Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 398
Filtrar
1.
BMC Biol ; 22(1): 27, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38317219

RESUMEN

BACKGROUND: Staphylococcus aureus (SA) and Pseudomonas aeruginosa (PA) cause a wide variety of bacterial infections and coinfections, showing a complex interaction that involves the production of different metabolites and metabolic changes. Temperature is a key factor for bacterial survival and virulence and within the host, bacteria could be exposed to an increment in temperature during fever development. We analyzed the previously unexplored effect of fever-like temperatures (39 °C) on S. aureus USA300 and P. aeruginosa PAO1 microaerobic mono- and co-cultures compared with 37 °C, by using RNAseq and physiological assays including in vivo experiments. RESULTS: In general terms both temperature and co-culturing had a strong impact on both PA and SA with the exception of the temperature response of monocultured PA. We studied metabolic and virulence changes in both species. Altered metabolic features at 39 °C included arginine biosynthesis and the periplasmic glucose oxidation in S. aureus and P. aeruginosa monocultures respectively. When PA co-cultures were exposed at 39 °C, they upregulated ethanol oxidation-related genes along with an increment in organic acid accumulation. Regarding virulence factors, monocultured SA showed an increase in the mRNA expression of the agr operon and hld, pmsα, and pmsß genes at 39 °C. Supported by mRNA data, we performed physiological experiments and detected and increment in hemolysis, staphyloxantin production, and a decrease in biofilm formation at 39 °C. On the side of PA monocultures, we observed an increase in extracellular lipase and protease and biofilm formation at 39 °C along with a decrease in the motility in correlation with changes observed at mRNA abundance. Additionally, we assessed host-pathogen interaction both in vitro and in vivo. S. aureus monocultured at 39οC showed a decrease in cellular invasion and an increase in IL-8-but not in IL-6-production by A549 cell line. PA also decreased its cellular invasion when monocultured at 39 °C and did not induce any change in IL-8 or IL-6 production. PA strongly increased cellular invasion when co-cultured at 37 and 39 °C. Finally, we observed increased lethality in mice intranasally inoculated with S. aureus monocultures pre-incubated at 39 °C and even higher levels when inoculated with co-cultures. The bacterial burden for P. aeruginosa was higher in liver when the mice were infected with co-cultures previously incubated at 39 °C comparing with 37 °C. CONCLUSIONS: Our results highlight a relevant change in the virulence of bacterial opportunistic pathogens exposed to fever-like temperatures in presence of competitors, opening new questions related to bacteria-bacteria and host-pathogen interactions and coevolution.


Asunto(s)
Infecciones por Pseudomonas , Infecciones Estafilocócicas , Ratones , Animales , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Virulencia/fisiología , Pseudomonas aeruginosa , Temperatura , Interleucina-6/metabolismo , Interleucina-6/farmacología , Interleucina-8/metabolismo , Interleucina-8/farmacología , Infecciones por Pseudomonas/microbiología , ARN Mensajero/metabolismo , Biopelículas , Infecciones Estafilocócicas/microbiología
2.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35007292

RESUMEN

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Asunto(s)
Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Macrófagos/microbiología , Vacuolas/microbiología , Virulencia/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Biomolecules ; 11(8)2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34439861

RESUMEN

BceF is a bacterial tyrosine kinase (BY-kinase) from Burkholderia cepacia, a Gram-negative bacterium accountable for respiratory infections in immunocompromised and cystic fibrosis patients. BceF is involved in the production of exopolysaccharides secreted to the biofilm matrix and promotes resistant and aggressive infections. BY-kinases share no homology with mammalian kinases, and thereby offer a means to develop novel and specific antivirulence drugs. Here, we report the crystal structure of the BceF kinase domain at 1.85 Å resolution. The isolated BceF kinase domain is assembled as a dimer in solution and crystallized as a dimer in the asymmetric unit with endogenous adenosine-diphosphate bound at the active sites. The low enzymatic efficiency measured in solution may be explained by the partial obstruction of the active sites at the crystallographic dimer interface. This study provides insights into self-assembly and the specific activity of isolated catalytic domains. Several unique variations around the active site compared to other BY-kinases may allow for structure-based design of specific inhibitors to target Burkholderia cepacia virulence.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Burkholderia cepacia/fisiología , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/fisiología , Cristalografía por Rayos X/métodos , Humanos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Virulencia/fisiología
4.
Semin Cancer Biol ; 76: 143-155, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33865991

RESUMEN

Helicobacter pylori is a human bacterial pathogen that causes peptic ulcers and has been designated a Class I carcinogen by the International Agency for Research on Cancer (IARC). Its ability to survive in the acid environment of the stomach, to colonize the stomach mucosa, and to cause cancer, are linked to two enzymes that require nickel-urease and hydrogenase. Thus, nickel is an important virulence factor and the proteins involved in nickel trafficking are potential antibiotic targets. This review summarizes the nickel biochemistry of H. pylori with a focus on the roles of nickel in virulence, nickel homeostasis, maturation of urease and hydrogenase, and the unique nickel trafficking that occurs between the hydrogenase maturation pathway and urease nickel incorporation that is mediated by the metallochaperone HypA and its partner, HypB.


Asunto(s)
Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Níquel/metabolismo , Factores de Virulencia/metabolismo , Virulencia/fisiología , Animales , Carcinógenos/metabolismo , Humanos
5.
PLoS Negl Trop Dis ; 15(4): e0009339, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33857149

RESUMEN

BACKGROUND: Scrub typhus is a neglected tropical disease that threatens more than one billion people. If antibiotic therapy is delayed, often due to mis- or late diagnosis, the case fatality rate can increase considerably. Scrub typhus is caused by the obligate intracellular bacterium, Orientia tsutsugamushi, which invades phagocytes and endothelial cells in vivo and diverse tissue culture cell types in vitro. The ability of O. tsutsugamushi to replicate in the cytoplasm indicates that it has evolved to counter eukaryotic host cell immune defense mechanisms. The transcription factor, NF-κB, is a tightly regulated initiator of proinflammatory and antimicrobial responses. Typically, the inhibitory proteins p105 and IκBα sequester the NF-κB p50:p65 heterodimer in the cytoplasm. Canonical activation of NF-κB via TNFα involves IKKß-mediated serine phosphorylation of IκBα and p105, which leads to their degradation and enables NF-κB nuclear translocation. A portion of p105 is also processed into p50. O. tsutsugamushi impairs NF-κB translocation into the nucleus, but how it does so is incompletely defined. PRINCIPAL FINDINGS: Western blot, densitometry, and quantitative RT-PCR analyses of O. tsutsugamushi infected host cells were used to determine if the pathogen's ability to inhibit NF-κB is linked to modulation of p105. Results demonstrate that p105 levels are elevated several-fold in O. tsutsugamushi infected HeLa and RF/6A cells with only a nominal increase in p50. The O. tsutsugamushi-stimulated increase in p105 is bacterial dose- and protein synthesis-dependent, but does not occur at the level of host cell transcription. While TNFα-induced phosphorylation of p105 serine 932 proceeds unhindered in infected cells, p105 levels remain elevated and NF-κB p65 is retained in the cytoplasm. CONCLUSIONS: O. tsutsugamushi specifically stabilizes p105 to inhibit the canonical NF-κB pathway, which advances understanding of how it counters host immunity to establish infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Orientia tsutsugamushi/metabolismo , Orientia tsutsugamushi/patogenicidad , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Orientia tsutsugamushi/inmunología , Tifus por Ácaros/inmunología , Tifus por Ácaros/microbiología , Activación Transcripcional , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia/genética , Virulencia/inmunología , Virulencia/fisiología
6.
PLoS Pathog ; 17(3): e1009481, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33788895

RESUMEN

TcpC is a virulence factor of uropathogenic E. coli (UPEC). It was found that TIR domain of TcpC impedes TLR signaling by direct association with MyD88. It has been a long-standing question whether bacterial pathogens have evolved a mechanism to manipulate MyD88 degradation by ubiquitin-proteasome pathway. Here, we show that TcpC is a MyD88-targeted E3 ubiquitin ligase. Kidney macrophages from mice with pyelonephritis induced by TcpC-secreting UPEC showed significantly decreased MyD88 protein levels. Recombinant TcpC (rTcpC) dose-dependently inhibited protein but not mRNA levels of MyD88 in macrophages. Moreover, rTcpC significantly promoted MyD88 ubiquitination and accumulation in proteasomes in macrophages. Cys12 and Trp106 in TcpC are crucial amino acids in maintaining its E3 activity. Therefore, TcpC blocks TLR signaling pathway by degradation of MyD88 through ubiquitin-proteasome system. Our findings provide not only a novel biochemical mechanism underlying TcpC-medicated immune evasion, but also the first example that bacterial pathogens inhibit MyD88-mediated signaling pathway by virulence factors that function as E3 ubiquitin ligase.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/fisiología , Escherichia coli Uropatógena/patogenicidad , Factores de Virulencia/metabolismo , Animales , Línea Celular , Femenino , Humanos , Evasión Inmune/fisiología , Macrófagos , Ratones , Ratones Endogámicos C57BL , Pielonefritis/inmunología , Pielonefritis/microbiología , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Escherichia coli Uropatógena/inmunología , Escherichia coli Uropatógena/metabolismo , Virulencia/fisiología
7.
PLoS One ; 16(2): e0247134, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33592038

RESUMEN

Infectious Bursal Disease Virus (IBDV), the etiological agent of Gumboro disease, causes mortality and immunosuppression in chickens and major losses to poultry industry worldwide. The IBDV major capsid protein VP2 is considered the best candidate for the production of novel subunit vaccines. This structural protein contains the major conformational epitopes responsible for the induction of IBDV neutralizing antibodies in chickens and has been demonstrated able to form supramolecular structures in yeast and insect cells. The aim of this study was to express an engineered version of the VP2 protein (His-pVP2) to verify its ability to self-assemble into virus-like particles in plants. The recombinant VP2 was transiently expressed by agroinfiltration in Nicotiana benthamiana and transmission electron microscopy of sucrose density gradient fractions revealed the presence of a mixed population of differently shaped particles ranging from spherical capsids, with a diameter between ~25 and ~70 nm, to tubular structures, with variable length (from 100 to 400 nm). The recombinant VP2-based particles when used for the intramuscular immunization of specific-pathogen-free chicks resulted able to induce the production of anti-IBDV specific antibodies at titers comparable to those induced by a commercial vaccine. Moreover, all the immunized birds survived to the challenge with a Moroccan very virulent IBDV strain with no major histomorphological alterations of the Bursa of Fabricius, similarly to what obtained with the commercial inactivated vaccine.


Asunto(s)
Virus de la Enfermedad Infecciosa de la Bolsa/patogenicidad , Nicotiana/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Western Blotting , Cápside/metabolismo , Pollos , Ensayo de Inmunoadsorción Enzimática , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Microscopía Electrónica de Transmisión , Proteínas Recombinantes/genética , Nicotiana/genética , Virulencia/genética , Virulencia/fisiología
8.
Sci Rep ; 11(1): 972, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441661

RESUMEN

Francisella tularensis is one of the most virulent pathogenic bacteria causing the acute human respiratory disease tularemia. While the mechanisms underlying F. tularensis pathogenesis are largely unknown, previous studies have shown that a F. novicida transposon mutant with insertions in a gene coding for a putative lysine decarboxylase was attenuated in mouse spleen, suggesting a possible role of its protein product as a virulence factor. Therefore, we set out to structurally and functionally characterize the F. novicida lysine decarboxylase, which we termed LdcF. Here, we investigate the genetic environment of ldcF as well as its evolutionary relationships with other basic AAT-fold amino acid decarboxylase superfamily members, known as key actors in bacterial adaptative stress response and polyamine biosynthesis. We determine the crystal structure of LdcF and compare it with the most thoroughly studied lysine decarboxylase, E. coli LdcI. We analyze the influence of ldcF deletion on bacterial growth under different stress conditions in dedicated growth media, as well as in infected macrophages, and demonstrate its involvement in oxidative stress resistance. Finally, our mass spectrometry-based quantitative proteomic analysis enables identification of 80 proteins with expression levels significantly affected by ldcF deletion, including several DNA repair proteins potentially involved in the diminished capacity of the F. novicida mutant to deal with oxidative stress. Taken together, we uncover an important role of LdcF in F. novicida survival in host cells through participation in oxidative stress response, thereby singling out this previously uncharacterized protein as a potential drug target.


Asunto(s)
Proteínas Bacterianas/metabolismo , Carboxiliasas/metabolismo , Francisella tularensis/metabolismo , Estrés Oxidativo/fisiología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Reparación del ADN/fisiología , Escherichia coli/metabolismo , Macrófagos/metabolismo , Ratones , Proteómica/métodos , Alineación de Secuencia , Tularemia/microbiología , Virulencia/fisiología
9.
Biochem Soc Trans ; 49(1): 455-465, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33492383

RESUMEN

The integrity of the plasma membranes is extremely crucial for the survival and proper functioning of the cells. Organisms from all kingdoms of life employ specialized pore-forming proteins and toxins (PFPs and PFTs) that perforate cell membranes, and cause detrimental effects. PFPs/PFTs exert their damaging actions by forming oligomeric pores in the membrane lipid bilayer. PFPs/PFTs play important roles in diverse biological processes. Many pathogenic bacteria secrete PFTs for executing their virulence mechanisms. The immune system of the higher vertebrates employs PFPs to kill pathogen-infected cells and transformed cancer cells. The most obvious consequence of membrane pore-formation by the PFPs/PFTs is the killing of the target cells due to the disruption of the permeability barrier function of the plasma membranes. PFPs/PFTs can also activate diverse cellular processes that include activation of the stress-response pathways, induction of programmed cell death, and inflammation. Upon attack by the PFTs, host cells may also activate pathways to repair the injured membranes, restore cellular homeostasis, and trigger inflammatory immune responses. In this article, we present an overview of the diverse cellular responses that are triggered by the PFPs/PFTs, and their implications in the process of pathogen infection and immunity.


Asunto(s)
Inmunidad , Infecciones/patología , Proteínas Citotóxicas Formadoras de Poros/farmacología , Toxinas Biológicas/farmacología , Animales , Membrana Celular/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/fisiología , Infecciones/inmunología , Membrana Dobles de Lípidos/metabolismo , Virulencia/fisiología
10.
EMBO J ; 40(2): e105699, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33347626

RESUMEN

Pathogen type 3 secretion systems (T3SS) manipulate host cell pathways by directly delivering effector proteins into host cells. In Vibrio parahaemolyticus, the leading cause of bacterial seafood-borne diarrheal disease, we showed that a T3SS effector, VgpA, localizes to the host cell nucleolus where it binds Epstein-Barr virus nuclear antigen 1-binding protein 2 (EBP2). An amino acid substitution in VgpA (VgpAL10A ) did not alter its translocation to the nucleus but abolished the effector's capacity to interact with EBP2. VgpA-EBP2 interaction led to the re-localization of c-Myc to the nucleolus and increased cellular rRNA expression and proliferation of cultured cells. The VgpA-EBP2 interaction elevated EBP2's affinity for c-Myc and prolonged the oncoprotein's half-life. Studies in infant rabbits demonstrated that VgpA is translocated into intestinal epithelial cells, where it interacts with EBP2 and leads to nucleolar re-localization of c-Myc. Moreover, the in vivo VgpA-EBP2 interaction during infection led to proliferation of intestinal cells and heightened V. parahaemolyticus' colonization and virulence. These observations suggest that direct effector stimulation of a c-Myc controlled host cell growth program can contribute to pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Nucléolo Celular/metabolismo , Proliferación Celular/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Vibrio parahaemolyticus/metabolismo , Virulencia/fisiología , Animales , Células CACO-2 , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Herpesvirus Humano 4/patogenicidad , Humanos , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/metabolismo , Conejos , Vibriosis/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-34977446

RESUMEN

PURPOSE: Family cancer history is an important component of genetic testing guidelines that estimate which patients with breast cancer are most likely to carry a germline pathogenic variant (PV). However, we do not know whether more extensive family history is differentially associated with PVs in specific genes. METHODS: All women diagnosed with breast cancer in 2013-2017 and reported to statewide SEER registries of Georgia and California were linked to clinical genetic testing results and family history from two laboratories. Family history was defined as strong (suggestive of PVs in high-penetrance genes such as BRCA1/2 or TP53, including male breast, ovarian, pancreatic, sarcoma, or multiple female breast cancers), moderate (any other cancer history), or none. Among established breast cancer susceptibility genes (ATM, BARD1, BRCA1, BRCA2, CDH1, CHEK2, NF1, PALB2, PTEN, RAD51C, RAD51D, and TP53), we evaluated PV prevalence according to family history extent and breast cancer subtype. We used a multivariable model to test for interaction between affected gene and family history extent for ATM, BRCA1/2, CHEK2, and PALB2. RESULTS: A total of 34,865 women linked to genetic results. Higher PV prevalence with increasing family history extent (P < .001) was observed only with BRCA1 (3.04% with none, 3.22% with moderate, and 4.06% with strong history) and in triple-negative breast cancer with PALB2 (0.75% with none, 2.23% with moderate, and 2.63% with strong history). In a multivariable model adjusted for age and subtype, there was no interaction between family history extent and PV prevalence for any gene except PALB2 (P = .037). CONCLUSION: Extent of family cancer history is not differentially associated with PVs across established breast cancer susceptibility genes and cannot be used to personalize genes selected for testing.


Asunto(s)
Neoplasias de la Mama/genética , Anamnesis/estadística & datos numéricos , Virulencia/genética , Adulto , Anciano , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/epidemiología , California/epidemiología , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Georgia/epidemiología , Humanos , Persona de Mediana Edad , Virulencia/fisiología
12.
Methods Mol Biol ; 2210: 97-112, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32815131

RESUMEN

Porphyromonas gingivalis is a gram-negative, rod-shaped, nonmotile bacterium belonging to the phylum Bacteroidetes. It produces abundant amounts of proteases in both cell-associated and secretory forms, including a group of cysteine proteases referred to as gingipains, which have attracted much attention due to their high proteolytic activity associated with pathogenicity. Gingipains are grouped into arginine (R)-specific (RgpA and RgpB) and lysine (K)-specific (Kgp) types. Both Rgp (collective term for RgpA and RgpB) and Kgp gingipains play crucial roles in the virulence of P. gingivalis, including the degradation of host periodontal tissues, disruption of host defense mechanisms, and loss of viability in host cells, such as fibroblasts and endothelial cells. In addition to their function in virulence, gingipains are also essential for the growth and survival of P. gingivalis in periodontal pockets through the acquisition of amino acids and heme groups. Furthermore, Rgp and Kgp gingipains are critical in processing fimbriae and several bacterial proteins that contribute to hemagglutination, coaggregation, and hemoglobin binding. This chapter describes the methods used to analyze gingipains.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cisteína-Endopeptidasas Gingipaínas/metabolismo , Porphyromonas gingivalis/metabolismo , Animales , Arginina/metabolismo , Cisteína Endopeptidasas/metabolismo , Células Endoteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Fimbrias Bacterianas/metabolismo , Cobayas , Hemaglutinación/fisiología , Hemaglutininas/metabolismo , Lisina/metabolismo , Virulencia/fisiología
13.
J Immunol ; 205(11): 3095-3106, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33148716

RESUMEN

Iron is an essential element for Mycobacterium tuberculosis; it has at least 40 enzymes that require iron as a cofactor. Accessibility of iron at the phagosomal surface inside macrophage is crucial for survival and virulence of M. tuberculosis ESAT-6, a 6-kDa-secreted protein of region of difference 1, is known to play a crucial role in virulence and pathogenesis of M. tuberculosis In our earlier study, we demonstrated that ESAT-6 protein interacts with ß-2-microglobulin (ß2M) and affects class I Ag presentation through sequestration of ß2M inside endoplasmic reticulum, which contributes toward inhibition of MHC class I:ß2M:peptide complex formation. The 6 aa at C-terminal region of ESAT-6 are essential for ESAT6:ß2M interaction. ß2M is essential for proper folding of HFE, CD1, and MHC class I and their surface expression. It is known that M. tuberculosis recruit holotransferrin at the surface of the phagosome. But the upstream mechanism by which it modulates holotransferrin-mediated iron uptake at the surface of macrophage is not well understood. In the current study, we report that interaction of the ESAT-6 protein with ß2M causes downregulation of surface HFE, a protein regulating iron homeostasis via interacting with transferrin receptor 1 (TFR1). We found that ESAT-6:ß2M interaction leads to sequestration of HFE in endoplasmic reticulum, causing poorer surface expression of HFE and HFE:TFR1 complex (nonfunctional TFR1) in peritoneal macrophages from C57BL/6 mice, resulting in increased holotransferrin-mediated iron uptake in these macrophages. These studies suggest that M. tuberculosis probably targets the ESAT-6 protein to increase iron uptake.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Regulación hacia Abajo/fisiología , Proteína de la Hemocromatosis/metabolismo , Macrófagos Peritoneales/metabolismo , Mycobacterium tuberculosis/metabolismo , Transferrina/metabolismo , Animales , Transporte Biológico/fisiología , Retículo Endoplásmico/metabolismo , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Transferrina/metabolismo , Virulencia/fisiología , Microglobulina beta-2/metabolismo
14.
Biomolecules ; 10(10)2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33050283

RESUMEN

Banana Fusarium wilt (BFW), which is one of the most important banana diseases worldwide, is mainly caused by Fusarium oxysporum f. sp. cubense tropic race 4 (Foc TR4). In this study, we conducted secretome analysis of Foc R1 and Foc TR4 and discovered a total of 120 and 109 secretory proteins (SPs) from Foc R1 cultured alone or with banana roots, respectively, and 129 and 105 SPs respectively from Foc TR4 cultured under the same conditions. Foc R1 and Foc TR4 shared numerous SPs associated with hydrolase activity, oxidoreductase activity, and transferase activity. Furthermore, in culture with banana roots, Foc R1 and Foc TR4 secreted many novel SPs, of which approximately 90% (Foc R1; 57/66; Foc TR4; 50/55) were unconventional SPs without signal peptides. Comparative analysis of SPs in Foc R1 and Foc TR4 revealed that Foc TR4 not only generated more specific SPs but also had a higher proportion of SPs involved in various metabolic pathways, such as phenylalanine metabolism and cysteine and methionine metabolism. The cysteine biosynthesis enzyme O-acetylhomoserine (thiol)-lyase (OASTL) was the most abundant root inducible Foc TR4-specific SP. In addition, knockout of the OASTL gene did not affect growth of Foc TR4; but resulted in the loss of pathogenicity in banana 'Brazil'. We speculated that OASTL functions in banana by interfering with the biosynthesis of cysteine, which is the precursor of an enormous number of sulfur-containing defense compounds. Overall, our studies provide a basic understanding of the SPs in Foc R1 and Foc TR4; including a novel effector in Foc TR4.


Asunto(s)
Liasas de Carbono-Oxígeno , Fusarium/patogenicidad , Musa/microbiología , Proteoma/metabolismo , Liasas de Carbono-Oxígeno/genética , Liasas de Carbono-Oxígeno/aislamiento & purificación , Liasas de Carbono-Oxígeno/metabolismo , Fusarium/química , Fusarium/genética , Fusarium/metabolismo , Perfilación de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Organismos Modificados Genéticamente , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/microbiología , Raíces de Plantas/microbiología , Proteoma/análisis , Proteoma/genética , Vías Secretoras/genética , Transcriptoma , Virulencia/fisiología , Factores de Virulencia/genética , Factores de Virulencia/aislamiento & purificación , Factores de Virulencia/metabolismo
15.
Methods Mol Biol ; 2134: 199-206, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32632871

RESUMEN

Macrophages are phagocytic cells that constitute the primary barrier against pathogens. After phagocytosis a single-membraned vesicle that contains the pathogen is formed. This phagosome undergoes a maturation process to acquire an increasingly antimicrobial environment. Leptospiral uptake by macrophages induces the formation of a Leptospira-containing phagosome (LCP). The kinetics of lysosomal marker recruitment by the LCP is correlated with virulence. This chapter presents a protocol to study the intracellular trafficking of Leptospira spp. within macrophages by fluorescent labeling bacteria and different markers of the phagocytic pathway. We also describe a method to evaluate the bacterial survival within macrophages.


Asunto(s)
Transporte Biológico/fisiología , Leptospira/fisiología , Macrófagos/microbiología , Animales , Línea Celular , Lisosomas/fisiología , Ratones , Fagocitosis/fisiología , Fagosomas/fisiología , Células RAW 264.7 , Virulencia/fisiología
16.
Sci Rep ; 10(1): 9421, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32523093

RESUMEN

Although shedding of zoonotic brucellae in milk has been demonstrated in natural hosts, these data are still missing for the standard murine infection model. We therefore analysed shedding kinetics and the niche of B. melitensis in murine milk. Pregnant Balb/cByJ mice were intraperitoneally infected with 105 CFU of the 16 M reference strain, a 16 M mCherry mutant or a human isolate. Milk was collected over the course of lactation, and subjected to culture and immunofluorescence assays. Bacteria were also quantified in spleen and mammary glands of maternal mice and in spleen of the litter. The shedding of the three strains did not differ significantly (p = 0.301), ranging from log10 1.5 to 4.04 CFU/ml. A total of 73% of the mice excreted B. melitensis into the milk with peak values at mid-lactation; up to 30 bacteria/cell were found in macrophages and neutrophils. While the bacterial counts in the spleen of lactating females confirmed a well-established infection, only 50% of the pups harboured brucellae in their spleen, including the spleen of an uninfected pup fed by an infected foster mother. In conclusion, the murine model of infection may contribute to a better understanding of the zoonotic transmission of brucellosis.


Asunto(s)
Brucella melitensis/fisiología , Brucelosis/microbiología , Macrófagos/microbiología , Leche/microbiología , Animales , Modelos Animales de Enfermedad , Femenino , Lactancia/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Embarazo , Bazo/microbiología , Virulencia/fisiología
17.
Biochem Biophys Res Commun ; 529(1): 97-103, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32560826

RESUMEN

Enterovirus A71 (EVA71) and Coxsackievirus A10 (CVA10) are representative types of Enterovirus A. Dependent on the host cell types, the EVA71 entry may utilize clathrin-, caveola-, and endophilin-A2-mediated endocytosis. However, the cell-entry and intracellular trafficking pathways of CVA10, using KREMEN1 as its receptor, are unclear. Here, we tested the relevant mechanisms through RNA interference (RNAi) and chemical inhibitors. We found that endocytosis of EVA71 and CVA10 in rhabdomyosarcoma (RD) cells engaged multiple pathways, and both viruses required Rac1. Interestingly, while CDC42 and Pak1 participated in EVA71 infection, PI3K played a role in CVA10 infection. The functions of Rab proteins in intracellular trafficking of CVA10 and EVA71 were examined by RNAi. Knockdown of Rab5 and Rab21 significantly reduced CVA10 infectivity, while knockdown of Rab5, Rab7 and Rab9 reduced EVA71 infectivity. Confocal microscopy confirmed the colocalization of CVA10 virions with Rab5 or Rab21, and colocalization of EVA71 virions with Rab5 or Rab7. Additionally, we observed that both CVA10 and EVA71 infections were inhibited by endosome acidification inhibitors, bafilomycin-A1 and NH4Cl. Together, our findings comparatively illustrate the entry and intracellular trafficking processes of representative Enterovirus A types and revealed novel enterovirus intervention targets.


Asunto(s)
Enterovirus Humano A/fisiología , Enterovirus Humano A/patogenicidad , Enterovirus/fisiología , Enterovirus/patogenicidad , Proteínas de Unión al GTP rab5/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Línea Celular Tumoral , Infecciones por Coxsackievirus/etiología , Infecciones por Coxsackievirus/virología , Endocitosis/fisiología , Endosomas/metabolismo , Infecciones por Enterovirus/etiología , Infecciones por Enterovirus/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Virulencia/fisiología , Internalización del Virus , Proteínas de Unión al GTP rab5/antagonistas & inhibidores , Proteínas de Unión al GTP rab5/genética
18.
J Med Microbiol ; 69(7): 1020-1033, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32589124

RESUMEN

Introduction. The incidence of Mycobacterium avium complex (MAC) pulmonary disease (MAC PD), a refractory chronic respiratory tract infection, is increasing worldwide. MAC has three predominant colony morphotypes: smooth opaque (SmO), smooth transparent (SmT) and rough (Rg).Aim. To determine whether colony morphotypes can predict the prognosis of MAC PD, we evaluated the virulence of SmO, SmT and Rg in mice and in human macrophages.Methodology. We compared the characteristics of mice and human macrophages infected with the SmO, SmT, or Rg morphotypes of M. avium subsp. hominissuis 104. C57BL/6 mice and human macrophages derived from peripheral mononuclear cells were used in these experiments.Results. In comparison to SmO- or SmT-infected mice, Rg-infected mice revealed severe pathologically confirmed pneumonia, increased lung weight and increased lung bacterial burden. Rg-infected macrophages revealed significant cytotoxicity, increased bacterial burden, secretion of proinflammatory cytokines (TNF-α and IL-6) and chemokines (CCL5 and CCL3), and formation of cell clusters. Rg formed larger bacterial aggregates than SmO and SmT. Cytotoxicity, bacterial burden and secretion of IL-6, CCL5 and CCL3 were induced strongly by Rg infection, and were decreased by disaggregation of the bacteria.Conclusion. M. avium Rg, which is associated with bacterial aggregation, has the highest virulence among the predominant colony morphotypes.


Asunto(s)
Macrófagos/metabolismo , Mycobacterium avium/genética , Mycobacterium avium/metabolismo , Animales , Citocinas , Femenino , Humanos , Incidencia , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mycobacterium avium/patogenicidad , Complejo Mycobacterium avium/metabolismo , Complejo Mycobacterium avium/patogenicidad , Infección por Mycobacterium avium-intracellulare/epidemiología , Infección por Mycobacterium avium-intracellulare/metabolismo , Fenotipo , Virulencia/fisiología
19.
Proc Natl Acad Sci U S A ; 117(20): 10989-10999, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32354997

RESUMEN

Staphylococcus aureus infections can lead to diseases that range from localized skin abscess to life-threatening toxic shock syndrome. The SrrAB two-component system (TCS) is a global regulator of S. aureus virulence and critical for survival under environmental conditions such as hypoxic, oxidative, and nitrosative stress found at sites of infection. Despite the critical role of SrrAB in S. aureus pathogenicity, the mechanism by which the SrrAB TCS senses and responds to these environmental signals remains unknown. Bioinformatics analysis showed that the SrrB histidine kinase contains several domains, including an extracellular Cache domain and a cytoplasmic HAMP-PAS-DHp-CA region. Here, we show that the PAS domain regulates both kinase and phosphatase enzyme activity of SrrB and present the structure of the DHp-CA catalytic core. Importantly, this structure shows a unique intramolecular cysteine disulfide bond in the ATP-binding domain that significantly affects autophosphorylation kinetics. In vitro data show that the redox state of the disulfide bond affects S. aureus biofilm formation and toxic shock syndrome toxin-1 production. Moreover, with the use of the rabbit infective endocarditis model, we demonstrate that the disulfide bond is a critical regulatory element of SrrB function during S. aureus infection. Our data support a model whereby the disulfide bond and PAS domain of SrrB sense and respond to the cellular redox environment to regulate S. aureus survival and pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cisteína/metabolismo , Proteínas Represoras/metabolismo , Staphylococcus aureus/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas , Secuencia de Bases , Biopelículas , Dominio Catalítico , Modelos Animales de Enfermedad , Endocarditis , Enterotoxinas , Femenino , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Masculino , Modelos Moleculares , Mutación , Oxidación-Reducción , Dominios Proteicos , Conejos , Proteínas Represoras/química , Proteínas Represoras/genética , Sepsis , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Superantígenos , Thermotoga maritima , Virulencia/genética , Virulencia/fisiología
20.
Infect Immun ; 88(5)2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32122940

RESUMEN

Bovine digital dermatitis (BDD), an infectious disease of the bovine foot with a predominant treponemal etiology, is a leading cause of lameness in dairy and beef herds worldwide. BDD is poorly responsive to antimicrobial therapy and exhibits a relapsing clinical course; an effective vaccine is therefore urgently sought. Using a reverse vaccinology approach, the present study surveyed the genomes of the three BDD-associated Treponema phylogroups for putative ß-barrel outer membrane proteins and considered their potential as vaccine candidates. Selection criteria included the presence of a signal peptidase I cleavage site, a predicted ß-barrel fold, and cross-phylogroup homology. Four candidate genes were overexpressed in Escherichia coli BL21(DE3), refolded, and purified. Consistent with their classification as ß-barrel OMPs, circular-dichroism spectroscopy revealed the adoption of a predominantly ß-sheet secondary structure. These recombinant proteins, when screened for their ability to adhere to immobilized extracellular matrix (ECM) components, exhibited a diverse range of ligand specificities. All four proteins specifically and dose dependently adhered to bovine fibrinogen. One recombinant protein was identified as a candidate diagnostic antigen (disease specificity, 75%). Finally, when adjuvanted with aluminum hydroxide and administered to BDD-naive calves using a prime-boost vaccination protocol, these proteins were immunogenic, eliciting specific IgG antibodies. In summary, we present the description of four putative treponemal ß-barrel OMPs that exhibit the characteristics of multispecific adhesins. The observed interactions with fibrinogen may be critical to host colonization and it is hypothesized that vaccination-induced antibody blockade of these interactions will impede treponemal virulence and thus be of therapeutic value.


Asunto(s)
Formación de Anticuerpos/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Dermatitis Digital/inmunología , Dermatitis Digital/microbiología , Treponema/inmunología , Treponema/patogenicidad , Adhesinas Bacterianas/metabolismo , Animales , Adhesión Bacteriana/fisiología , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , ADN Bacteriano/genética , Escherichia coli/inmunología , Escherichia coli/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/microbiología , Inmunoglobulina G/metabolismo , Proteínas de la Membrana/metabolismo , Filogenia , Conformación Proteica en Lámina beta , Serina Endopeptidasas/metabolismo , Virulencia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA