Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 25(7): 3064-3089, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38866980

RESUMEN

Type I interferons (IFN-I) are implicated in exacerbation of tuberculosis (TB), but the mechanisms are unclear. Mouse macrophages infected with Mycobacterium tuberculosis (Mtb) produce IFN-I, which contributes to their death. Here we investigate whether the same is true for human monocyte-derived macrophages (MDM). MDM prepared by a conventional method markedly upregulate interferon-stimulated genes (ISGs) upon Mtb infection, while MDM prepared to better restrict Mtb do so much less. A mixture of antibodies inhibiting IFN-I signaling prevents ISG induction. Surprisingly, secreted IFN-I are undetectable until nearly two days after ISG induction. These same antibodies do not diminish Mtb-infected MDM death. MDM induce ISGs in response to picogram/mL levels of exogenous IFN-I while depleting similar quantities from the medium. Exogenous IFN-I increase the proportion of dead MDM. We speculate that Mtb-infected MDM produce and respond to minute levels of IFN-I, and that only some of the resultant signaling is susceptible to neutralizing antibodies. Many types of cells may secrete IFN-I in patients with TB, where IFN-I is likely to promote the death of infected macrophages.


Asunto(s)
Muerte Celular , Interferón Tipo I , Macrófagos , Mycobacterium tuberculosis , Humanos , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Macrófagos/microbiología , Macrófagos/metabolismo , Macrófagos/inmunología , Interferón Tipo I/metabolismo , Transducción de Señal , Tuberculosis/microbiología , Tuberculosis/inmunología , Tuberculosis/metabolismo , Animales , Ratones , Células Cultivadas
2.
Eur J Immunol ; 54(3): e2350666, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38161237

RESUMEN

Mycobacterium tuberculosis (Mtb) can cause a latent infection that sometimes progresses to clinically active tuberculosis (TB). Type I interferons (IFN-I) have been implicated in initiating the progression from latency to active TB, in part because IFN-I stimulated genes are the earliest genes to be upregulated in patients as they advance to active TB. Plasmacytoid dendritic cells (pDCs) are major producers of IFN-I during viral infections and in response to autoimmune-induced neutrophil extracellular traps. pDCs have also been suggested to be the major producers of IFN-I during Mtb infection of mice and nonhuman primates, but direct evidence has been lacking. Here, we found that Mtb did not stimulate isolated human pDCs to produce IFN-I, but human neutrophils infected with Mtb-activated co-cultured pDCs to do so. Mtb-infected neutrophils produced neutrophil extracellular traps, whose exposed DNA is a well-known mechanism to activate pDCs to secrete IFN-I. We conclude that pDCs contribute to the IFN-I response during Mtb infection by interacting with infected neutrophils which may then promote Mtb pathogenesis.


Asunto(s)
Interferón Tipo I , Mycobacterium tuberculosis , Tuberculosis , Animales , Humanos , Neutrófilos/metabolismo , Interferón Tipo I/metabolismo , Células Dendríticas/metabolismo
3.
J Immunol ; 206(7): 1631-1641, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33674446

RESUMEN

Inhibitors of the immunoproteasome (i-20S) have shown promise in mouse models of autoimmune diseases and allograft rejection. In this study, we used a novel inhibitor of the immunoproteasome, PKS3053, that is reversible, noncovalent, tight-binding, and highly selective for the ß5i subunit of the i-20S to evaluate the role that i-20S plays in regulating immune responses in vitro and in vivo. In contrast to irreversible, less-selective inhibitors, PKS3053 did not kill any of the primary human cell types tested, including plasmacytoid dendritic cells, conventional dendritic cells, macrophages, and T cells, all of which expressed genes encoding both the constitutive proteasome (c-20S) and i-20S. PKS3053 reduced TLR-dependent activation of plasmacytoid dendritic cells, decreasing their maturation and IFN-α response and reducing their ability to activate allogenic T cells. In addition, PKS3053 reduced T cell proliferation directly and inhibited TLR-mediated activation of conventional dendritic cells and macrophages. In a mouse model of skin injury that shares some features of cutaneous lupus erythematosus, blocking i-20S decreased inflammation, cellular infiltration, and tissue damage. We conclude that the immunoproteasome is involved in the activation of innate and adaptive immune cells, that their activation can be suppressed with an i-20S inhibitor without killing them, and that selective inhibition of ß5i holds promise as a potential therapy for inflammatory skin diseases such as psoriasis, cutaneous lupus erythematosus, and systemic sclerosis.


Asunto(s)
Células Dendríticas/inmunología , Inflamación/tratamiento farmacológico , Lupus Eritematoso Cutáneo/tratamiento farmacológico , Macrófagos/inmunología , Inhibidores de Proteasoma/uso terapéutico , Piel/patología , Linfocitos T/inmunología , Animales , Movimiento Celular , Células Cultivadas , Citotoxicidad Inmunológica , Células Dendríticas/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Linfocitos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 115(41): E9560-E9569, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30257943

RESUMEN

The protein disaggregase ClpB hexamer is conserved across evolution and has two AAA+-type nucleotide-binding domains, NBD1 and NBD2, in each protomer. In M. tuberculosis (Mtb), ClpB facilitates asymmetric distribution of protein aggregates during cell division to help the pathogen survive and persist within the host, but a mechanistic understanding has been lacking. Here we report cryo-EM structures at 3.8- to 3.9-Šresolution of Mtb ClpB bound to a model substrate, casein, in the presence of the weakly hydrolyzable ATP mimic adenosine 5'-[γ-thio]triphosphate. Mtb ClpB existed in solution in two closed-ring conformations, conformers 1 and 2. In both conformers, the 12 pore-loops on the 12 NTDs of the six protomers (P1-P6) were arranged similarly to a staircase around the bound peptide. Conformer 1 is a low-affinity state in which three of the 12 pore-loops (the protomer P1 NBD1 and NBD2 loops and the protomer P2 NBD1 loop) are not engaged with peptide. Conformer 2 is a high-affinity state because only one pore-loop (the protomer P2 NBD1 loop) is not engaged with the peptide. The resolution of the two conformations, along with their bound substrate peptides and nucleotides, enabled us to propose a nucleotide-driven peptide translocation mechanism of a bacterial ClpB that is largely consistent with several recent unfoldase structures, in particular with the eukaryotic Hsp104. However, whereas Hsp104's two NBDs move in opposing directions during one step of peptide translocation, in Mtb ClpB the two NBDs move only in the direction of translocation.


Asunto(s)
Adenosina Trifosfato/química , Proteínas Bacterianas/química , Endopeptidasa Clp/química , Mycobacterium tuberculosis/enzimología , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/metabolismo , Endopeptidasa Clp/metabolismo , Hidrólisis , Dominios Proteicos , Transporte de Proteínas
5.
Proc Natl Acad Sci U S A ; 115(29): E6863-E6870, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29967165

RESUMEN

We describe noncovalent, reversible asparagine ethylenediamine (AsnEDA) inhibitors of the Plasmodium falciparum proteasome (Pf20S) ß5 subunit that spare all active subunits of human constitutive and immuno-proteasomes. The compounds are active against erythrocytic, sexual, and liver-stage parasites, against parasites resistant to current antimalarials, and against P. falciparum strains from patients in Africa. The ß5 inhibitors synergize with a ß2 inhibitor in vitro and in mice and with artemisinin. P. falciparum selected for resistance to an AsnEDA ß5 inhibitor surprisingly harbored a point mutation in the noncatalytic ß6 subunit. The ß6 mutant was resistant to the species-selective Pf20S ß5 inhibitor but remained sensitive to the species-nonselective ß5 inhibitors bortezomib and carfilzomib. Moreover, resistance to the Pf20S ß5 inhibitor was accompanied by increased sensitivity to a Pf20S ß2 inhibitor. Finally, the ß5 inhibitor-resistant mutant had a fitness cost that was exacerbated by irradiation. Thus, used in combination, multistage-active inhibitors of the Pf20S ß5 and ß2 subunits afford synergistic antimalarial activity with a potential to delay the emergence of resistance to artemisinins and each other.


Asunto(s)
Antimaláricos/química , Plasmodium falciparum/enzimología , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Proteínas Protozoarias/antagonistas & inhibidores , Artemisininas/química , Bortezomib/química , Farmacorresistencia Microbiana , Humanos , Lactonas/química , Oligopéptidos/química , Proteínas Protozoarias/química
6.
Angew Chem Int Ed Engl ; 60(17): 9279-9283, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33433953

RESUMEN

Plasmodium falciparum proteasome (Pf20S) inhibitors are active against Plasmodium at multiple stages-erythrocytic, gametocyte, liver, and gamete activation stages-indicating that selective Pf20S inhibitors possess the potential to be therapeutic, prophylactic, and transmission-blocking antimalarials. Starting from a reported compound, we developed a noncovalent, macrocyclic peptide inhibitor of the malarial proteasome with high species selectivity and improved pharmacokinetic properties. The compound demonstrates specific, time-dependent inhibition of the ß5 subunit of the Pf20S, kills artemisinin-sensitive and artemisinin-resistant P. falciparum isolates in vitro and reduces parasitemia in humanized, P. falciparum-infected mice.


Asunto(s)
Antimaláricos/farmacología , Desarrollo de Medicamentos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Animales , Antimaláricos/síntesis química , Antimaláricos/química , Malaria Falciparum/metabolismo , Ratones , Modelos Moleculares , Conformación Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/enzimología , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/química
7.
Proc Natl Acad Sci U S A ; 114(24): E4832-E4840, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28559332

RESUMEN

Mycobacterium tuberculosis (Mtb) encounters stresses during the pathogenesis and treatment of tuberculosis (TB) that can suppress replication of the bacteria and render them phenotypically tolerant to most available drugs. Where studied, the majority of Mtb in the sputum of most untreated subjects with active TB have been found to be nonreplicating by the criterion that they do not grow as colony-forming units (cfus) when plated on agar. However, these cells are viable because they grow when diluted in liquid media. A method for generating such "differentially detectable" (DD) Mtb in vitro would aid studies of the biology and drug susceptibility of this population, but lack of independent confirmation of reported methods has contributed to skepticism about their existence. Here, we identified confounding artifacts that, when avoided, allowed development of a reliable method of producing cultures of ≥90% DD Mtb in starved cells. We then characterized several drugs according to whether they contribute to the generation of DD Mtb or kill them. Of the agents tested, rifamycins led to DD Mtb generation, an effect lacking in a rifampin-resistant strain with a mutation in rpoB, which encodes the canonical rifampin target, the ß subunit of RNA polymerase. In contrast, thioridazine did not generate DD Mtb from starved cells but killed those generated by rifampin.


Asunto(s)
Antibióticos Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Rifamicinas/farmacología , Antituberculosos/farmacología , Proteínas Bacterianas/genética , ARN Polimerasas Dirigidas por ADN/genética , Farmacorresistencia Bacteriana/genética , Humanos , Isoniazida/farmacología , Mutación , Mycobacterium tuberculosis/genética , Tioridazina/farmacología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología
8.
Eur J Immunol ; 48(4): 612-620, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436711

RESUMEN

Genetic deficiency of protein kinase R (PKR) in mice was reported to enhance macrophage activation in vitro in response to interferon-γ (IFNγ) and to reduce the burden of Mycobacterium tuberculosis (Mtb) in vivo (Wu et al. PloS One. 2012 7:e30512). Consistent with this, treatment of wild-type (WT) macrophages in vitro with a novel PKR inhibitor (Bryk et al., Bioorg. Med. Chem. Lett. 2011 21:4108-4114) also enhanced IFN-γ-dependent macrophage activation (Wu et al. PloS One. 2012 7:e30512). Here we show that co-treatment with IFN-γ and a new PKR inhibitor identified herein to be highly but not completely selective likewise induced macrophages to produce more reactive nitrogen intermediates (RNI) and tumor necrosis factor alpha (TNF-α) and less interleukin 10 (IL-10) than seen with IFN-γ alone. Unexpectedly, however, this new PKR inhibitor had a comparable effect on PKR-deficient macrophages. Retrospective investigation revealed that the PKR-deficient mice in (Wu et al. PloS One. 2012 7:e30512) had not been backcrossed. On comparing genetically matched PKR-deficient and WT mice, we saw no impact of PKR deficiency on macrophage activation in vitro or during the course of Mtb infection in vivo. In addition, although 129S1/SvImJ macrophage responses to IFN-γ were greater than those of C57BL/6J macrophages, PKR was not required to mediate the IFN-γ-dependent production of IL-10, RNI or TNF-α in either strain. Together the data cast doubt on PKR as a potential therapeutic target for tuberculosis.


Asunto(s)
Interferón gamma/farmacología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Células Cultivadas , Femenino , Interleucina-10/biosíntesis , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies de Nitrógeno Reactivo/biosíntesis , Tuberculosis/microbiología , Factor de Necrosis Tumoral alfa/biosíntesis , eIF-2 Quinasa/genética
9.
Proc Natl Acad Sci U S A ; 113(49): E7947-E7956, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27872278

RESUMEN

During host infection, Mycobacterium tuberculosis (Mtb) encounters several types of stress that impair protein integrity, including reactive oxygen and nitrogen species and chemotherapy. The resulting protein aggregates can be resolved or degraded by molecular machinery conserved from bacteria to eukaryotes. Eukaryotic Hsp104/Hsp70 and their bacterial homologs ClpB/DnaK are ATP-powered chaperones that restore toxic protein aggregates to a native folded state. DnaK is essential in Mycobacterium smegmatis, and ClpB is involved in asymmetrically distributing damaged proteins during cell division as a mechanism of survival in Mtb, commending both proteins as potential drug targets. However, their molecular partners in protein reactivation have not been characterized in mycobacteria. Here, we reconstituted the activities of the Mtb ClpB/DnaK bichaperone system with the cofactors DnaJ1, DnaJ2, and GrpE and the small heat shock protein Hsp20. We found that DnaJ1 and DnaJ2 activate the ATPase activity of DnaK differently. A point mutation in the highly conserved HPD motif of the DnaJ proteins abrogates their ability to activate DnaK, although the DnaJ2 mutant still binds to DnaK. The purified Mtb ClpB/DnaK system reactivated a heat-denatured model substrate, but the DnaJ HPD mutants inhibited the reaction. Finally, either DnaJ1 or DnaJ2 is required for mycobacterial viability, as is the DnaK-activating activity of a DnaJ protein. These studies lay the groundwork for strategies to target essential chaperone-protein interactions in Mtb, the leading cause of death from a bacterial infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chaperonas Moleculares/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteostasis , Adenosina Trifosfatasas/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/metabolismo , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium smegmatis/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(52): E8425-E8432, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27956634

RESUMEN

Constitutive proteasomes (c-20S) are ubiquitously expressed cellular proteases that degrade polyubiquitinated proteins and regulate cell functions. An isoform of proteasome, the immunoproteasome (i-20S), is highly expressed in human T cells, dendritic cells (DCs), and B cells, suggesting that it could be a potential target for inflammatory diseases, including those involving autoimmunity and alloimmunity. Here, we describe DPLG3, a rationally designed, noncovalent inhibitor of the immunoproteasome chymotryptic subunit ß5i that has thousands-fold selectivity over constitutive ß5c. DPLG3 suppressed cytokine release from blood mononuclear cells and the activation of DCs and T cells, diminished accumulation of effector T cells, promoted expression of exhaustion and coinhibitory markers on T cells, and synergized with CTLA4-Ig to promote long-term acceptance of cardiac allografts across a major histocompatibility barrier. These findings demonstrate the potential value of using brief posttransplant immunoproteasome inhibition to entrain a long-term response favorable to allograft survival as part of an immunomodulatory regimen that is neither broadly immunosuppressive nor toxic.


Asunto(s)
Supervivencia de Injerto , Trasplante de Corazón/métodos , Inmunosupresores/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Citocinas/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Hep G2 , Humanos , Memoria Inmunológica , Leucocitos Mononucleares/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología
11.
Proc Natl Acad Sci U S A ; 113(31): E4523-30, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27432954

RESUMEN

The rising incidence of antimicrobial resistance (AMR) makes it imperative to understand the underlying mechanisms. Mycobacterium tuberculosis (Mtb) is the single leading cause of death from a bacterial pathogen and estimated to be the leading cause of death from AMR. A pyrido-benzimidazole, 14, was reported to have potent bactericidal activity against Mtb. Here, we isolated multiple Mtb clones resistant to 14. Each had mutations in the putative DNA-binding and dimerization domains of rv2887, a gene encoding a transcriptional repressor of the MarR family. The mutations in Rv2887 led to markedly increased expression of rv0560c. We characterized Rv0560c as an S-adenosyl-L-methionine-dependent methyltransferase that N-methylates 14, abolishing its mycobactericidal activity. An Mtb strain lacking rv0560c became resistant to 14 by mutating decaprenylphosphoryl-ß-d-ribose 2-oxidase (DprE1), an essential enzyme in arabinogalactan synthesis; 14 proved to be a nanomolar inhibitor of DprE1, and methylation of 14 by Rv0560c abrogated this activity. Thus, 14 joins a growing list of DprE1 inhibitors that are potently mycobactericidal. Bacterial methylation of an antibacterial agent, 14, catalyzed by Rv0560c of Mtb, is a previously unreported mechanism of AMR.


Asunto(s)
Antituberculosos/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Mycobacterium tuberculosis/metabolismo , Antituberculosos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bencimidazoles/química , Bencimidazoles/metabolismo , Regulación Bacteriana de la Expresión Génica , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Metiltransferasas/metabolismo , Modelos Moleculares , Estructura Molecular , Mutación , Mycobacterium tuberculosis/genética , Dominios Proteicos , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , S-Adenosilmetionina/metabolismo
12.
Proc Natl Acad Sci U S A ; 112(43): E5834-43, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26430237

RESUMEN

Enzymes of central carbon metabolism (CCM) in Mycobacterium tuberculosis (Mtb) make an important contribution to the pathogen's virulence. Evidence is emerging that some of these enzymes are not simply playing the metabolic roles for which they are annotated, but can protect the pathogen via additional functions. Here, we found that deficiency of 2-hydroxy-3-oxoadipate synthase (HOAS), the E1 component of the α-ketoglutarate (α-KG) dehydrogenase complex (KDHC), did not lead to general metabolic perturbation or growth impairment of Mtb, but only to the specific inability to cope with glutamate anaplerosis and nitroxidative stress. In the former role, HOAS acts to prevent accumulation of aldehydes, including growth-inhibitory succinate semialdehyde (SSA). In the latter role, HOAS can participate in an alternative four-component peroxidase system, HOAS/dihydrolipoyl acetyl transferase (DlaT)/alkylhydroperoxide reductase colorless subunit gene (ahpC)-neighboring subunit (AhpD)/AhpC, using α-KG as a previously undescribed source of electrons for reductase action. Thus, instead of a canonical role in CCM, the E1 component of Mtb's KDHC serves key roles in situational defense that contribute to its requirement for virulence in the host. We also show that pyruvate decarboxylase (AceE), the E1 component of pyruvate dehydrogenase (PDHC), can participate in AceE/DlaT/AhpD/AhpC, using pyruvate as a source of electrons for reductase action. Identification of these systems leads us to suggest that Mtb can recruit components of its CCM for reactive nitrogen defense using central carbon metabolites.


Asunto(s)
Ácido Glutámico/metabolismo , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Mycobacterium tuberculosis/metabolismo , Nitrosación , Estrés Oxidativo , Animales , Ratones , Ratones Endogámicos C57BL
13.
Proc Natl Acad Sci U S A ; 110(45): E4256-65, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24145454

RESUMEN

In high enough concentrations, such as produced by inducible nitric oxide synthase (iNOS), reactive nitrogen species (RNS) can kill Mycobacterium tuberculosis (Mtb). Lesional macrophages in macaques and humans with tuberculosis express iNOS, and mice need iNOS to avoid succumbing rapidly to tuberculosis. However, Mtb's own ability to produce RNS is rarely considered, perhaps because nitrate reduction to nitrite is only prominent in axenic Mtb cultures at oxygen tensions ≤1%. Here we found that cultures of Mtb-infected human macrophages cultured at physiologic oxygen tensions produced copious nitrite. Surprisingly, the nitrite arose from the Mtb, not the macrophages. Mtb responded to nitrite by ceasing growth; elevating levels of ATP through reduced consumption; and altering the expression of 120 genes associated with adaptation to acid, hypoxia, nitric oxide, oxidative stress, and iron deprivation. The transcriptomic effect of endogenous nitrite was distinct from that of nitric oxide. Thus, whether or not Mtb is hypoxic, the host expresses iNOS, or hypoxia impairs the action of iNOS, Mtb in vivo is likely to encounter RNS by producing nitrite. Endogenous nitrite may slow Mtb's growth and prepare it to resist host stresses while the pathogen waits for immunopathology to promote its transmission.


Asunto(s)
Adenosina Trifosfato/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Macrófagos/microbiología , Mycobacterium tuberculosis/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Oxígeno/metabolismo , Secuencia de Bases , Perfilación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Mycobacterium tuberculosis/crecimiento & desarrollo , Nitroimidazoles , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
14.
Proc Natl Acad Sci U S A ; 110(49): E4790-7, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24248369

RESUMEN

Mycobacterium tuberculosis (Mtb) restrains immune responses well enough to escape eradication but elicits enough immunopathology to ensure its transmission. Here we provide evidence that this host-pathogen relationship is regulated in part by a cytosolic, membrane-associated protein with a unique structural fold, encoded by the Mtb gene rv0431. The protein acts by regulating the quantity of Mtb-derived membrane vesicles bearing Toll-like receptor 2 ligands, including the lipoproteins LpqH and SodC. We propose that rv0431 be named "vesiculogenesis and immune response regulator."


Asunto(s)
Proteínas Bacterianas/química , Inmunomodulación/fisiología , Lipoproteínas/metabolismo , Proteínas de la Membrana/química , Modelos Moleculares , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Vesículas Transportadoras/fisiología , Animales , Proteínas Bacterianas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Inmunomodulación/genética , Macrófagos , Espectroscopía de Resonancia Magnética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Rastreo , Pliegue de Proteína , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/genética , Vesículas Transportadoras/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(40): 16004-11, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23012453

RESUMEN

Existing drugs are slow to eradicate Mycobacterium tuberculosis (Mtb) in patients and have failed to control tuberculosis globally. One reason may be that host conditions impair Mtb's replication, reducing its sensitivity to most antiinfectives. We devised a high-throughput screen for compounds that kill Mtb when its replication has been halted by reactive nitrogen intermediates (RNIs), acid, hypoxia, and a fatty acid carbon source. At concentrations routinely achieved in human blood, oxyphenbutazone (OPB), an inexpensive anti-inflammatory drug, was selectively mycobactericidal to nonreplicating (NR) Mtb. Its cidal activity depended on mild acid and was augmented by RNIs and fatty acid. Acid and RNIs fostered OPB's 4-hydroxylation. The resultant 4-butyl-4-hydroxy-1-(4-hydroxyphenyl)-2-phenylpyrazolidine-3,5-dione (4-OH-OPB) killed both replicating and NR Mtb, including Mtb resistant to standard drugs. 4-OH-OPB depleted flavins and formed covalent adducts with N-acetyl-cysteine and mycothiol. 4-OH-OPB killed Mtb synergistically with oxidants and several antituberculosis drugs. Thus, conditions that block Mtb's replication modify OPB and enhance its cidal action. Modified OPB kills both replicating and NR Mtb and sensitizes both to host-derived and medicinal antimycobacterial agents.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Oxifenilbutazona/farmacología , Animales , Cromatografía Líquida de Alta Presión , Farmacorresistencia Microbiana/fisiología , Ácidos Grasos/metabolismo , Femenino , Hidroxilación , Espectroscopía de Resonancia Magnética , Ratones , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/fisiología , Oxifenilbutazona/metabolismo , Oxifenilbutazona/farmacocinética , Especies de Nitrógeno Reactivo/metabolismo
16.
J Biol Chem ; 288(30): 21688-702, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23760263

RESUMEN

Allosteric regulation often controls key branch points in metabolic processes. Mycobacterium tuberculosis 2-hydroxy-3-oxoadipate synthase (HOAS), a thiamin diphosphate (ThDP)-dependent enzyme, produces 2-hydroxy-3-oxoadipate using 2-ketoglutarate and glyoxylate. The proposed chemical mechanism in analogy with other ThDP-dependent carboligases involves multiple ThDP-bound covalent intermediates. Acetyl coenzyme A is an activator, and GarA, a forkhead association domain-containing protein known to regulate glutamate metabolism, is an allosteric inhibitor of HOAS. Steady state kinetics using assays to study the first half and the full catalytic cycle suggested that the regulators act at different steps in the overall mechanism. To explore the modes of regulation and to test the effects on individual catalytic steps, we performed circular dichroism (CD) studies using a non-decarboxylatable 2-ketoglutarate analog and determined the distribution of ThDP-bound covalent intermediates during the steady state of the HOAS reaction using one-dimensional (1)H gradient carbon heteronuclear single quantum coherence NMR. The results suggest that acetyl coenzyme A acts as a mixed V and K type activator and predominantly affects the predecarboxylation steps. GarA does not inhibit the formation of the predecarboxylation analog and does not affect the accumulation of the postdecarboxylation covalent intermediate derived from 2-ketoglutarate; however, it decreases the abundance of the product ThDP adduct in the HOAS pathway. Thus, the two regulators act on different halves of the catalytic cycle in an unusual regulatory regime.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Oxo-Ácido-Liasas/metabolismo , Tiamina Pirofosfato/metabolismo , Acetilcoenzima A/metabolismo , Adipatos/química , Adipatos/metabolismo , Transferasas de Aldehído-Cetona , Algoritmos , Regulación Alostérica , Proteínas Bacterianas/genética , Biocatálisis , Dicroismo Circular , Clonación Molecular , Inhibidores Enzimáticos/metabolismo , Glioxilatos/química , Glioxilatos/metabolismo , Concentración de Iones de Hidrógeno , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Cinética , Espectroscopía de Resonancia Magnética , Modelos Químicos , Estructura Molecular , Mycobacterium tuberculosis/genética , Oxo-Ácido-Liasas/genética , Especificidad por Sustrato , Tiamina Pirofosfato/química
18.
Science ; 384(6693): eadl2016, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38635718

RESUMEN

Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.


Asunto(s)
Enfermedades Transmisibles , Interferón gamma , Humanos , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/terapia , Interferón gamma/genética , Interferón gamma/metabolismo , Interferón gamma/uso terapéutico , Inmunidad/genética
19.
Antimicrob Agents Chemother ; 57(6): 2834-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23507275

RESUMEN

Nitazoxanide (NTZ) has bactericidal activity against the H37Rv laboratory strain of Mycobacterium tuberculosis with a MIC of 16 µg/ml. However, its efficacy against clinical isolates of M. tuberculosis has not been determined. We found that NTZ's MIC against 50 clinical isolates ranged from 12 to 28 µg/ml with a median of 16 µg/ml and was unaffected by resistance to first- or second-line antituberculosis drugs or a diversity of spoligotypes.


Asunto(s)
Antituberculosos/farmacología , Pruebas de Sensibilidad Microbiana/normas , Mycobacterium tuberculosis/efectos de los fármacos , Tiazoles/farmacología , Farmacorresistencia Bacteriana , Humanos , Isoniazida/farmacología , Mycobacterium tuberculosis/clasificación , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/aislamiento & purificación , Nitrocompuestos , Esputo/microbiología , Tuberculosis Pulmonar/microbiología
20.
ACS Med Chem Lett ; 14(7): 970-976, 2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37465309

RESUMEN

4'-Phosphopantetheinyl transferase (PptT) is an essential enzyme for Mycobacterium tuberculosis (Mtb) survival and virulence and therefore an attractive target for a tuberculosis therapeutic. In this work, two modeling-informed approaches toward the isosteric replacement of the amidinourea moiety present in the previously reported PptT inhibitor AU 8918 are reported. Although a designed 3,5-diamino imidazole unexpectedly adopted an undesired tautomeric form and was inactive, replacement of the amidinourea moiety afforded a series of active PptT inhibitors containing 2,6-diaminopyridine scaffolds.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA