Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Toxins (Basel) ; 15(9)2023 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-37755954

RESUMEN

Anthrax toxin is a critical virulence factor of Bacillus anthracis. The toxin comprises protective antigen (PA) and two enzymatic moieties, edema factor (EF) and lethal factor (LF), forming bipartite lethal toxin (LT) and edema toxin (ET). PA binds cellular surface receptors and is required for intracellular translocation of the enzymatic moieties. For this reason, anti-PA antibodies have been developed as therapeutics for prophylaxis and treatment of human anthrax infection. Assays described publicly for the control of anti-PA antibody potency quantify inhibition of LT-mediated cell death or the ET-induced increase in c-AMP levels. These assays do not fully reflect and/or capture the pathological functions of anthrax toxin in humans. Herein, we report the development of a cell-based gene reporter potency assay for anti-PA antibodies based on the rapid LT-induced degradation of c-Jun protein, a pathogenic effect that occurs in human cells. This new assay was developed by transducing Hepa1c1c7 cells with an AP-1 reporter lentiviral construct and has been qualified for specificity, accuracy, repeatability, intermediate precision, and linearity. This assay not only serves as a bioassay for LT activity, but has applications for characterization and quality control of anti-PA therapeutic antibodies or other products that target the AP-1 signaling pathway.


Asunto(s)
Carbunco , Toxinas Bacterianas , Humanos , Factor de Transcripción AP-1/genética , Toxinas Bacterianas/genética , Exotoxinas
2.
Viruses ; 15(8)2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37632039

RESUMEN

The recent global COVID-19 pandemic caused by SARS-CoV-2 lasted for over three years. A key measure in combatting this pandemic involved the measurement of the monoclonal antibody (mAb)-mediated inhibition of binding between the spike receptor-binding domain (RBD) and hACE2 receptor. Potency assessments of therapeutic anti-SARS-CoV-2 mAbs typically include binding or cell-based neutralization assays. We assessed the inhibitory activity of five anti-SARS-CoV-2 mAbs using ELISA, surface plasmon resonance (SPR), and four cell-based neutralization assays using different pseudovirus particles and 293T or A549 cells expressing hACE2 with or without TMPRSS2. We assessed the interchangeability between cell-based and binding assays by applying the Bland-Altman method under certain assumptions. Our data demonstrated that the IC50 [nM] values determined by eight neutralization assays are independent of the cell line, presence of TMPRSS2 enzyme on the cell surface, and pseudovirus backbone used. Moreover, the Bland-Altman analysis showed that the IC50 [nM] and KD [nM] values determined by neutralization/ELISA or by SPR are equivalent and that the anti-spike mAb activity can be attributed to one variable directly related to its tertiary conformational structure conformation, rate dissociation constant Koff. This parameter is independent from the concentrations of the components of the mAb:RBD:hACE2 complexes and can be used for a comparison between the activities of the different mAbs.


Asunto(s)
COVID-19 , Humanos , Pandemias , SARS-CoV-2 , Células A549 , Anticuerpos Monoclonales , Anticuerpos Antivirales
3.
Int J Mol Sci ; 22(14)2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34299155

RESUMEN

Proinflammatory cytokine production following infection with severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) is associated with poor clinical outcomes. Like SARS CoV-1, SARS CoV-2 enters host cells via its spike protein, which attaches to angiotensin-converting enzyme 2 (ACE2). As SARS CoV-1 spike protein is reported to induce cytokine production, we hypothesized that this pathway could be a shared mechanism underlying pathogenic immune responses. We herein compared the capabilities of Middle East Respiratory Syndrome (MERS), SARS CoV-1 and SARS CoV-2 spike proteins to induce cytokine expression in human peripheral blood mononuclear cells (PBMC). We observed that only specific commercial lots of SARS CoV-2 induce cytokine production. Surprisingly, recombinant SARS CoV-2 spike proteins from different vendors and batches exhibited different patterns of cytokine induction, and these activities were not inhibited by blockade of spike protein-ACE2 binding using either soluble ACE2 or neutralizing anti-S1 antibody. Moreover, commercial spike protein reagents contained varying levels of lipopolysaccharide (LPS), which correlated directly with their abilities to induce cytokine production. The LPS inhibitor, polymyxin B, blocked this cytokine induction activity. In addition, SARS CoV-2 spike protein avidly bound soluble LPS in vitro, rendering it a cytokine inducer. These results not only suggest caution in monitoring the purity of SARS CoV-2 spike protein reagents, but they indicate the possibility that interactions of SARS CoV-2 spike protein with LPS from commensal bacteria in virally infected mucosal tissues could promote pathogenic inflammatory cytokine production.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Citocinas/metabolismo , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Modelos Biológicos , Glicoproteína de la Espiga del Coronavirus/farmacología , Voluntarios Sanos , Humanos , Técnicas In Vitro , Leucocitos Mononucleares/efectos de los fármacos
4.
Int J Mol Sci ; 22(8)2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918729

RESUMEN

Constitutive photomorphogenic 1 (COP1) is the ubiquitin E3 ligase that mediates degradation of c-Jun protein upon Erk1/2 inactivation. It remains unknown how this protein degradation pathway is regulated. In this study, we investigated the roles of protein phosphatases, ubiquitin-conjugating E2 enzymes (UBE2), and an intrinsic motif of c-Jun in regulating this degradation pathway. By using pharmacological inhibitors and/or gene knockdown techniques, we identified protein phosphatase 1 (PP1) and PP2A as the phosphatases and UBE23d as the UBE2 promoting c-Jun degradation, triggered by Erk1/2 inactivation. In addition, we report that the C-terminus of c-Jun protein facilitates its degradation. The addition of a C-terminal tag or deletion of the last four amino acid residues from the C-terminus of c-Jun protects it from degradation under Erk1/2-inactivating conditions. Taken together, this study reveals that the Erk1/2 inactivation-triggered and COP1-mediated c-Jun degradation is extrinsically and intrinsically regulated, providing a new understanding of the mechanisms underlying this protein degradation pathway.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Línea Celular Tumoral , Activación Enzimática , Humanos , Ratones , Modelos Biológicos , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Proteolisis
5.
Proc Natl Acad Sci U S A ; 117(8): 4078-4087, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32041890

RESUMEN

Anthrax lethal toxin (LT) is a protease virulence factor produced by Bacillus anthracis that is required for its pathogenicity. LT treatment causes a rapid degradation of c-Jun protein that follows inactivation of the MEK1/2-Erk1/2 signaling pathway. Here we identify COP1 as the ubiquitin E3 ligase that is essential for LT-induced c-Jun degradation. COP1 knockdown using siRNA prevents degradation of c-Jun, ETV4, and ETV5 in cells treated with either LT or the MEK1/2 inhibitor, U0126. Immunofluorescence staining reveals that COP1 preferentially localizes to the nuclear envelope, but it is released from the nuclear envelope into the nucleoplasm following Erk1/2 inactivation. At baseline, COP1 attaches to the nuclear envelope via interaction with translocated promoter region (TPR), a component of the nuclear pore complex. Disruption of this COP1-TPR interaction, through Erk1/2 inactivation or TPR knockdown, leads to rapid COP1 release from the nuclear envelope into the nucleoplasm where it degrades COP1 substrates. COP1-mediated degradation of c-Jun protein, combined with LT-mediated blockade of the JNK1/2 signaling pathway, inhibits cellular proliferation. This effect on proliferation is reversed by COP1 knockdown and ectopic expression of an LT-resistant MKK7-4 fusion protein. Taken together, this study reveals that the nuclear envelope acts as a reservoir, maintaining COP1 poised for action. Upon Erk1/2 inactivation, COP1 is rapidly released from the nuclear envelope, promoting the degradation of its nuclear substrates, including c-Jun, a critical transcription factor that promotes cellular proliferation. This regulation allows mammalian cells to respond rapidly to changes in extracellular cues and mediates pathogenic mechanisms in disease states.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Proliferación Celular , Humanos , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 6 Activada por Mitógenos/genética , Proteínas Nucleares/genética , Ubiquitina-Proteína Ligasas/genética
6.
Sci Rep ; 10(1): 2476, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32051479

RESUMEN

PEGylated recombinant human granulocyte colony stimulating factor (pegfilgrastim) is used clinically to accelerate immune reconstitution following chemotherapy and is being pursued for biosimilar development. One challenge to overcome in pegfilgrastim biosimilar development is establishing pharmacokinetic (PK) similarity, which is partly due to the degree of PK variability. We herein report that commercially available G-CSF and PEG ELISA detection kits have different capacities to detect pegfilgrastim aggregates that rapidly form in vitro in physiological conditions. These aggregates can be observed using SDS-PAGE, size-exclusion chromatography, dynamic light scattering, and real-time NMR analysis and are associated with decreased bioactivity as reflected by reduced drug-induced cellular proliferation and STAT3 phosphorylation. Furthermore, individual variability in the stability and detectability of pegfilgrastim in human sera is also observed. Pegfilgrastim levels display marked subject variability in sera from healthy donors incubated at 37 °C. The stability patterns of pegfilgrastim closely match the stability patterns of filgrastim, consistent with a key role for pegfilgrastim's G-CSF moiety in driving formation of inactive aggregates. Taken together, our results indicate that individual variability and ELISA specificity for inactive aggregates are key factors to consider when designing and interpreting studies involving the measurement of serum pegfilgrastim concentrations.


Asunto(s)
Variación Biológica Individual , Filgrastim/farmacocinética , Polietilenglicoles/farmacocinética , Animales , Línea Celular Tumoral , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática/normas , Humanos , Ratones , Factor de Transcripción STAT3/metabolismo
7.
J Infect Dis ; 216(11): 1471-1475, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-28968672

RESUMEN

Gastrointestinal (GI) anthrax is the most prevalent form of naturally acquired Bacillus anthracis infection, which is associated with exposure to vegetative bacteria in infected meat (carnivores) or to fermented rumen contents (herbivores). We assessed whether key host and pathogen factors modulate infectivity and progression of infection using a mouse model of GI infection. Gastric acid neutralization increases infectivity, but 30%-40% of mice succumb to infection without neutralization. Mice either fed or fasted before exposure showed similar infectivity rates. Finally, the pathogen's anthrax lethal factor is required to establish lethal infection, whereas its edema factor modulates progression and dissemination of infection.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/metabolismo , Progresión de la Enfermedad , Ácido Gástrico , Enfermedades Gastrointestinales/microbiología , Factores de Virulencia , Animales , Carbunco/microbiología , Carbunco/patología , Bacillus anthracis/fisiología , Modelos Animales de Enfermedad , Femenino , Enfermedades Gastrointestinales/patología , Corazón/microbiología , Concentración de Iones de Hidrógeno , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Pulmón/microbiología , Pulmón/patología , Ratones , Virulencia
8.
J Biol Chem ; 292(43): 17919-17927, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28893904

RESUMEN

Anthrax is a life-threatening disease caused by infection with Bacillus anthracis, which expresses lethal factor and the receptor-binding protective antigen. These two proteins combine to form anthrax lethal toxin (LT), whose proximal targets are mitogen-activated kinase kinases (MKKs). However, the downstream mediators of LT toxicity remain elusive. Here we report that LT exposure rapidly reduces the levels of c-Jun, a key regulator of cell proliferation and survival. Blockade of proteasome-dependent protein degradation with the 26S proteasome inhibitor MG132 largely restored c-Jun protein levels, suggesting that LT promotes degradation of c-Jun protein. Using the MKK1/2 inhibitor U0126, we further show that MKK1/2-Erk1/2 pathway inactivation similarly reduces c-Jun protein, which was also restored by MG132 pre-exposure. Interestingly, c-Jun protein rebounded to normal levels 4 h following U0126 exposure but not after LT exposure. The restoration of c-Jun in U0126-exposed cells was associated with increased c-Jun mRNA levels and was blocked by inactivation of the JNK1/2 signaling pathway. These results indicate that LT reduces c-Jun both by promoting c-Jun protein degradation via inactivation of MKK1/2-Erk1/2 signaling and by blocking c-Jun gene transcription via inactivation of MKK4-JNK1/2 signaling. In line with the known functions of c-Jun, LT also inhibited cell proliferation. Ectopic expression of LT-resistant MKK2 and MKK4 variants partially restored Erk1/2 and JNK1/2 signaling in LT-exposed cells, enabling the cells to maintain relatively normal c-Jun protein levels and cell proliferation. Taken together, these findings indicate that LT reduces c-Jun protein levels via two distinct mechanisms, thereby inhibiting critical cell functions, including cellular proliferation.


Asunto(s)
Antígenos Bacterianos/farmacología , Bacillus anthracis/química , Toxinas Bacterianas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Antígenos Bacterianos/química , Toxinas Bacterianas/química , Butadienos/farmacología , Células Hep G2 , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-jun/genética
9.
Toxins (Basel) ; 7(10): 3960-76, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26426050

RESUMEN

The principal portal for anthrax infection in natural animal outbreaks is the digestive tract. Enteric exposure to anthrax, which is difficult to detect or prevent in a timely manner, could be exploited as an act of terror through contamination of human or animal food. Our group has developed a novel animal model of gastrointestinal (GI) anthrax for evaluation of disease pathogenesis and experimental therapeutics, utilizing vegetative Bacillus anthracis (Sterne strain) administered to A/J mice (a complement-deficient strain) by oral gavage. We hypothesized that a humanized recombinant monoclonal antibody (mAb) * that neutralizes the protective antigen (PA) component of B. anthracis lethal toxin (LT) and edema toxin (ET) could be an effective treatment. Although the efficacy of this anti-anthrax PA mAb has been shown in animal models of inhalational anthrax, its activity in GI infection had not yet been ascertained. We hereby demonstrate that passive immunotherapy with anti-anthrax PA mAb, administered at the same time as gastrointestinal exposure to B. anthracis, prevents lethal sepsis in nearly all cases (>90%), while a delay of up to forty-eight hours in treatment still greatly reduces mortality following exposure (65%). Moreover, passive immunotherapy protects against enteric invasion, associated mucosal injury and subsequent dissemination by gastrointestinal B. anthracis, indicating that it acts to prevent the initial stages of infection. * Expired raxibacumab being cycled off the Strategic National Stockpile; biological activity confirmed by in vitro assay.


Asunto(s)
Carbunco/terapia , Anticuerpos Monoclonales Humanizados/uso terapéutico , Gastroenteritis/terapia , Inmunización Pasiva/métodos , Sepsis/prevención & control , Animales , Carbunco/inmunología , Carbunco/microbiología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antígenos Bacterianos , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Esquema de Medicación , Femenino , Gastroenteritis/inmunología , Gastroenteritis/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Endogámicos , Sepsis/inmunología
10.
Science ; 345(6204): 1623-1627, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25213377

RESUMEN

Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an inhibitory receptor found on immune cells. The consequences of mutations in CTLA4 in humans are unknown. We identified germline heterozygous mutations in CTLA4 in subjects with severe immune dysregulation from four unrelated families. Whereas Ctla4 heterozygous mice have no obvious phenotype, human CTLA4 haploinsufficiency caused dysregulation of FoxP3(+) regulatory T (Treg) cells, hyperactivation of effector T cells, and lymphocytic infiltration of target organs. Patients also exhibited progressive loss of circulating B cells, associated with an increase of predominantly autoreactive CD21(lo) B cells and accumulation of B cells in nonlymphoid organs. Inherited human CTLA4 haploinsufficiency demonstrates a critical quantitative role for CTLA-4 in governing T and B lymphocyte homeostasis.


Asunto(s)
Antígeno CTLA-4/genética , Mutación de Línea Germinal , Haploinsuficiencia , Enfermedades del Sistema Inmune/genética , Inmunidad/genética , Adulto , Animales , Linfocitos B/inmunología , Femenino , Factores de Transcripción Forkhead/inmunología , Humanos , Masculino , Ratones , Ratones Mutantes , Linaje , Linfocitos T Reguladores/inmunología , Adulto Joven
11.
J Clin Immunol ; 34(4): 436-43, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24682681

RESUMEN

OBJECTIVE: Patients with hypomorphic mutations in Nuclear Factor-κB Essential Modulator (NEMO) are immunodeficient (ID) and most display ectodermal dysplasia and anhidrosis (EDA). We compared cytokine production by NEMO-ID patients with and without EDA. METHODS: PBMCs of NEMO-ID patients, four with EDA carrying E315A, C417R, D311N and Q403X, and three without EDA carrying E315A, E311_L333del and R254G, were cultured with PHA, PHA plus IL-12p70, LPS, LPS plus IFN-γ, TNF and IL-1ß. The production of various cytokines was measured in the supernatants. Fifty-nine healthy individuals served as controls. RESULTS: PBMCs of NEMO-ID patients without EDA produce subnormal amounts of IFN-γ after stimulation with PHA, but normal amounts of IFN-γ after PHA plus IL-12p70. In contrast, IFN-γ production by patients with EDA was low in both cases. Patients with EDA also generate lower PHA-stimulated IL-10 and IL-1ß than controls, whereas the production of these cytokines by patients without EDA was normal. CONCLUSION: Responses of PBMCs in NEMO-ID patients with EDA to PHA with and without IL-12p70 appear less robust than in NEMO-ID patients without EDA. This possibly indicates a better preserved NEMO function in our patients without EDA.


Asunto(s)
Displasia Ectodérmica/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Quinasa I-kappa B/inmunología , Síndromes de Inmunodeficiencia/inmunología , Interferón gamma/biosíntesis , Interleucina-12/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Adulto , Estudios de Casos y Controles , Displasia Ectodérmica/complicaciones , Displasia Ectodérmica/genética , Displasia Ectodérmica/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Quinasa I-kappa B/genética , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Interleucina-10/biosíntesis , Interleucina-1beta/biosíntesis , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Lipopolisacáridos/farmacología , Masculino , Persona de Mediana Edad , Fitohemaglutininas/farmacología , Cultivo Primario de Células , Enfermedades de Inmunodeficiencia Primaria , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
12.
J Biol Chem ; 289(7): 4180-90, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24366872

RESUMEN

Hypoxia is considered to be a contributor to the pathology associated with administration of anthrax lethal toxin (LT). However, we report here that serum lactate levels in LT-treated mice are reduced, a finding inconsistent with the anaerobic metabolism expected to occur during hypoxia. Reduced lactate levels are also observed in the culture supernatants of LT-treated cells. LT inhibits the accumulation of hypoxia-inducible factor (HIF)-1α, a subunit of HIF-1, the master regulator directing cellular responses to hypoxia. The toxin has no effect on the transcription or protein turnover of HIF-1α, but instead it acts to inhibit HIF-1α translation. LT treatment diminishes phosphorylation of eIF4B, eIF4E, and rpS6, critical components of the intracellular machinery required for HIF-1α translation. Moreover, blockade of MKK1/2-ERK1/2, but not p38 or JNK signaling, lowers HIF-1α protein levels in both normoxic and hypoxic conditions, consistent with a role for MKK1 and MKK2 as the major targets of LT responsible for the inhibition of HIF-1α translation. The physiological importance of the LT-induced translation blockade is demonstrated by the finding that LT treatment decreases the survival of hepatocyte cell lines grown in hypoxic conditions, an effect that is overcome by preinduction of HIF-1α. Taken together, these data support a role for LT in dysregulating HIF-1α and thereby disrupting homeostatic responses to hypoxia, an environmental characteristic of certain tissues at baseline and/or during disseminated infection with Bacillus anthracis.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Hipoxia/metabolismo , Biosíntesis de Proteínas , Animales , Carbunco/genética , Carbunco/patología , Hipoxia de la Célula/genética , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Hep G2 , Humanos , Hipoxia/genética , Hipoxia/microbiología , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Sistema de Señalización de MAP Quinasas/genética , Ratones , Fosforilación/genética , Proteína S6 Ribosómica/genética , Proteína S6 Ribosómica/metabolismo
13.
Nat Immunol ; 15(1): 88-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24165795

RESUMEN

The p110δ subunit of phosphatidylinositol-3-OH kinase (PI(3)K) is selectively expressed in leukocytes and is critical for lymphocyte biology. Here we report fourteen patients from seven families who were heterozygous for three different germline, gain-of-function mutations in PIK3CD (which encodes p110δ). These patients presented with sinopulmonary infections, lymphadenopathy, nodular lymphoid hyperplasia and viremia due to cytomegalovirus (CMV) and/or Epstein-Barr virus (EBV). Strikingly, they had a substantial deficiency in naive T cells but an over-representation of senescent effector T cells. In vitro, T cells from patients exhibited increased phosphorylation of the kinase Akt and hyperactivation of the metabolic checkpoint kinase mTOR, enhanced glucose uptake and terminal effector differentiation. Notably, treatment with rapamycin to inhibit mTOR activity in vivo partially restored the abundance of naive T cells, largely 'rescued' the in vitro T cell defects and improved the clinical course.


Asunto(s)
Senescencia Celular/genética , Mutación de Línea Germinal , Síndromes de Inmunodeficiencia/genética , Fosfatidilinositol 3-Quinasas/genética , Linfocitos T/metabolismo , Antibióticos Antineoplásicos/uso terapéutico , Diferenciación Celular/genética , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Infecciones por Citomegalovirus/sangre , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Infecciones por Virus de Epstein-Barr/sangre , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Femenino , Genes Dominantes , Humanos , Immunoblotting , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Masculino , Linaje , Fosfatidilinositol 3-Quinasas/química , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo , Viremia/tratamiento farmacológico , Viremia/genética , Viremia/virología
14.
PLoS One ; 8(6): e66943, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825096

RESUMEN

The scientific community has been restricted by the lack of a practical and informative animal model of gastrointestinal infection with vegetative Bacillus anthracis. We herein report the development of a murine model of gastrointestinal anthrax infection by gavage of vegetative Sterne strain of Bacillus anthracis into the complement-deficient A/J mouse strain. Mice infected in this manner developed lethal infections in a dose-dependent manner and died 30 h-5 d following gavage. Histological findings were consistent with penetration and growth of the bacilli within the intestinal villi, with subsequent dissemination into major organs including the spleen, liver, kidney and lung. Blood cultures confirmed anthrax bacteremia in all moribund animals, with approximately 1/3 showing co-infection with commensal enteric organisms. However, no evidence of immune activation was observed during infection. Time-course experiments revealed early compromise of the intestinal epithelium, characterized by villus blunting and ulceration in the ileum and jejunum. A decrease in body temperature was most predictive of near-term lethality. Antibiotic treatment of infected animals 24 h following high-dose bacterial gavage protected all animals, demonstrating the utility of this animal model in evaluating potential therapeutics.


Asunto(s)
Carbunco/fisiopatología , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/fisiopatología , Animales , Bacillus anthracis/inmunología , Bacillus anthracis/fisiología , Mucosa Intestinal/microbiología , Ratones , Esporas Bacterianas
15.
PLoS One ; 7(3): e33583, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438953

RESUMEN

A variety of intestinal pathogens have virulence factors that target mitogen activated protein kinase (MAPK) signaling pathways, including Bacillus anthracis. Anthrax lethal toxin (LT) has specific proteolytic activity against the upstream regulators of MAPKs, the MAPK kinases (MKKs). Using a murine model of intoxication, we show that LT causes the dose-dependent disruption of intestinal epithelial integrity, characterized by mucosal erosion, ulceration, and bleeding. This pathology correlates with an LT-dependent blockade of intestinal crypt cell proliferation, accompanied by marked apoptosis in the villus tips. C57BL/6J mice treated with intravenous LT nearly uniformly develop systemic infections with commensal enteric organisms within 72 hours of administration. LT-dependent intestinal pathology depends upon its proteolytic activity and is partially attenuated by co-administration of broad spectrum antibiotics, indicating that it is both a cause and an effect of infection. These findings indicate that targeting of MAPK signaling pathways by anthrax LT compromises the structural integrity of the mucosal layer, serving to undermine the effectiveness of the intestinal barrier. Combined with the well-described immunosuppressive effects of LT, this disruption of the intestinal barrier provides a potential mechanism for host invasion via the enteric route, a common portal of entry during the natural infection cycle of Bacillus anthracis.


Asunto(s)
Antígenos Bacterianos/toxicidad , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/toxicidad , Infecciones por Enterobacteriaceae/etiología , Animales , Carbunco/etiología , Carbunco/patología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/patología , Femenino , Interacciones Huésped-Patógeno , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
16.
Toxins (Basel) ; 3(6): 591-607, 2011 06.
Artículo en Inglés | MEDLINE | ID: mdl-22069727

RESUMEN

The pathological actions of anthrax toxin require the activities of its edema factor (EF) and lethal factor (LF) enzyme components, which gain intracellular access via its receptor-binding component, protective antigen (PA). LF is a metalloproteinase with specificity for selected mitogen-activated protein kinase kinases (MKKs), but its activity is not directly lethal to many types of primary and transformed cells in vitro. Nevertheless, in vivo treatment of several animal species with the combination of LF and PA (termed lethal toxin or LT) leads to morbidity and mortality, suggesting that LT-dependent toxicity is mediated by cellular interactions between host cells. Decades of research have revealed that a central hallmark of this toxicity is the disruption of key cellular barriers required to maintain homeostasis. This review will focus on the current understanding of the effects of LT on barrier function, highlighting recent progress in establishing the molecular mechanisms underlying these effects.


Asunto(s)
Carbunco/microbiología , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Barrera Hematoacuosa/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Epitelio/efectos de los fármacos , Animales , Carbunco/patología , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/farmacocinética , Barrera Hematoacuosa/metabolismo , Barrera Hematoacuosa/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Epitelio/metabolismo , Epitelio/patología , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología
17.
Blood ; 118(10): 2653-5, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21670465

RESUMEN

The syndrome of monocytopenia, B-cell and NK-cell lymphopenia, and mycobacterial, fungal, and viral infections is associated with myelodysplasia, cytogenetic abnormalities, pulmonary alveolar proteinosis, and myeloid leukemias. Both autosomal dominant and sporadic cases occur. We identified 12 distinct mutations in GATA2 affecting 20 patients and relatives with this syndrome, including recurrent missense mutations affecting the zinc finger-2 domain (R398W and T354M), suggesting dominant interference of gene function. Four discrete insertion/deletion mutations leading to frame shifts and premature termination implicate haploinsufficiency as a possible mechanism of action as well. These mutations were found in hematopoietic and somatic tissues, and several were identified in families, indicating germline transmission. Thus, GATA2 joins RUNX1 and CEBPA not only as a familial leukemia gene but also as a cause of a complex congenital immunodeficiency that evolves over decades and combines predisposition to infection and myeloid malignancy.


Asunto(s)
Factor de Transcripción GATA2/genética , Predisposición Genética a la Enfermedad , Monocitos/patología , Mutación/genética , Infecciones por Mycobacterium/etiología , Infecciones por Mycobacterium/patología , Mycobacterium/patogenicidad , Genes Dominantes , Humanos , Síndrome
18.
J Immunol ; 185(9): 5463-7, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20921524

RESUMEN

Neutrophils isolated from BALB/c or C57BL/6 mice and treated in vitro with anthrax lethal toxin release bioactive neutrophil elastase, a proinflammatory mediator of tissue destruction. Similarly, neutrophils isolated from mice treated with anthrax lethal toxin in vivo and cultured ex vivo release greater amounts of elastase than neutrophils from vehicle-treated controls. Direct measurements from murine intestinal tissue samples demonstrate an anthrax lethal toxin-dependent increase in neutrophil elastase activity in vivo as well. These findings correlate with marked lethal toxin-induced intestinal ulceration and bleeding in neutrophil elastase(+/+) animals, but not in neutrophil elastase(-/-) animals. Moreover, neutrophil elastase(-/-) mice have a significant survival advantage over neutrophil elastase(+/+) animals following exposure to anthrax lethal toxin, thereby establishing a key role for neutrophil elastase in mediating the deleterious effects of anthrax lethal toxin.


Asunto(s)
Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Intestinos/enzimología , Intestinos/patología , Neutrófilos/enzimología , Elastasa Pancreática/inmunología , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Intestinos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Elastasa Pancreática/biosíntesis
19.
Blood ; 115(8): 1519-29, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20040766

RESUMEN

We identified 18 patients with the distinct clinical phenotype of susceptibility to disseminated nontuberculous mycobacterial infections, viral infections, especially with human papillomaviruses, and fungal infections, primarily histoplasmosis, and molds. This syndrome typically had its onset in adulthood (age range, 7-60 years; mean, 31.1 years; median, 32 years) and was characterized by profound circulating monocytopenia (mean, 13.3 cells/microL; median, 14.5 cells/microL), B lymphocytopenia (mean, 9.4 cells/microL; median, 4 cells/microL), and NK lymphocytopenia (mean, 16 cells/microL; median, 5.5 cells/microL). T lymphocytes were variably affected. Despite these peripheral cytopenias, all patients had macrophages and plasma cells at sites of inflammation and normal immunoglobulin levels. Ten of these patients developed 1 or more of the following malignancies: 9 myelodysplasia/leukemia, 1 vulvar carcinoma and metastatic melanoma, 1 cervical carcinoma, 1 Bowen disease of the vulva, and 1 multiple Epstein-Barr virus(+) leiomyosarcoma. Five patients developed pulmonary alveolar proteinosis without mutations in the granulocyte-macrophage colony-stimulating factor receptor or anti-granulocyte-macrophage colony-stimulating factor autoantibodies. Among these 18 patients, 5 families had 2 generations affected, suggesting autosomal dominant transmission as well as sporadic cases. This novel clinical syndrome links susceptibility to mycobacterial, viral, and fungal infections with malignancy and can be transmitted in an autosomal dominant pattern.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Predisposición Genética a la Enfermedad/genética , Leucopenia/genética , Infecciones por Mycobacterium/genética , Micosis/genética , Síndromes Mielodisplásicos/genética , Infecciones por Papillomavirus/genética , Linaje , Adolescente , Adulto , Niño , Femenino , Hongos , Enfermedades Genéticas Congénitas/sangre , Enfermedades Genéticas Congénitas/complicaciones , Humanos , Recuento de Leucocitos , Leucopenia/sangre , Leucopenia/complicaciones , Masculino , Persona de Mediana Edad , Mycobacterium , Infecciones por Mycobacterium/sangre , Infecciones por Mycobacterium/etiología , Micosis/sangre , Micosis/etiología , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/etiología , Neoplasias/sangre , Neoplasias/etiología , Neoplasias/genética , Papillomaviridae , Infecciones por Papillomavirus/sangre , Infecciones por Papillomavirus/etiología
20.
J Exp Med ; 205(12): 2803-12, 2008 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19001140

RESUMEN

Tpl2 (Tumor progression locus 2), also known as Cot/MAP3K8, is a hematopoietically expressed serine-threonine kinase. Tpl2 is known to have critical functions in innate immunity in regulating tumor necrosis factor-alpha, Toll-like receptor, and G protein-coupled receptor signaling; however, our understanding of its physiological role in T cells is limited. We investigated the potential roles of Tpl2 in T cells and found that it was induced by interleukin-12 in human and mouse T cells in a Stat4-dependent manner. Deficiency of Tpl2 was associated with impaired interferon (IFN)-gamma production. Accordingly, Tpl2(-/-) mice had impaired host defense against Toxoplasma gondii with reduced parasite clearance and decreased IFN-gamma production. Furthermore, reconstitution of Rag2(-/-) mice with Tpl2-deficient T cells followed by T. gondii infection recapitulated the IFN-gamma defect seen in the Tpl2-deficient mice, confirming a T cell-intrinsic defect. CD4(+) T cells isolated from Tpl2(-/-) mice showed poor induction of T-bet and failure to up-regulate Stat4 protein, which is associated with impaired TCR-dependent extracellular signal-regulated kinase activation. These data underscore the role of Tpl2 as a regulator of T helper cell lineage decisions and demonstrate that Tpl2 has an important functional role in the regulation of Th1 responses.


Asunto(s)
Interferón gamma/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Proteínas Proto-Oncogénicas/inmunología , Linfocitos T/inmunología , Toxoplasma/inmunología , Animales , Linaje de la Célula , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Perfilación de la Expresión Génica , Humanos , Interleucina-12/inmunología , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Análisis por Micromatrices , Proteínas Proto-Oncogénicas/genética , Factor de Transcripción STAT4/inmunología , Proteínas de Dominio T Box/inmunología , Células TH1/citología , Células TH1/inmunología , Toxoplasmosis Animal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA