Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
EMBO Rep ; 25(7): 3064-3089, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38866980

RESUMO

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.


Assuntos
Morte Celular , Interferon Tipo I , Macrófagos , Mycobacterium tuberculosis , Humanos , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Macrófagos/microbiologia , Macrófagos/metabolismo , Macrófagos/imunologia , Interferon Tipo I/metabolismo , Transdução de Sinais , Tuberculose/microbiologia , Tuberculose/imunologia , Tuberculose/metabolismo , Animais , Camundongos , Células Cultivadas
2.
J Med Chem ; 65(13): 9350-9375, 2022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-35727231

RESUMO

With over 200 million cases and close to half a million deaths each year, malaria is a threat to global health, particularly in developing countries. Plasmodium falciparum, the parasite that causes the most severe form of the disease, has developed resistance to all antimalarial drugs. Resistance to the first-line antimalarial artemisinin and to artemisinin combination therapies is widespread in Southeast Asia and is emerging in sub-Saharan Africa. The P. falciparum proteasome is an attractive antimalarial target because its inhibition kills the parasite at multiple stages of its life cycle and restores artemisinin sensitivity in parasites that have become resistant through mutation in Kelch K13. Here, we detail our efforts to develop noncovalent, macrocyclic peptide malaria proteasome inhibitors, guided by structural analysis and pharmacokinetic properties, leading to a potent, species-selective, metabolically stable inhibitor.


Assuntos
Antimaláricos , Artemisininas , Malária Falciparum , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Artemisininas/farmacologia , Resistência a Medicamentos , Humanos , Malária Falciparum/tratamento farmacológico , Peptídeos/uso terapêutico , Plasmodium falciparum , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Proteínas de Protozoários/genética
3.
J Immunol ; 206(7): 1631-1641, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33674446

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Inflamação/tratamento farmacológico , Lúpus Eritematoso Cutâneo/tratamento farmacológico , Macrófagos/imunologia , Inibidores de Proteassoma/uso terapêutico , Pele/patologia , Linfócitos T/imunologia , Animais , Movimento Celular , Células Cultivadas , Citotoxicidade Imunológica , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Ativação Linfocitária , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T/efeitos dos fármacos
4.
J Exp Med ; 218(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33125053

RESUMO

Macrophages help defend the host against Mycobacterium tuberculosis (Mtb), the major cause of tuberculosis (TB). Once phagocytized, Mtb resists killing by macrophages, replicates inside them, and leads to their death, releasing Mtb that can infect other cells. We found that the death of Mtb-infected mouse macrophages in vitro does not appear to proceed by a currently known pathway. Through genome-wide CRISPR-Cas9 screening, we identified a critical role for autocrine or paracrine signaling by macrophage-derived type I IFNs in the death of Mtb-infected macrophages in vitro, and blockade of type I IFN signaling augmented the effect of rifampin, a first-line TB drug, in Mtb-infected mice. Further definition of the pathway of type I IFN-mediated macrophage death may allow for host-directed therapy of TB that is more selective than systemic blockade of type I IFN signaling.


Assuntos
Morte Celular/fisiologia , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Transdução de Sinais/fisiologia , Tuberculose/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/fisiologia , Sistemas CRISPR-Cas/efeitos dos fármacos , Sistemas CRISPR-Cas/fisiologia , Morte Celular/efeitos dos fármacos , Linhagem Celular , Células HEK293 , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/fisiologia , Células RAW 264.7 , Rifampina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
5.
J Med Chem ; 63(21): 13103-13123, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33095579

RESUMO

The immunoproteasome (i-20S) has emerged as a therapeutic target for autoimmune and inflammatory disorders and hematological malignancies. Inhibition of the chymotryptic ß5i subunit of i-20S inhibits T cell activation, B cell proliferation, and dendritic cell differentiation in vitro and suppresses immune responses in animal models of autoimmune disorders and allograft rejection. However, cytotoxicity to immune cells has accompanied the use of covalently reactive ß5i inhibitors, whose activity against the constitutive proteasome (c-20S) is cumulative with the time of exposure. Herein, we report a structure-activity relationship study of a class of noncovalent proteasome inhibitors with picomolar potencies and 1000-fold selectivity for i-20S over c-20S. Furthermore, these inhibitors are specific for ß5i over the other five active subunits of i-20S and c-20S, providing useful tools to study the functions of ß5i in immune responses. The potency of these compounds in inhibiting human T cell activation suggests that they may have therapeutic potential.


Assuntos
Dipeptídeos/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química , Sítios de Ligação , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/metabolismo , Dipeptídeos/farmacologia , Células HeLa , Humanos , Concentração Inibidora 50 , Cinética , Ativação Linfocitária/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ligação Proteica , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Relação Estrutura-Atividade , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
6.
J Exp Med ; 216(8): 1725, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31262843
7.
Proc Natl Acad Sci U S A ; 115(41): E9560-E9569, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30257943

RESUMO

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.


Assuntos
Trifosfato de Adenosina/química , Proteínas de Bactérias/química , Endopeptidase Clp/química , Mycobacterium tuberculosis/enzimologia , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Endopeptidase Clp/metabolismo , Hidrólise , Domínios Proteicos , Transporte Proteico
8.
Eur J Immunol ; 48(4): 612-620, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436711

RESUMO

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.


Assuntos
Interferon gama/farmacologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , eIF-2 Quinase/antagonistas & inibidores , Animais , Células Cultivadas , Feminino , Interleucina-10/biossíntese , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Nitrogênio/biossíntese , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , eIF-2 Quinase/genética
9.
ACS Infect Dis ; 4(5): 771-787, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29465985

RESUMO

The success of Mycobacterium tuberculosis (Mtb) as a pathogen depends on the redundant and complex mechanisms it has evolved for resisting nitrosative and oxidative stresses inflicted by host immunity. Improving our understanding of these defense pathways can reveal vulnerable points in Mtb pathogenesis. In this study, we combined genetic, structural, computational, biochemical, and biophysical approaches to identify a novel enzyme class represented by Rv2466c. We show that Rv2466c is a mycothiol-dependent nitroreductase of Mtb and can reduce the nitro group of a novel mycobactericidal compound using mycothiol as a cofactor. In addition to its function as a nitroreductase, Rv2466c confers partial protection to menadione stress.


Assuntos
Cisteína/metabolismo , Glicopeptídeos/metabolismo , Inositol/metabolismo , Mycobacterium tuberculosis/enzimologia , Nitrorredutases/genética , Nitrorredutases/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cisteína/química , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Glicopeptídeos/química , Inositol/química , Camundongos , Modelos Moleculares , Mutação , Mycobacterium tuberculosis/classificação , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Nitrorredutases/química , Oxirredução , Estresse Oxidativo , Filogenia , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Tuberculose/microbiologia
10.
Nat Commun ; 8(1): 1692, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29167449

RESUMO

Proteasome inhibitors benefit patients with multiple myeloma and B cell-dependent autoimmune disorders but exert toxicity from inhibition of proteasomes in other cells. Toxicity should be minimized by reversible inhibition of the immunoproteasome ß5i subunit while sparing the constitutive ß5c subunit. Here we report ß5i-selective inhibition by asparagine-ethylenediamine (AsnEDA)-based compounds and present the high-resolution cryo-EM structural analysis of the human immunoproteasome. Despite inhibiting noncompetitively, an AsnEDA inhibitor binds the active site. Hydrophobic interactions are accompanied by hydrogen bonding with ß5i and ß6 subunits. The inhibitors are far more cytotoxic for myeloma and lymphoma cell lines than for hepatocarcinoma or non-activated lymphocytes. They block human B-cell proliferation and promote apoptotic cell death selectively in antibody-secreting B cells, and to a lesser extent in activated human T cells. Reversible, ß5i-selective inhibitors may be useful for treatment of diseases involving activated or neoplastic B cells or activated T cells.


Assuntos
Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/imunologia , Inibidores de Proteassoma/química , Asparagina/análogos & derivados , Asparagina/química , Asparagina/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Microscopia Crioeletrônica , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Linfócitos/metabolismo , Modelos Moleculares , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/metabolismo , Subunidades Proteicas , Eletricidade Estática , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Proc Natl Acad Sci U S A ; 113(52): E8425-E8432, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27956634

RESUMO

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.


Assuntos
Sobrevivência de Enxerto , Transplante de Coração/métodos , Imunossupressores/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Hep G2 , Humanos , Memória Imunológica , Leucócitos Mononucleares/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
12.
Proc Natl Acad Sci U S A ; 113(49): E7947-E7956, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27872278

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Chaperonas Moleculares/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteostase , Adenosina Trifosfatases/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/metabolismo , Mycobacterium smegmatis/crescimento & desenvolvimento , Mycobacterium smegmatis/metabolismo
13.
Proc Natl Acad Sci U S A ; 113(31): E4523-30, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27432954

RESUMO

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.


Assuntos
Antituberculosos/metabolismo , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana , Mycobacterium tuberculosis/metabolismo , Antituberculosos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Benzimidazóis/química , Benzimidazóis/metabolismo , Regulação Bacteriana da Expressão Gênica , Metilação , Metiltransferases/química , Metiltransferases/genética , Metiltransferases/metabolismo , Modelos Moleculares , Estrutura Molecular , Mutação , Mycobacterium tuberculosis/genética , Domínios Proteicos , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , S-Adenosilmetionina/metabolismo
14.
ACS Med Chem Lett ; 5(4): 405-10, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24900849

RESUMO

Selective inhibitors for the human immunoproteasome LMP7 (ß5i) subunit over the constitutive proteasome hold promise for the treatment of autoimmune and inflammatory diseases and hematologic malignancies. Here we report that oxathiazolones inhibit the immunoproteasome ß5i with up to 4700-fold selectivity over the constitutive proteasome, are cell permeable, and inhibit proteasomes inside cells.

15.
Eur J Med Chem ; 75: 336-53, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24556148

RESUMO

Tuberculosis remains one of the deadliest infectious diseases, killing 1.4 million people annually and showing a rapid increase in cases resistant to multiple drugs. New antibiotics against tuberculosis are urgently needed. Here we describe the design, synthesis and structure-activity relationships of a series of benzimidazole-based compounds with activity against Mycobacterium tuberculosis (Mtb) in a replicating state, a physiologically-induced non-replicating state, or both. Compounds 49, 67, 68, 69, 70, and 72, which shared a 5-nitrofuranyl moiety, exhibited high potency and acceptable selectivity indices (SI). As illustrated by compound 70 (MIC90 < 0.049 µg/mL, SI > 512), the 5-nitrofuranyl group was compatible with minimal cytotoxicity and good intra-macrophage killing, although it lacked non-replicating activity when assessed by CFU assays. Compound 70 had low mutagenic potential by SOS Chromotest assay, making this class of compounds good candidates for further evaluation and target identification.


Assuntos
Antituberculosos/química , Antituberculosos/farmacologia , Benzimidazóis/química , Benzimidazóis/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Antituberculosos/síntese química , Benzimidazóis/síntese química , Células Cultivadas , Desenho de Fármacos , Humanos , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Relação Estrutura-Atividade , Tuberculose/tratamento farmacológico
16.
Proc Natl Acad Sci U S A ; 110(49): E4790-7, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24248369

RESUMO

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."


Assuntos
Proteínas de Bactérias/química , Imunomodulação/fisiologia , Lipoproteínas/metabolismo , Proteínas de Membrana/química , Modelos Moleculares , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Vesículas Transportadoras/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Feminino , Interações Hospedeiro-Patógeno , Imunomodulação/genética , Macrófagos , Espectroscopia de Ressonância Magnética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Dobramento de Proteína , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Vesículas Transportadoras/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(45): E4256-65, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24145454

RESUMO

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.


Assuntos
Trifosfato de Adenosina/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Oxigênio/metabolismo , Sequência de Bases , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Mycobacterium tuberculosis/crescimento & desenvolvimento , Nitroimidazóis , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA
18.
Microbiologyopen ; 2(6): 901-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24019302

RESUMO

When access to molecular oxygen is restricted, Mycobacterium tuberculosis (Mtb) can respire an alternative electron acceptor, nitrate. We found that Mtb within infected primary human macrophages in vitro at physiologic tissue oxygen tensions respired nitrate, generating copious nitrite. A strain of Mtb lacking a functioning nitrate reductase was more susceptible than wild-type Mtb to treatment with isoniazid during infection of macrophages. Likewise, nitrate reductase-deficient Mtb was more susceptible to isoniazid than wild-type Mtb in axenic culture, and more resistant to hydrogen peroxide. These phenotypes were reversed by the addition of exogenous nitrite. Further investigation suggested that nitrite might inhibit the bacterial catalase. To the extent that Mtb itself is the most relevant source of nitrite acting within Mtb, these findings suggest that inhibitors of Mtb's nitrate transporter or nitrate reductase could enhance the efficacy of isoniazid.


Assuntos
Antibacterianos/farmacologia , Peróxido de Hidrogênio/farmacologia , Isoniazida/farmacologia , Macrófagos/microbiologia , Viabilidade Microbiana/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Nitritos/metabolismo , Células Cultivadas , Humanos , Macrófagos/imunologia , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/fisiologia , Nitrato Redutase/genética , Nitrato Redutase/metabolismo
19.
Proc Natl Acad Sci U S A ; 109(40): 16004-11, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-23012453

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Oxifenilbutazona/farmacologia , Animais , Cromatografia Líquida de Alta Pressão , Resistência Microbiana a Medicamentos/fisiologia , Ácidos Graxos/metabolismo , Feminino , Hidroxilação , Espectroscopia de Ressonância Magnética , Camundongos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/fisiologia , Oxifenilbutazona/metabolismo , Oxifenilbutazona/farmacocinética , Espécies Reativas de Nitrogênio/metabolismo
20.
Tuberculosis (Edinb) ; 91(5): 343-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21778115

RESUMO

The gene Rv2136c is annotated to encode the Mycobacterium tuberculosis (Mtb) homolog of Escherichia coli's undecaprenyl pyrophosphate phosphatase. In previous work, a genetic screen of 10,100 Mtb transposon mutants identified Rv2136c as being involved in acid resistance in Mtb. The Rv2136c:Tn strain was also sensitive to sodium dodecyl sulfate, lipophilic antibiotics, elevated temperature and reactive oxygen and nitrogen intermediates and was attenuated for growth and persistence in mice. However, none of these phenotypes could be genetically complemented, leading us to generate an Rv2136c knockout strain to test its role in Mtb pathogenicity. Genetic deletion revealed that Rv2136c is not responsible for any of the phenotypes observed in the transposon mutant strain. An independent genomic mutation is likely to have accounted for the extreme attenuation of this strain. Identification of the mutated gene will further our understanding of acid resistance mechanisms in Mtb and may offer a target for anti-tuberculosis chemotherapy.


Assuntos
Alquil e Aril Transferases/genética , Proteínas de Bactérias/genética , Macrófagos/metabolismo , Mutação/genética , Fagocitose/genética , Fagossomos/genética , Alquil e Aril Transferases/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Southern Blotting , Feminino , Deleção de Genes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA