machinery. This difference suggests that adaptations to evolutionarily distinct hosts may be reflected in the effector protein repertoires displayed by these two pathogens. Here we provide evidence in support of this hypothesis. We show that a unique C. burnetii effector from the ankyrin repeat (Ank) family called AnkG interferes with the mammalian apoptosis pathway. AnkG was found to interact with the host protein gC1qR (p32). Either the addition of AnkG to the repertoire of L. pneumophila effector proteins or the silencing of p32 in mouse dendritic cells resulted in a gain of function that allowed intracellular replication of L. pneumophila in these normally restrictive mammalian host cells by preventing rapid pathogen-induced apoptosis. These data indicate that p32 regulates pathogen-induced apoptosis and that AnkG functions to block this pathway. Thus, emergence of an effector protein that interferes with a proapoptotic signaling pathway directed against intracellular bacteria correlates with adaptation of a pathogen to mammalian hosts.


Asunto(s)
Apoptosis/inmunología , Proteínas Bacterianas/inmunología , Coxiella burnetii/fisiología , Interacciones Huésped-Patógeno/inmunología , Receptores de Hialuranos/inmunología , Legionella pneumophila/fisiología , Fiebre Q/inmunología , Secuencias de Aminoácidos , Animales , Apoptosis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coxiella burnetii/patogenicidad , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células HEK293 , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Legionella pneumophila/patogenicidad , Ratones , Proteínas Mitocondriales , Fiebre Q/genética , Fiebre Q/metabolismo
13.
PLoS Pathog ; 6(1): e1000720, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20084115

RESUMEN

Toxoplasma gondii is an obligate intracellular parasite that enters cells by a process of active penetration. Host cell penetration and parasite motility are driven by a myosin motor complex consisting of four known proteins: TgMyoA, an unconventional Class XIV myosin; TgMLC1, a myosin light chain; and two membrane-associated proteins, TgGAP45 and TgGAP50. Little is known about how the activity of the myosin motor complex is regulated. Here, we show that treatment of parasites with a recently identified small-molecule inhibitor of invasion and motility results in a rapid and irreversible change in the electrophoretic mobility of TgMLC1. While the precise nature of the TgMLC1 modification has not yet been established, it was mapped to the peptide Val46-Arg59. To determine if the TgMLC1 modification is responsible for the motility defect observed in parasites after compound treatment, the activity of myosin motor complexes from control and compound-treated parasites was compared in an in vitro motility assay. TgMyoA motor complexes containing the modified TgMLC1 showed significantly decreased motor activity compared to control complexes. This change in motor activity likely accounts for the motility defects seen in the parasites after compound treatment and provides the first evidence, in any species, that the mechanical activity of Class XIV myosins can be modulated by posttranslational modifications to their associated light chains.


Asunto(s)
Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Procesamiento Proteico-Postraduccional , Toxoplasma/fisiología , Secuencia de Aminoácidos , Autorradiografía , Western Blotting , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Espectrometría de Masas en Tándem
14.
Eukaryot Cell ; 3(5): 1320-30, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15470260

RESUMEN

Many intracellular pathogens are separated from the cytosol of their host cells by a vacuole membrane. This membrane serves as a critical interface between the pathogen and the host cell, across which nutrients are imported, wastes are excreted, and communication between the two cells takes place. Very little is known about the vacuole membrane proteins mediating these processes in any host-pathogen interaction. During a screen for monoclonal antibodies against novel surface or secreted proteins of Toxoplasma gondii, we identified ROP4, a previously uncharacterized member of the ROP2 family of proteins. We report here on the sequence, posttranslational processing, and subcellular localization of ROP4, a type I transmembrane protein. Mature, processed ROP4 is localized to the rhoptries, secretory organelles at the apical end of the parasite, and is secreted from the parasite during host cell invasion. Released ROP4 associates with the vacuole membrane and becomes phosphorylated in the infected cell. Similar results are seen with ROP2. Further analysis of ROP4 showed it to be phosphorylated on multiple sites, a subset of which result from the action of either host cell protein kinase(s) or parasite kinase(s) activated by host cell factors. The localization and posttranslational modification of ROP4 and other members of the ROP2 family of proteins within the infected cell make them well situated to play important roles in vacuole membrane function.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Antígenos de Protozoos/genética , Secuencia de Bases , ADN Protozoario/genética , Genes Protozoarios , Interacciones Huésped-Parásitos/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Toxoplasma/genética , Toxoplasma/patogenicidad , Toxoplasma/ultraestructura , Vacuolas/metabolismo
15.
Proc Natl Acad Sci U S A ; 101(19): 7433-8, 2004 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-15123807

RESUMEN

Toxoplasma gondii is the most common protozoan parasite of humans. Infection with T. gondii can lead to life-threatening disease as a result of repeated cycles of host cell invasion, parasite replication, and host cell lysis. Relatively little is known about the invasive mechanisms of T. gondii and related parasites within the Phylum Apicomplexa (including Plasmodium spp., the causative agents of malaria), due to difficulties associated with studying genes essential to invasion in haploid obligate intracellular organisms. To circumvent this problem, we have developed a high-throughput microscope-based assay, which we have used to screen a collection of 12,160 structurally diverse small molecules for inhibitors of T. gondii invasion. A total of 24 noncytotoxic invasion inhibitors were identified. Secondary assays demonstrated that different inhibitors perturb different aspects of invasion, including gliding motility, secretion of host cell adhesins from apical organelles (the micronemes), and extension of a unique tubulin-based structure at the anterior of the parasite (the conoid). Unexpectedly, the screen also identified six small molecules that dramatically enhance invasion, gliding motility, and microneme secretion. The small molecules identified here reveal a previously unrecognized complexity in the control of parasite motility and microneme secretion, and they constitute a set of useful probes for dissecting the invasive mechanisms of T. gondii and related parasites. Small-molecule-based approaches provide a powerful means to address experimentally challenging problems in host-pathogen interaction, while simultaneously identifying new potential targets for drug development.


Asunto(s)
Toxoplasma/fisiología , Animales , Antiprotozoarios/farmacología , Orgánulos/fisiología , Toxoplasma/efectos de los fármacos
16.
Cell Microbiol ; 4(8): 471-82, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12174082

RESUMEN

High-throughput screening of small molecules is used extensively in pharmaceutical settings for the purpose of drug discovery. In the case of antimicrobials, this involves the identification of small molecules that are significantly more toxic to the microbe than to the host. Only a small percentage of the small molecules identified in these screens have been studied in sufficient detail to explain the molecular basis of their antimicrobial effect. Rarer still are small molecule screens undertaken with the explicit goal of learning more about the biology of a particular microbe or the mechanism of its interaction with its host. Recent technological advances in small molecule synthesis and high-throughput screening have made such mechanism-directed small molecule approaches a powerful and accessible experimental option. In this article, we provide an overview of the methods and technical requirements and we discuss the potential of small molecule approaches to address important and often otherwise experimentally intractable problems in cellular microbiology.


Asunto(s)
Evaluación Preclínica de Medicamentos , Microbiología , Animales , Diseño de Fármacos , Técnicas Microbiológicas , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...