Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Circ Res ; 133(8): 704-719, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37698017

RESUMEN

BACKGROUND: Epigenetic regulation of vascular remodeling in pulmonary hypertension (PH) is poorly understood. Transcription regulating, histone acetylation code alters chromatin accessibility to promote transcriptional activation. Our goal was to identify upstream mechanisms that disrupt epigenetic equilibrium in PH. METHODS: Human pulmonary artery smooth muscle cells (PASMCs), human idiopathic pulmonary arterial hypertension (iPAH):human PASMCs, iPAH lung tissue, failed donor lung tissue, human pulmonary microvascular endothelial cells, iPAH:PASMC and non-iPAH:PASMC RNA-seq databases, NanoString nCounter, and cleavage under targets and release using nuclease were utilized to investigate histone acetylation, hyperacetylation targets, protein and gene expression, sphingolipid activation, cell proliferation, and gene target identification. SPHK2 (sphingosine kinase 2) knockout was compared with control C57BL/6NJ mice after 3 weeks of hypoxia and assessed for indices of PH. RESULTS: We identified that Human PASMCs are vulnerable to the transcription-promoting epigenetic mediator histone acetylation resulting in alterations in transcription machinery and confirmed its pathological existence in PH:PASMC cells. We report that SPHK2 is elevated as much as 20-fold in iPAH lung tissue and is elevated in iPAH:PASMC cells. During PH pathogenesis, nuclear SPHK2 activates nuclear bioactive lipid S1P (sphingosine 1-phosphate) catalyzing enzyme and mediates transcription regulating histone H3K9 acetylation (acetyl histone H3 lysine 9 [Ac-H3K9]) through EMAP (endothelial monocyte activating polypeptide) II. In iPAH lungs, we identified a 4-fold elevation of the reversible epigenetic transcription modulator Ac-H3K9:H3 ratio. Loss of SPHK2 inhibited hypoxic-induced PH and Ac-H3K9 in mice. We discovered that pulmonary vascular endothelial cells are a priming factor of the EMAP II/SPHK2/S1P axis that alters the acetylome with a specificity for PASMC, through hyperacetylation of histone H3K9. Using cleavage under targets and release using nuclease, we further show that EMAP II-mediated SPHK2 has the potential to modify the local transcription machinery of pluripotency factor KLF4 (Krüppel-like factor 4) by hyperacetylating KLF4 Cis-regulatory elements while deletion and targeted inhibition of SPHK2 rescues transcription altering Ac-H3K9. CONCLUSIONS: SPHK2 expression and its activation of the reversible histone H3K9 acetylation in human pulmonary artery smooth muscle cell represent new therapeutic targets that could mitigate PH vascular remodeling.


Asunto(s)
Hipertensión Pulmonar , Humanos , Ratones , Animales , Hipertensión Pulmonar/metabolismo , Histonas/metabolismo , Epigénesis Genética , Células Endoteliales/metabolismo , Remodelación Vascular , Ratones Endogámicos C57BL , Arteria Pulmonar/metabolismo , Proliferación Celular , Hipoxia/complicaciones , Miocitos del Músculo Liso/metabolismo , Células Cultivadas
2.
J Microbiol Biotechnol ; 33(3): 299-309, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36788458

RESUMEN

Glutathione peroxidases (Gpx) are a group of antioxidant enzymes that protect cells or tissues against damage from reactive oxygen species (ROS). The Gpx proteins identified in mammals exhibit high catalytic activity toward glutathione (GSH). In contrast, a variety of non-mammalian Gpx proteins from diverse organisms, including fungi, plants, insects, and rodent parasites, show specificity for thioredoxin (TRX) rather than GSH and are designated as TRX-dependent peroxiredoxins. However, the study of the properties of Gpx in the environmental microbiome or isolated bacteria is limited. In this study, we analyzed the Gpx sequences, identified the characteristics of sequences and structures, and found that the environmental microbiome Gpx proteins should be classified as TRX-dependent, Gpx-like peroxiredoxins. This classification is based on the following three items of evidence: i) the conservation of the peroxidatic Cys residue; ii) the existence and conservation of the resolving Cys residue that forms the disulfide bond with the peroxidatic cysteine; and iii) the absence of dimeric and tetrameric interface domains. The conservation/divergence pattern of all known bacterial Gpx-like proteins in public databases shows that they share common characteristics with that from the environmental microbiome and are also TRX-dependent. Moreover, phylogenetic analysis shows that the bacterial Gpx-like proteins exhibit a star-like radiating phylogenetic structure forming a highly diverse genetic pool of TRX-dependent, Gpx-like peroxidases.


Asunto(s)
Peroxidasas , Peroxirredoxinas , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/química , Glutatión Peroxidasa/metabolismo , Peroxidasas/genética , Peroxidasas/metabolismo , Filogenia , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Glutatión/metabolismo , Bacterias/genética , Bacterias/metabolismo , Oxidación-Reducción
3.
J Biol Chem ; 298(6): 101940, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35430253

RESUMEN

Trafficking of M-protein (Mprt) from the cytosol of Group A Streptococcus pyogenes (GAS) occurs via Sec translocase membrane channels that associate with Sortase A (SrtA), an enzyme that catalyzes cleavage of Mprt at the proximal C-terminal [-LPST355∗GEAA-] motif and subsequent transpeptidation of the Mprt-containing product to the cell wall (CW). These steps facilitate stable exposure of the N-terminus of Mprt to the extracellular milieu where it interacts with ligands. Previously, we found that inactivation of SrtA in GAS cells eliminated Mprt CW transpeptidation but effected little reduction in its cell surface exposure, indicating that the C-terminus of Mprt retained in the cytoplasmic membrane (CM) extends its N-terminus to the cell surface. Herein, we assessed the effects of mutating the Thr355 residue in the WT SrtA consensus sequence (LPST355∗GEAA-) in a specific Mprt, PAM. In vitro, we found that synthetic peptides with mutations (LPSX355GEAA) in the SrtA cleavage site displayed slower cleavage activities with rSrtA than the WT peptide. Aromatic residues at X had the lowest activities. Nonetheless, PAM/[Y355G] still transpeptidated the CW in vivo. However, when using isolated CMs from srtA-inactivated GAS cells, rapid cleavage of PAM/[LPSY355GEAA] occurred at E357∗ but transpeptidation did not take place. These results show that another CM-resident enzyme nonproductively cleaved PAM/[LPSYGE357∗AA]. However, SrtA associated with the translocon channel in vivo cleaved and transpeptidated PAM/[LPSX355∗GEAA] variants. These CM features allow diverse cleavage site variants to covalently attach to the CW despite the presence of other potent nonproductive CM proteases.


Asunto(s)
Aminoaciltransferasas , Proteínas Bacterianas , Pared Celular , Streptococcus pyogenes , Aminoaciltransferasas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Evolución Biológica , Pared Celular/metabolismo , Cisteína Endopeptidasas , Mutación , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/enzimología
4.
J Bacteriol ; 202(11)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32205460

RESUMEN

Streptococcus pyogenes, or group A Streptococcus (GAS), is both a pathogen and an asymptomatic colonizer of human hosts and produces a large number of surface-expressed and secreted factors that contribute to a variety of infection outcomes. The GAS-secreted cysteine protease SpeB has been well studied for its effects on the human host; however, despite its broad proteolytic activity, studies on how this factor is utilized in polymicrobial environments are lacking. Here, we utilized various forms of SpeB protease to evaluate its antimicrobial and antibiofilm properties against the clinically important human colonizer Staphylococcus aureus, which occupies niches similar to those of GAS. For our investigation, we used a skin-tropic GAS strain, AP53CovS+, and its isogenic ΔspeB mutant to compare the production and activity of native SpeB protease. We also generated active and inactive forms of recombinant purified SpeB for functional studies. We demonstrate that SpeB exhibits potent biofilm disruption activity at multiple stages of S. aureus biofilm formation. We hypothesized that the surface-expressed adhesin SdrC in S. aureus was cleaved by SpeB, which contributed to the observed biofilm disruption. Indeed, we found that SpeB cleaved recombinant SdrC in vitro and in the context of the full S. aureus biofilm. Our results suggest an understudied role for the broadly proteolytic SpeB as an important factor for GAS colonization and competition with other microorganisms in its niche.IMPORTANCEStreptococcus pyogenes (GAS) causes a range of diseases in humans, ranging from mild to severe, and produces many virulence factors in order to be a successful pathogen. One factor produced by many GAS strains is the protease SpeB, which has been studied for its ability to cleave and degrade human proteins, an important factor in GAS pathogenesis. An understudied aspect of SpeB is the manner in which its broad proteolytic activity affects other microorganisms that co-occupy niches similar to that of GAS. The significance of the research reported herein is the demonstration that SpeB can degrade the biofilms of the human pathogen Staphylococcus aureus, which has important implications for how SpeB may be utilized by GAS to successfully compete in a polymicrobial environment.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas , Exotoxinas/metabolismo , Staphylococcus aureus/enzimología , Staphylococcus aureus/fisiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Proteínas Bacterianas/genética , Exotoxinas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Staphylococcus aureus/genética , Streptococcus pyogenes/genética
5.
J Bacteriol ; 202(10)2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32123038

RESUMEN

Streptococcus pyogenes (Lancefield group A Streptococcus [GAS]) is a ß-hemolytic human-selective pathogen that is responsible for a large number of morbid and mortal infections in humans. For efficient infection, GAS requires different types of surface proteins that provide various mechanisms for evading human innate immune responses, thus enhancing pathogenicity of the bacteria. Many such virulence-promoting proteins, including the major surface signature M protein, are translocated after biosynthesis through the cytoplasmic membrane and temporarily tethered to this membrane via a type 1 transmembrane domain (TMD) positioned near the COOH terminus. In these proteins, a sorting signal, LPXTG, is positioned immediately upstream of the TMD, which is cleaved by the membrane-associated transpeptidase, sortase A (SrtA), leading to the covalent anchoring of these proteins to newly emerging l-Ala-l-Ala cross-bridges of the growing peptidoglycan cell wall. Herein, we show that inactivation of the srtA gene in a skin-tropic pattern D GAS strain (AP53) results in retention of the M protein in the cell membrane. However, while the isogenic AP53 ΔsrtA strain is attenuated in overall pathogenic properties due to effects on the integrity of the cell membrane, our data show that the M protein nonetheless can extend from the cytoplasmic membrane through the cell wall and then to the surface of the bacteria and thereby retain its important properties of productively binding and activating fluid-phase host plasminogen (hPg). The studies presented herein demonstrate an underappreciated additional mechanism of cell surface display of bacterial virulence proteins via their retention in the cell membrane and extension to the GAS surface.IMPORTANCE Group A Streptococcus pyogenes (GAS) is a human-specific pathogen that produces many surface factors, including its signature M protein, that contribute to its pathogenicity. M proteins undergo specific membrane localization and anchoring to the cell wall via the transpeptidase sortase A. Herein, we explored the role of sortase A function on M protein localization, architecture, and function, employing, a skin-tropic GAS isolate, AP53, which expresses a human plasminogen (hPg)-binding M (PAM) Protein. We showed that PAM anchored in the cell membrane, due to the targeted inactivation of sortase A, was nonetheless exposed on the cell surface and functionally interacted with host hPg. We demonstrate that M proteins, and possibly other sortase A-processed proteins that are retained in the cell membrane, can still function to initiate pathogenic processes by this underappreciated mechanism.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Plasminógeno/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/genética , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/genética
6.
Semin Thromb Hemost ; 45(4): 354-372, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31108555

RESUMEN

The relationship between malignancy and coagulopathy is one that is well documented yet incompletely understood. Clinicians have attempted to quantify the hypercoagulable state produced in various malignancies using common coagulation tests such as prothrombin time, activated partial thromboplastin time, and platelet count; however, due to these tests' focus on individual aspects of coagulation during one specific time point, they have failed to provide clinicians the complete picture of malignancy-associated coagulopathy (MAC). Viscoelastic tests (VETs), such as thromboelastography (TEG) and rotational thromboelastometry (ROTEM), are whole blood analyses that have the advantage of providing information related to the cumulative effects of plasma clotting factors, platelets, leukocytes, and red cells during all stages of the coagulation and fibrinolytic processes. VETs have gained popularity in the care of trauma patients to objectively measure trauma-induced coagulopathy (TIC), but the utility of VETs remains yet unrealized in many other medical specialties. The authors discuss the similarities and differences between TIC and MAC, and propose a mechanism for the hypercoagulable state of MAC that revolves around the thrombomodulin-thrombin complex as it switches between activating the protein C anticoagulation pathway or the thrombin activatable fibrinolysis inhibitor coagulation pathway. Additionally, they review the current literature on the use of TEG and ROTEM in patients with various malignancies. Although limited research is currently available, early results demonstrate the utility of both TEG and ROTEM in the prediction of hypercoagulable states and thromboembolic complications in oncologic patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea/diagnóstico , Pruebas de Coagulación Sanguínea/métodos , Neoplasias/complicaciones , Trombosis/diagnóstico , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/complicaciones , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Tromboelastografía/métodos , Tromboembolia/sangre , Tromboembolia/diagnóstico , Tromboembolia/etiología , Trombosis/sangre , Trombosis/complicaciones
7.
Biomed Res Int ; 2018: 1878964, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29977908

RESUMEN

A murine genetic model of LDL-cholesterol- (LDL-C-) driven atherosclerosis, based on complete deficiencies of both the LDL-receptor (Ldlr-/-) and key catalytic component of an apolipoprotein B-edisome complex (Apobec1-/-), which converts apoB-100 to apoB-48, has been extensively characterized. These gene deficiencies allow high levels of apoB-100 to be present and inefficiently cleared, thus leading to very high levels of LDL-C in mice on a normal diet. Many key features of atherosclerotic plaques observed in human familial hypercholesterolemia are found in these mice as they are allowed to age through 72 weeks. The general characteristics include the presence of high levels of LDL-C in plasma and macrophage-related fatty streak formation in the aortic tree, which progressively worsens with age. More specifically, plaque found in the aortic sinuses contains a lipid core with relatively high numbers of macrophages and a smooth muscle cell α-actin- and collagen-containing cap, which thins with age. These critical features of plaque progression suggest that the Ldlr-/-/Apobec1-/- mouse line presents a superior model of LDL-C-driven atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Modelos Animales de Enfermedad , Hipercolesterolemia/complicaciones , Desaminasas APOBEC-1/genética , Animales , Aterosclerosis/etiología , Humanos , Hiperlipoproteinemia Tipo II , Masculino , Ratones , Ratones Noqueados , Receptores de LDL
8.
J Antibiot (Tokyo) ; 71(6): 592-600, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463889

RESUMEN

Bacteriocins hold unprecedented promise as a largely untapped source of antibiotic alternatives in the age of multidrug resistance. Here, we describe the first approach to systematically design variants of a novel AS-48 bacteriocin homologue, which we have termed safencin AS-48, from Bacillus safensis, to gain insights into engineering improved activity of bacteriocins. A library of synthetic peptides in which systematic amino acid substitutions to vary the periodicity and abundance of polar, acidic, aliphatic, and hydrophobic residues were generated for a total of 96 novel peptide variants of a single bacteriocin candidate. Using this method, we identified nine synthetic safencin (syn-safencin) variants with broad and potent antimicrobial activities with minimal inhibitory concentrations (MIC) as low as 250 nM against E. coli, P. aeruginosa, X. axonopodis, and S. pyogenes with minimal cytotoxicity to mammalian cells. It is anticipated that the strategies we have developed will serve as general guides for tuning the specificity of a given natural bacteriocin compound for therapeutic specificity.


Asunto(s)
Antibacterianos/síntesis química , Péptidos Catiónicos Antimicrobianos/síntesis química , Bacteriocinas/síntesis química , Secuencia de Aminoácidos , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Bacillus/química , Bacterias/efectos de los fármacos , Bacteriocinas/farmacología , Simulación por Computador , Diseño de Fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Biblioteca de Péptidos
9.
Biochemistry ; 55(51): 7112-7122, 2016 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-27981829

RESUMEN

Conantokins (con) are short γ-carboxyglutamate (Gla)-containing polypeptides expressed by marine snails that function as antagonists of N-methyl-d-aspartate receptor (NMDAR) ion channels. The Gla residues govern structural conformations and antagonistic activities of the conantokins. In addition to Gla, some conantokins, e.g., conRl-B, also contain a hydroxyproline (HyP or O) residue, which in this case is centrally located in the peptide at position 10. Because conRl-B specifically inhibits ion channels of GluN2B subunit-containing heterotetrameric NMDARs, we evaluated the unusual role of HyP10 in this effect. To accomplish this goal, we examined synthetic variants of conRl-B in which HyP10 was either deleted (conRl-B[ΔO10]) or replaced with alanine (conRl-B[O10A]) or proline (conRl-B[O10P]). The solution structures of these variants were determined by nuclear magnetic resonance spectroscopy. Deletion of HyP10, or replacement of HyP10 with Ala10, attenuated the distortion in the central region of the apo-conRl-B helix and allowed Mg2+-complexed end-to-end α-helix formation. The inhibitory properties of these variants were assessed by measuring NMDA/Gly-stimulated intracellular Ca2+ influx in mice neurons. ConRl-B[O10P] retained its NMDAR ion channel inhibitory activity in wild-type (WT) neurons but lost its GluN2B specificity, whereas conRl-B[ΔO10] showed overall diminished inhibitory function. ConRl-B[O10A] showed attenuated inhibitory function but retained its GluN2B specificity. Thus, HyP10 plays a critical role in maintaining the structural integrity of conRl-B, which can be correlated with its GluN2B subunit-selective inhibition. Weakened inhibition by conRl-B was also observed in neurons lacking either the GluN2C or GluN2D subunit, compared to WT neurons. This suggests that GluN2C and GluN2D are also required for inhibition by conRl-B.


Asunto(s)
Ácido 1-Carboxiglutámico/química , Hidroxiprolina/química , Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Ácido 1-Carboxiglutámico/genética , Ácido 1-Carboxiglutámico/metabolismo , Alanina/química , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Células Cultivadas , Caracol Conus/química , Hidroxiprolina/genética , Hidroxiprolina/metabolismo , Espectroscopía de Resonancia Magnética , Ratones Noqueados , Modelos Moleculares , Mutación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Péptidos/química , Péptidos/genética , Prolina/química , Prolina/genética , Prolina/metabolismo , Multimerización de Proteína , Estructura Secundaria de Proteína , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/genética , Soluciones
10.
Biochem Biophys Res Commun ; 477(3): 503-8, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27317488

RESUMEN

While inflammation is often associated with increased Plasminogen Activator Inhibitor-1 (PAI-1), the functional consequences of PAI-1 in inflammation have yet to be fully determined. The aim of this study was to establish the in vivo relevance of PAI-1 in inflammation. A mouse model of systemic inflammation was employed in wild-type (WT) and PAI-1 deficient (PAI-1(-/-)) mice. Mice survival, macrophage infiltration into the lungs, and plasma levels of pro-inflammatory cytokines were assessed after lipopolysaccharide (LPS) infusion. In vitro experiments were conducted to examine changes in LPS-induced inflammatory responses after PAI-1 exposure. PAI-1 was shown to regulate inflammation, in vivo, and affect macrophage infiltration into lungs. Further, PAI-1 activated macrophages, and increased pro-inflammatory cytokines at both the mRNA and protein levels in these cells. The effect of PAI-1 on macrophage activation was dose-dependent and LPS-independent. Proteolytic inhibitory activity and Lipoprotein Receptor-related Protein (LRP) and vitronectin (VN) binding functions, were not involved in PAI-1-mediated activation of macrophages. However, the effect of PAI-1 on macrophage activation was partially blocked by a TLR4 neutralizing antibody. Furthermore, PAI-1-induced Tumor Necrosis Factor-alpha (TNF-α) and Macrophage Inflammatory Protein-2 (MIP-2) expression was reduced in TLR4(-/-) macrophages compared to WT macrophages. These results demonstrate that PAI-1 is involved in the regulation of host inflammatory responses through Toll-like Receptor-4 (TLR4)-mediated macrophage activation.


Asunto(s)
Activación de Macrófagos/inmunología , Macrófagos/inmunología , Inhibidor 1 de Activador Plasminogénico/fisiología , Receptor Toll-Like 4/fisiología
11.
J Bacteriol ; 198(12): 1712-24, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27044623

RESUMEN

UNLABELLED: The genome of an invasive skin-tropic strain (AP53) of serotype M53 group A Streptococcus pyogenes (GAS) is composed of a circular chromosome of 1,860,554 bp and carries genetic markers for infection at skin locales, viz, emm gene family pattern D and FCT type 3. Through genome-scale comparisons of AP53 with other GAS genomes, we identified 596 candidate single-nucleotide polymorphisms (SNPs) that reveal a potential genetic basis for skin tropism. The genome of AP53 differed by ∼30 point mutations from a noninvasive pattern D serotype M53 strain (Alab49), 4 of which are located in virulence genes. One pseudogene, yielding an inactive sensor kinase (CovS(-)) of the two-component transcriptional regulator CovRS, a major determinant for invasiveness, severely attenuated the expression of the secreted cysteine protease SpeB and enhanced the expression of the hyaluronic acid capsule compared to the isogenic noninvasive AP53/CovS(+) strain. The collagen-binding protein transcript sclB differed in the number of 5'-pentanucleotide repeats in the signal peptides of AP53 and Alab49 (9 versus 15), translating into different lengths of their signal peptides, which nonetheless maintained a full-length translatable coding frame. Furthermore, GAS strain AP53 acquired two phages that are absent in Alab49. One such phage (ΦAP53.2) contains the known virulence factor superantigen exotoxin gene tandem speK-slaA Overall, we conclude that this bacterium has evolved in multiple ways, including mutational variations of regulatory genes, short-tandem-repeat polymorphisms, large-scale genomic alterations, and acquisition of phages, all of which may be involved in shaping the adaptation of GAS in specific infectious environments and contribute to its enhanced virulence. IMPORTANCE: Infectious strains of S. pyogenes (GAS) are classified by their serotypes, relating to the surface M protein, the emm-like subfamily pattern, and their tropicity toward the nasopharynx and/or skin. It is generally agreed that M proteins from pattern D strains, which also directly bind human host plasminogen, are skin tropic. We have sequenced and characterized the genome of an invasive pattern D GAS strain (AP53) in comparison to a very similar strain (Alab49) that is noninvasive and developed a genomic rationale as to possible reasons for the skin tropicity of these two strains and the greater invasiveness of AP53.


Asunto(s)
Proteínas Bacterianas/genética , Genoma Bacteriano , Enfermedades de la Piel/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Animales , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Genómica , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/patogenicidad , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
12.
Neurosci Lett ; 617: 240-6, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-26917100

RESUMEN

The N-Methyl-D-Aspartate Receptors (NMDARs) are heteromeric cation channels involved in learning, memory, and synaptic plasticity, and their dysregulation leads to various neurodegenerative disorders. Recent evidence has shown that apart from the GluN1/GluN2A and GluN1/GluN2B diheteromeric ion channels, the NMDAR also exists as a GluN1/GluN2A/GluN2B triheteromeric channel that occupies the majority of the synaptic space. These GluN1/GluN2A/GluN2B triheteromers exhibit pharmacological and electrophysiological properties that are distinct from the GluN1/GluN2A and GluN1/GluN2B diheteromeric subtypes. However, these receptors have not been characterized with regards to their inhibition by conantokins, as well as their allosteric modulation by polyamines and extracellular protons. Here, we show that the GluN1/GluN2A/GluN2B triheteromeric channels showed less sensitivity to GluN2B-specific conantokin (con)-G and con-RlB, and subunit non-specific con-T, compared to the GluN2A-specific inhibitor TCN-201. Also, spermine modulation of GluN1/GluN2A/GluN2B triheteromers switched its nature from potentiation to inhibition in a pH dependent manner, and was 2.5-fold slower compared to the GluN1/GluN2B diheteromeric channels. Unraveling the distinctive functional attributes of the GluN1/GluN2A/GluN2B triheteromers is physiologically relevant since they form an integral part of the synapse, which will aid in understanding spermine/pH-dependent potentiation of these receptors in pathological settings.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Conotoxinas/farmacología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular , Venenos de Moluscos/farmacología , Técnicas de Placa-Clamp , Péptidos/farmacología , Multimerización de Proteína , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Espermina/farmacología , Sulfonamidas/farmacología
13.
Blood ; 127(9): 1085-96, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26647393

RESUMEN

Extravascular fibrin deposition accompanies many human diseases and causes chronic inflammation and organ damage, unless removed in a timely manner. Here, we used intravital microscopy to investigate how fibrin is removed from extravascular space. Fibrin placed into the dermis of mice underwent cellular endocytosis and lysosomal targeting, revealing a novel intracellular pathway for extravascular fibrin degradation. A C-C chemokine receptor type 2 (CCR2)-positive macrophage subpopulation constituted the majority of fibrin-uptaking cells. Consequently, cellular fibrin uptake was diminished by elimination of CCR2-expressing cells. The CCR2-positive macrophage subtype was different from collagen-internalizing M2-like macrophages. Cellular fibrin uptake was strictly dependent on plasminogen and plasminogen activator. Surprisingly, however, fibrin endocytosis was unimpeded by the absence of the fibrin(ogen) receptors, αMß2 and ICAM-1, the myeloid cell integrin-binding site on fibrin or the endocytic collagen receptor, the mannose receptor. The study identifies a novel fibrin endocytic pathway engaged in extravascular fibrin clearance and shows that interstitial fibrin and collagen are cleared by different subsets of macrophages employing distinct molecular pathways.


Asunto(s)
Endocitosis , Fibrina/metabolismo , Macrófagos/metabolismo , Receptores CCR2/metabolismo , Animales , Bioensayo , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Fibrinolisina/metabolismo , Ratones , Células Mieloides/metabolismo , Plasminógeno/metabolismo , Activadores Plasminogénicos/metabolismo , Proteolisis , Receptores de Quimiocina/metabolismo , Receptores de Péptidos/metabolismo
14.
J Biol Chem ; 290(29): 18156-18172, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26048991

RESUMEN

Conantokins are ~20-amino acid peptides present in predatory marine snail venoms that function as allosteric antagonists of ion channels of the N-methyl-d-aspartate receptor (NMDAR). These peptides possess a high percentage of post-/co-translationally modified amino acids, particularly γ-carboxyglutamate (Gla). Appropriately spaced Gla residues allow binding of functional divalent cations, which induces end-to-end α-helices in many conantokins. A smaller number of these peptides additionally contain 4-hydroxyproline (Hyp). Hyp should prevent adoption of the metal ion-induced full α-helix, with unknown functional consequences. To address this disparity, as well as the role of Hyp in conantokins, we have solved the high resolution three-dimensional solution structure of a Gla/Hyp-containing 18-residue conantokin, conRl-B, by high field NMR spectroscopy. We show that Hyp(10) disrupts only a small region of the α-helix of the Mn(2+)·peptide complex, which displays cation-induced α-helices on each terminus of the peptide. The function of conRl-B was examined by measuring its inhibition of NMDA/Gly-mediated current through NMDAR ion channels in mouse cortical neurons. The conRl-B displays high inhibitory selectivity for subclasses of NMDARs that contain the functionally important GluN2B subunit. Replacement of Hyp(10) with N(8)Q results in a Mg(2+)-complexed end-to-end α-helix, accompanied by attenuation of NMDAR inhibitory activity. However, replacement of Hyp(10) with Pro(10) allowed the resulting peptide to retain its inhibitory property but diminished its GluN2B specificity. Thus, these modified amino acids, in specific peptide backbones, play critical roles in their subunit-selective inhibition of NMDAR ion channels, a finding that can be employed to design NMDAR antagonists that function at ion channels of distinct NMDAR subclasses.


Asunto(s)
Hidroxiprolina/química , Hidroxiprolina/farmacología , Venenos de Moluscos/química , Venenos de Moluscos/farmacología , Péptidos/química , Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Células Cultivadas , Conotoxinas , Caracol Conus/química , Magnesio/metabolismo , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estructura Secundaria de Proteína , Receptores de N-Metil-D-Aspartato/metabolismo
15.
J Bacteriol ; 196(23): 4089-102, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25225265

RESUMEN

The first genome sequence of a group A Streptococcus pyogenes serotype M23 (emm23) strain (M23ND), isolated from an invasive human infection, has been completed. The genome of this opacity factor-negative (SOF(-)) strain is composed of a circular chromosome of 1,846,477 bp. Gene profiling showed that this strain contained six phage-encoded and 24 chromosomally inherited well-known virulence factors, as well as 11 pseudogenes. The bacterium has acquired four large prophage elements, ΦM23ND.1 to ΦM23ND.4, harboring genes encoding streptococcal superantigen (ssa), streptococcal pyrogenic exotoxins (speC, speH, and speI), and DNases (spd1 and spd3), with phage integrase genes being present at one flank of each phage insertion, suggesting that the phages were integrated by horizontal gene transfer. Comparative analyses revealed unique large-scale genomic rearrangements that result in genomic rearrangements that differ from those of previously sequenced GAS strains. These rearrangements resulted in an imbalanced genomic architecture and translocations of chromosomal virulence genes. The covS sensor in M23ND was identified as a pseudogene, resulting in the attenuation of speB function and increased expression of the genes for the chromosomal virulence factors multiple-gene activator (mga), M protein (emm23), C5a peptidase (scpA), fibronectin-binding proteins (sfbI and fbp54), streptolysin O (slo), hyaluronic acid capsule (hasA), streptokinase (ska), and DNases (spd and spd3), which were verified by PCR. These genes are responsible for facilitating host epithelial cell binding and and/or immune evasion, thus further contributing to the virulence of M23ND. In conclusion, strain M23ND has become highly pathogenic as the result of a combination of multiple genetic factors, particularly gene composition and mutations, prophage integrations, unique genomic rearrangements, and regulated expression of critical virulence factors.


Asunto(s)
Orden Génico , Genoma Bacteriano , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/genética , Adhesión Bacteriana , Células Epiteliales/microbiología , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Transferencia de Gen Horizontal , Reacción en Cadena de la Polimerasa , Profagos/genética , Seudogenes , Recombinación Genética , Serogrupo , Streptococcus pyogenes/fisiología , Virulencia , Factores de Virulencia/genética
16.
J Neurosci Res ; 92(12): 1767-74, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25043917

RESUMEN

Retinal ganglion cells (RGCs), which are the sole output neurons of the retina, express N-methyl-D-aspartate receptors (NMDARs), rendering these cells susceptible to glutamate excitotoxicity, with implications for loss of normal RGC excitatory responses in disorders such as glaucoma and diabetic retinopathy. Therefore, antagonists that inhibit NMDAR-mediated currents specifically by targeting the GluN2B component of the ion channel have the potential to serve as a basis for developing potential therapeutics. The roles of peptidic conantokins, which are potent brain neuronal NMDAR inhibitors, were studied. By using patch-clamp whole-cell analyses in dissociated RGCs and retinal whole-mount RGCs, we evaluated the effects of synthetic conantokin-G (conG) and conantokin-T (conT), which are small γ-carboxyglutamate-containing peptides, on NMDA-mediated excitatory responses in mouse RGCs. Both conG and conT inhibited the NMDA-mediated currents of dark-adapted dissociated and whole-mount RGCs in a dose-dependent, reversible, noncompetitive manner. Inhibition of NMDA-mediated steady-state currents by NMDAR nonsubunit-selective conT was approximately threefold greater than GluN2B-selective conG or ifenprodil, demonstrating its potential ability to inhibit both GluN2A- and GluN2B-containing ion channels in RGCs. Because the extent of inhibition of NMDA-evoked currents by conG and the pharmacologic GluN2B-selective inhibitor ifenprodil were similar (40-45%) to that of the GluN2A-selective antagonist NVP-AAM0077, we conclude that the levels of GluN2A and GluN2B subunits are similar in RGCs. These results provide a novel basis for developing effective neuroprotective agents to aid in the prevention of undesired glutamatergic excitotoxicity in neurodegenerative diseases of the retina and demonstrate functional assembly of NMDARs in RGCs.


Asunto(s)
Conotoxinas/farmacología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Venenos de Moluscos/farmacología , Péptidos/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , N-Metilaspartato/farmacología , Técnicas de Placa-Clamp , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Retina/citología
17.
J Trauma Acute Care Surg ; 76(5): 1169-76, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24747445

RESUMEN

BACKGROUND: Coagulopathy in traumatic brain injury (CTBI) is a well-established phenomenon, but its mechanism is poorly understood. Various studies implicate protein C activation related to the global insult of hemorrhagic shock or brain tissue factor release with resultant platelet dysfunction and depletion of coagulation factors. We hypothesized that the platelet dysfunction of CTBI is a distinct phenomenon from the coagulopathy following hemorrhagic shock. METHODS: We used thrombelastography with platelet mapping as a measure of platelet function, assessing the degree of inhibition of the adenosine diphosphate (ADP) and arachidonic acid (AA) receptor pathways. First, we studied the early effect of TBI on platelet inhibition by performing thrombelastography with platelet mapping on rats. We then conducted an analysis of admission blood samples from trauma patients with isolated head injury (n = 70). Patients in shock or on clopidogrel or aspirin were excluded. RESULTS: In rats, ADP receptor inhibition at 15 minutes after injury was 77.6% ± 6.7% versus 39.0% ± 5.3% for controls (p < 0.0001). Humans with severe TBI (Glasgow Coma Scale [GCS] score ≤ 8) showed an increase in ADP receptor inhibition at 93.1% (interquartile range [IQR], 44.8-98.3%; n = 29) compared with 56.5% (IQR, 35-79.1%; n = 41) in milder TBI and 15.5% (IQR, 13.2-29.1%) in controls (p = 0.0014 and p < 0.0001, respectively). No patient had significant hypotension or acidosis. Parallel trends were noted in AA receptor inhibition. CONCLUSION: Platelet ADP and AA receptor inhibition is a prominent early feature of CTBI in humans and rats and is linked to the severity of brain injury in patients with isolated head trauma. This phenomenon is observed in the absence of hemorrhagic shock or multisystem injury. Thus, TBI alone is shown to be sufficient to induce a profound platelet dysfunction.


Asunto(s)
Ácido Araquidónico/antagonistas & inhibidores , Trastornos de la Coagulación Sanguínea/etiología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/complicaciones , Antagonistas del Receptor Purinérgico P2Y/sangre , Choque Hemorrágico/sangre , Choque Hemorrágico/etiología , Adulto , Animales , Ácido Araquidónico/sangre , Trastornos de la Coagulación Sanguínea/sangre , Pruebas de Coagulación Sanguínea , Plaquetas/fisiología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Escala de Coma de Glasgow , Humanos , Masculino , Persona de Mediana Edad , Activación Plaquetaria/fisiología , Pruebas de Función Plaquetaria , Estudios Prospectivos , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Medición de Riesgo , Estadísticas no Paramétricas , Tromboelastografía
18.
J Neurotrauma ; 31(19): 1672-5, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24605991

RESUMEN

Coagulopathy after severe traumatic brain injury (TBI) has been extensively reported. Clinical studies have identified a strong relationship between diminished platelet-rich thrombus formation, responsiveness to adenosine diphosphate agonism, and severity of TBI. The mechanisms that lead to platelet dysfunction in the acute response to TBI are poorly understood. The development of a rodent model of TBI that mimics the coagulopathy observed clinically has recently been reported. Using immunohistochemical techniques and thromboelastography platelet mapping, the current study demonstrated that the expression of coagulation (tissue factor and fibrin) and platelet activation (P-selectin) markers in the injured brain paralleled the alteration in systemic platelet responsiveness to the agonists, adenosine diphosphate and arachodonic acid. Results of this study demonstrate that local procoagulant changes in the injured brain have profound effects on systemic platelet function.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Lesiones Encefálicas/sangre , Encéfalo/patología , Activación Plaquetaria/fisiología , Animales , Coagulación Sanguínea/fisiología , Trastornos de la Coagulación Sanguínea/etiología , Plaquetas/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley
19.
J Neurotrauma ; 31(4): 404-10, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24040968

RESUMEN

Acute coagulopathy is a serious complication of traumatic brain injury (TBI) and is of uncertain etiology because of the complex nature of TBI. However, recent work has shown a correlation between mortality and abnormal hemostasis resulting from early platelet dysfunction. The aim of the current study was to develop and characterize a rodent model of TBI that mimics the human coagulopathic condition so that mechanisms of the early acute coagulopathy in TBI can be more readily assessed. Studies utilizing a highly reproducible constrained blunt-force brain injury in rats demonstrate a strong correlation with important postinjury pathological changes that are observed in human TBI patients, namely, diminished platelet responses to agonists, especially adenosine diphosphate (ADP), and subarachnoid bleeding. Additionally, administration of a direct thrombin inhibitor, preinjury, recovers platelet functionality to ADP stimulation, indicating a direct role for excess thrombin production in TBI-induced early platelet dysfunction.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Plaquetas/efectos de los fármacos , Lesiones Encefálicas/sangre , Heridas no Penetrantes/sangre , Enfermedad Aguda , Adenosina Difosfato/farmacología , Animales , Recuento de Células Sanguíneas , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/patología , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Hirudinas/farmacología , Cinética , Masculino , Tiempo de Tromboplastina Parcial , Agregación Plaquetaria/fisiología , Protrombina/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Hemorragia Subaracnoidea/sangre , Tromboelastografía , Trombina/antagonistas & inhibidores , Heridas no Penetrantes/patología
20.
J Biol Chem ; 289(5): 2992-3000, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24338014

RESUMEN

Plasma plasminogen is the precursor of the tumor angiogenesis inhibitor, angiostatin. Generation of angiostatin in blood involves activation of plasminogen to the serine protease plasmin and facilitated cleavage of two disulfide bonds and up to three peptide bonds in the kringle 5 domain of the protein. The mechanism of reduction of the two allosteric disulfides has been explored in this study. Using thiol-alkylating agents, mass spectrometry, and an assay for angiostatin formation, we show that the Cys(462)-Cys(541) disulfide bond is already cleaved in a fraction of plasma plasminogen and that this reduced plasminogen is the precursor for angiostatin formation. From the crystal structure of plasminogen, we propose that plasmin ligands such as phosphoglycerate kinase induce a conformational change in reduced kringle 5 that leads to attack by the Cys(541) thiolate anion on the Cys(536) sulfur atom of the Cys(512)-Cys(536) disulfide bond, resulting in reduction of the bond by thiol/disulfide exchange. Cleavage of the Cys(512)-Cys(536) allosteric disulfide allows further conformational change and exposure of the peptide backbone to proteolysis and angiostatin release. The Cys(462)-Cys(541) and Cys(512)-Cys(536) disulfides have -/+RHHook and -LHHook configurations, respectively, which are two of the 20 different measures of the geometry of a disulfide bond. Analysis of the structures of the known allosteric disulfide bonds identified six other bonds that have these configurations, and they share some functional similarities with the plasminogen disulfides. This suggests that the -/+RHHook and -LHHook disulfides, along with the -RHStaple bond, are potential allosteric configurations.


Asunto(s)
Angiostatinas/metabolismo , Disulfuros/metabolismo , Fibrinolisina/metabolismo , Plasminógeno/metabolismo , Precursores de Proteínas/metabolismo , Regulación Alostérica , Angiostatinas/química , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Fibrinolisina/química , Humanos , Oxidación-Reducción , Plasminógeno/química , Precursores de Proteínas/química , Estructura Terciaria de Proteína , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA