Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 187(6): 1363-1373.e12, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38366591

RESUMO

In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).


Assuntos
Vírus da Varíola dos Macacos , Varíola dos Macacos , Vacina Antivariólica , Animais , Humanos , Camundongos , Macaca fascicularis , Vírus da Varíola dos Macacos/genética , Varíola dos Macacos/imunologia , Varíola dos Macacos/prevenção & controle , Vacinas Combinadas , Vírus Vaccinia/genética
2.
Nat Aging ; 3(10): 1251-1268, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37723209

RESUMO

Aging is characterized by gradual immune dysfunction and increased disease risk. Genomic instability is considered central to the aging process, but the underlying mechanisms of DNA damage are insufficiently defined. Cells in confined environments experience forces applied to their nucleus, leading to transient nuclear envelope rupture (NER) and DNA damage. Here, we show that Lamin A/C protects lung alveolar macrophages (AMs) from NER and hallmarks of aging. AMs move within constricted spaces in the lung. Immune-specific ablation of lamin A/C results in selective depletion of AMs and heightened susceptibility to influenza virus-induced pathogenesis and lung cancer growth. Lamin A/C-deficient AMs that persist display constitutive NER marks, DNA damage and p53-dependent senescence. AMs from aged wild-type and from lamin A/C-deficient mice share a lysosomal signature comprising CD63. CD63 is required to limit damaged DNA in macrophages. We propose that NER-induced genomic instability represents a mechanism of aging in AMs.


Assuntos
Lamina Tipo A , Macrófagos Alveolares , Animais , Camundongos , Lamina Tipo A/genética , Membrana Nuclear , Pulmão , Envelhecimento/genética , Instabilidade Genômica
3.
iScience ; 24(12): 103425, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34877491

RESUMO

We previously showed stabilization of NIK-induced activation of NF-κB non-canonical signaling suppresses MLL-AF9-induced AML. In the current study, we demonstrate that deletion of NF-κB non-canonical RelB prevents the inhibitory effect of NIK stabilization in MLL-AF9 AML. Mechanistically, RelB suppresses its direct target, TIFAB, which is upregulated in human AML and correlates negatively with the survival of AML patients. Forced expression of TIFAB reverses NIK-induced impaired AML development through downregulation of RelB and upregulation of HOXA9. Consistent with upregulation of HOXA9, gene set enrichment analysis shows that forced expression of TIFAB blocks myeloid cell development, upregulates leukemia stem cell signature and induces similar gene expression patterns to those of HOXA9-MEIS1 and HOXA9-NUP98, and upregulates oxidative phosphorylation. Accordingly, forced expression of HOXA9 also largely releases the inhibitory impact of NIK stabilization via downregulation of RelB and upregulation of RelA. Our data suggest that NIK/RelB suppresses MLL-AF9-induced AML mainly through downregulation of TIFAB/HOXA9.

4.
Sci Signal ; 14(697)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429383

RESUMO

Modified vaccinia Ankara (MVA) is a live, attenuated human smallpox vaccine and a vector for the development of new vaccines against infectious diseases and cancer. Efficient activation of the immune system by MVA partially relies on its encounter with dendritic cells (DCs). MVA infection of DCs leads to multiple outcomes, including cytokine production, activation of costimulatory molecules for T cell stimulation, and cell death. Here, we examined how these diverse responses are orchestrated in human DCs. Single-cell analyses revealed that the response to MVA infection in DCs was limited to early viral gene expression. In response to the early events in the viral cycle, we found that DCs grouped into three distinct clusters. A cluster of infected cells sensed the MVA genome by the intracellular innate immunity pathway mediated by cGAS, STING, TBK1, and IRF3 and subsequently produced inflammatory cytokines. In response to these cytokines, a cluster of noninfected bystander cells increased costimulatory molecule expression. A separate cluster of infected cells underwent caspase-dependent apoptosis. Induction of apoptosis persisted after inhibition of innate immunity pathway mediators independently of previously described IRF-dependent or replication-dependent pathways and was a response to early MVA gene expression. Together, our study identified multiple mechanisms that underlie the interactions of MVA with human DCs.


Assuntos
Vaccinia , Vacinas Virais , Células Dendríticas , Humanos , Análise de Célula Única , Vacinas de DNA
5.
Methods Mol Biol ; 2366: 305-319, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34236647

RESUMO

The NF-κB signal transduction pathway has crucial functions in cell growth, survival, and the development of lymphocytes and other immune cells. Upon activation of the pathway, five distinct NF-κB transcription factor subunits that occur as homodimers or heterodimers comprise the downstream mediators that transcribe NF-κB target genes. A major quest in NF-κB research is to understand the biology of the separate subunits. However, determining the functions of the individual subunits using constitutional knockout mice is often hampered by the marked cell type and/or developmental stage-specific activation of the NF-κB pathway. To overcome these problems, we and others have generated loxP-flanked alleles of the genes encoding the different NF-κB subunits that upon crossing to suitable Cre-expressing mouse lines can be conditionally deleted in the desired cell type or developmental stage. We here describe the basic characteristics of conditional NF-κB subunit alleles rel (encoding c-REL), rela (RELA), relb (RELB), and nfkb2 (NF-κB2) generated in our laboratory that are available to the research community through a repository, and provide basic methods to study the consequences of tissue-specific ablation of NF-κB transcription factors in lymphocytes.


Assuntos
Linfócitos , Animais , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/metabolismo
7.
Cell Rep ; 26(9): 2377-2393.e13, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30811988

RESUMO

Cytosolic DNA activates cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS), an innate immune sensor pivotal in anti-microbial defense, senescence, auto-immunity, and cancer. cGAS is considered to be a sequence-independent DNA sensor with limited access to nuclear DNA because of compartmentalization. However, the nuclear envelope is a dynamic barrier, and cGAS is present in the nucleus. Here, we identify determinants of nuclear cGAS localization and activation. We show that nuclear-localized cGAS synthesizes cGAMP and induces innate immune activation of dendritic cells, although cGAMP levels are 200-fold lower than following transfection with exogenous DNA. Using cGAS ChIP-seq and a GFP-cGAS knockin mouse, we find nuclear cGAS enrichment on centromeric satellite DNA, confirmed by imaging, and to a lesser extent on LINE elements. The non-enzymatic N-terminal domain of cGAS determines nucleo-cytoplasmic localization, enrichment on centromeres, and activation of nuclear-localized cGAS. These results reveal a preferential functional association of nuclear cGAS with centromeres.


Assuntos
Centrômero/enzimologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Imunidade Inata/genética , Nucleotidiltransferases/metabolismo , Adulto , Animais , Linhagem Celular , Núcleo Celular/enzimologia , DNA , DNA Satélite , Feminino , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nucleotidiltransferases/química , Domínios Proteicos
8.
J Immunol ; 200(7): 2362-2371, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29459403

RESUMO

CD4+Foxp3+ regulatory T cells (Tregs) are essential regulators of immune responses. Perturbation of Treg homeostasis or function can lead to uncontrolled inflammation and autoimmunity. Therefore, understanding the molecular mechanisms involved in Treg biology remains an active area of investigation. It has been shown previously that the NF-κB family of transcription factors, in particular, the canonical pathway subunits, c-Rel and p65, are crucial for the development, maintenance, and function of Tregs. However, the role of the alternative NF-κB pathway components, p100 and RelB, in Treg biology remains unclear. In this article, we show that conditional deletion of the p100 gene, nfkb2, in Tregs, resulted in massive inflammation because of impaired suppressive function of nfkb2-deficient Tregs. Surprisingly, mice lacking RelB in Tregs did not exhibit the same phenotype. Instead, deletion of both relb and nfkb2 rescued the inflammatory phenotype, demonstrating an essential role for p100 as an inhibitor of RelB in Tregs. Our data therefore illustrate a new role for the alternative NF-κB signaling pathway in Tregs that has implications for the understanding of molecular pathways driving tolerance and immunity.


Assuntos
Tolerância Imunológica/imunologia , Subunidade p52 de NF-kappa B/genética , Proteínas Nucleares/genética , Linfócitos T Reguladores/imunologia , Fator de Transcrição RelB/genética , Animais , Autoimunidade/imunologia , Diferenciação Celular , Células Cultivadas , Endonucleases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade p52 de NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Linfócitos T Reguladores/citologia , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelB/metabolismo
9.
Immunol Cell Biol ; 95(3): 261-271, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27649781

RESUMO

Signaling through the canonical nuclear factor-κB (NF-κB) pathway is critical for the generation and maintenance of mature B cells and for antigen-dependent B-cell activation. c-REL (rel) and RELA (rela) are the downstream transcriptional activators of the canonical NF-κB pathway. Studies of B cells derived from constitutional rel knockout mice and chimeric mice repopulated with rela-/- fetal liver cells provided evidence that the subunits can have distinct roles during B-cell development. However, the B cell-intrinsic functions of c-REL and RELA during B-cell generation and antigen-dependent B-cell activation have not been determined in vivo. To clarify this issue, we crossed mice with conditional rel and rela alleles individually or in combination to mice that express Cre-recombinase in B cells. We here report that, whereas single deletion of rel or rela did not impair mature B-cell generation and maintenance, their simultaneous deletion led to a dramatic reduction of follicular and marginal zone B cells. Upon T cell-dependent immunization, B cell-specific deletion of the c-REL subunit alone abrogated the formation of germinal centers (GCs), whereas rela deletion did not affect GC formation. T-independent responses were strongly impaired in mice with B cell-specific deletion of rel, and only modestly in mice with RELA-deficient B cells. Our findings identify differential requirements for the canonical NF-κB subunits c-REL and RELA at distinct stages of mature B-cell development. The subunits are jointly required for the generation of mature B cells. During antigen-dependent B-cell activation, c-REL is the critical subunit required for the initiation of the GC reaction and for optimal T-independent antibody responses, with RELA being largely dispensable at this stage.


Assuntos
Linfócitos B/citologia , Linfócitos B/metabolismo , Ativação Linfocitária/imunologia , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Formação de Anticorpos/imunologia , Fator Ativador de Células B/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Sobrevivência Celular , Deleção de Genes , Centro Germinativo/citologia , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Baço/citologia
10.
Proc Natl Acad Sci U S A ; 113(32): 9063-8, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27457956

RESUMO

The NF-κB signaling cascade relays external signals essential for B-cell growth and survival. This cascade is frequently hijacked by cancers that arise from the malignant transformation of germinal center (GC) B cells, underscoring the importance of deciphering the function of NF-κB in these cells. The NF-κB signaling cascade is comprised of two branches, the canonical and alternative NF-κB pathways, mediated by distinct transcription factors. The expression and function of the transcription factors of the alternative pathway, RELB and NF-κB2, in late B-cell development is incompletely understood. Using conditional deletion of relb and nfkb2 in GC B cells, we here report that ablation of both RELB and NF-κB2, but not of the single transcription factors, resulted in the collapse of established GCs. RELB/NF-κB2 deficiency in GC B cells was associated with impaired cell-cycle entry and reduced expression of the cell-surface receptor inducible T-cell costimulator ligand that promotes optimal interactions between B and T cells. Analysis of human tonsillar tissue revealed that plasma cells and their precursors in the GC expressed high levels of NF-κB2 relative to surrounding lymphocytes. Accordingly, deletion of nfkb2 in murine GC B cells resulted in a dramatic reduction of antigen-specific antibody-secreting cells, whereas deletion of relb had no effect. These results demonstrate that the transcription factors of the alternative NF-κB pathway control distinct stages of late B-cell development, which may have implications for B-cell malignancies that aberrantly activate this pathway.


Assuntos
Linfócitos B/fisiologia , Centro Germinativo/fisiologia , NF-kappa B/fisiologia , Fatores de Transcrição/fisiologia , Animais , Antígenos CD40/fisiologia , Células Cultivadas , Humanos , Camundongos , Transdução de Sinais/fisiologia , Fator de Transcrição RelB/fisiologia
11.
J Immunol ; 196(6): 2591-601, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26851215

RESUMO

BAFF is critical for the survival and maturation of mature B cells. BAFF, via BAFFR, activates multiple signaling pathways in B cells, including the alternative NF-κB pathway. The transcription factors RELB and NF-κB2 (p100/p52) are the downstream mediators of the alternative pathway; however, the B cell-intrinsic functions of these NF-κB subunits have not been studied in vivo using conditional alleles, either individually or in combination. We in this study report that B cell-specific deletion of relb led to only a slight decrease in the fraction of mature splenic B cells, whereas deletion of nfkb2 caused a marked reduction. This phenotype was further exacerbated upon combined deletion of relb and nfkb2 and most dramatically affected the maintenance of marginal zone B cells. BAFF stimulation, in contrast to CD40 activation, was unable to rescue relb/nfkb2-deleted B cells in vitro. RNA-sequencing analysis of BAFF-stimulated nfkb2-deleted versus normal B cells suggests that the alternative NF-κB pathway, in addition to its critical role in BAFF-mediated cell survival, may control the expression of genes involved in the positioning of B cells within the lymphoid microenvironment and in the establishment of T cell-B cell interactions. Thus, by ablating the downstream transcription factors of the alternative NF-κB pathway specifically in B cells, we identify in this study a critical role for the combined activity of the RELB and NF-κB2 subunits in B cell homeostasis that cannot be compensated for by the canonical NF-κB pathway under physiological conditions.


Assuntos
Linfócitos B/citologia , Homeostase/imunologia , Subunidade p52 de NF-kappa B/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Separação Celular , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Subunidade p52 de NF-kappa B/metabolismo , Fator de Transcrição RelB/imunologia , Fator de Transcrição RelB/metabolismo
12.
Nat Rev Immunol ; 15(3): 137-48, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25656706

RESUMO

Humoral immunity depends on the germinal centre (GC) reaction during which somatically mutated high-affinity memory B cells and plasma cells are generated. Recent studies have uncovered crucial cues that are required for the formation and the maintenance of GCs and for the selection of high-affinity antibody mutants. In addition, it is now clear that these events are promoted by the dynamic movements of cells within and between GCs. These findings have resolved the complexities of the GC reaction in greater detail than ever before. This Review focuses on these recent advances and discusses their implications for the establishment of humoral immunity.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Imunidade Humoral/imunologia , Afinidade de Anticorpos/imunologia , Humanos , Plasmócitos/imunologia
13.
J Exp Med ; 211(10): 2103-18, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25180063

RESUMO

Germinal centers (GCs) are the sites where memory B cells and plasma cells producing high-affinity antibodies are generated during T cell-dependent immune responses. The molecular control of GC B cell maintenance and differentiation remains incompletely understood. Activation of the NF-κB signaling pathway has been implicated; however, the distinct roles of the individual NF-κB transcription factor subunits are unknown. We report that GC B cell-specific deletion of the NF-κB subunits c-REL or RELA, which are both activated by the canonical NF-κB pathway, abolished the generation of high-affinity B cells via different mechanisms acting at distinct stages during the GC reaction. c-REL deficiency led to the collapse of established GCs immediately after the formation of dark and light zones at day 7 of the GC reaction and was associated with the failure to activate a metabolic program that promotes cell growth. Conversely, RELA was dispensable for GC maintenance but essential for the development of GC-derived plasma cells due to impaired up-regulation of BLIMP1. These results indicate that activation of the canonical NF-κB pathway in GC B cells controls GC maintenance and differentiation through distinct transcription factor subunits. Our findings have implications for the role of NF-κB in GC lymphomagenesis.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular/imunologia , Centro Germinativo/imunologia , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fluoresceínas , Deleção de Genes , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Proto-Oncogênicas c-rel/genética , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Succinimidas , Fator de Transcrição RelA/genética , Fatores de Transcrição/metabolismo
14.
Biochem J ; 458(3): 513-23, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24428730

RESUMO

Plasmodium parasites possess two endosymbiotic organelles: a mitochondrion and a relict plastid called the apicoplast. To accommodate the translational requirements of these organelles in addition to its cytosolic translation apparatus, the parasite must maintain a supply of charged tRNA molecules in each of these compartments. In the present study we investigate how the parasite manages these translational requirements for charged tRNACys with only a single gene for CysRS (cysteinyl-tRNA synthetase). We demonstrate that the single PfCysRS (Plasmodium falciparum CysRS) transcript is alternatively spliced, and, using a combination of endogenous and heterologous tagging experiments in both P. falciparum and Toxoplasma gondii, we show that CysRS isoforms traffic to the cytosol and apicoplast. PfCysRS can recognize and charge the eukaryotic tRNACys encoded by the Plasmodium nucleus as well as the bacterial-type tRNA encoded by the apicoplast genome, albeit with a preference for the eukaryotic type cytosolic tRNA. The results of the present study indicate that apicomplexan parasites have lost their original plastidic cysteinyl-tRNA synthetase, and have replaced it with a dual-targeted eukaryotic type CysRS that recognizes plastid and nuclear tRNACys. Inhibitors of the Plasmodium dual-targeted CysRS would potentially offer a therapy capable of the desirable immediate effects on parasite growth as well as the irreversibility of inhibitors that disrupt apicoplast inheritance.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Citosol/metabolismo , Plasmodium falciparum/enzimologia , Processamento Alternativo , Aminoacil-tRNA Sintetases/genética , Apicoplastos/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Mutação , Plasmodium falciparum/genética , Transporte Proteico , Temperatura , Toxoplasma/genética
15.
J Exp Med ; 210(13): 2887-902, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24323359

RESUMO

The transcription factor interferon regulatory factor-4 (IRF4) is expressed in B cells at most developmental stages. In antigen-activated B cells, IRF4 controls germinal center formation, class-switch recombination, and the generation of plasma cells. Here we describe a novel function for IRF4 in the homeostasis of mature B cells. Inducible deletion of irf4 specifically in B cells in vivo led to the aberrant accumulation of irf4-deleted follicular B cells in the marginal zone (MZ) area. IRF4-deficient B cells showed elevated protein expression and activation of NOTCH2, a transmembrane receptor and transcriptional regulator known to be required for MZ B cell development. Administration of a NOTCH2-inhibitory antibody abolished nuclear translocation of NOTCH2 in B cells within 12 h and caused a rapid and progressive disintegration of the MZ that was virtually complete 48 h after injection. The disappearance of the MZ was accompanied by a transient increase of MZ-like B cells in the blood rather than increased B cell apoptosis, demonstrating that continued NOTCH2 activation is critical for the retention of B cells in the MZ. Our results suggest that IRF4 controls the positioning of mature B cells in the lymphoid microenvironments by regulating NOTCH2 expression. These findings may have implications for the understanding of B cell malignancies with dysregulated IRF4 and NOTCH2 activity.


Assuntos
Linfócitos B/metabolismo , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/fisiologia , Receptor Notch2/metabolismo , Animais , Apoptose , Linfócitos B/citologia , Citometria de Fluxo , Deleção de Genes , Centro Germinativo/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Fatores Reguladores de Interferon/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Fenótipo , Transdução de Sinais , Baço/metabolismo
16.
Immunol Rev ; 247(1): 73-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22500833

RESUMO

Interferon regulatory factor 4 (IRF4) is a member of the IRF family of transcription factors and is expressed in most cell types of the immune system. Within the B-cell lineage, IRF4 is expressed in all developmental stages except during the germinal center (GC) reaction. IRF4 expression, however, is upregulated during exit from the GC reaction and has been demonstrated to have critical functions in at least three key developmental processes: the termination of the GC B-cell transcriptional program, immunoglobulin (Ig) class switch recombination (CSR), and plasma cell development. Herein, we attempt to reconcile the often contradictory findings regarding IRF4 into a model to explain the role of IRF4 in the transcription factor networks that operate within exiting GC B cells. In addition, a deregulation of the biological programs controlled by IRF4 has recently been implicated in the pathogenesis of various B-cell-derived malignancies. Determining the specific functions of IRF4 in the markedly diverse developmental processes that coordinate B-cell development is therefore likely to have important implications for understanding these malignancies and devising therapeutic interventions.


Assuntos
Linfócitos B/citologia , Diferenciação Celular , Centro Germinativo , Fatores Reguladores de Interferon/metabolismo , Animais , Centro Germinativo/fisiologia , Humanos , Fatores Reguladores de Interferon/química , Modelos Biológicos
17.
Int J Parasitol ; 42(2): 177-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22222968

RESUMO

The causative agent of malaria, Plasmodium, possesses three translationally active compartments: the cytosol, the mitochondrion and a relic plastid called the apicoplast. Aminoacyl-tRNA synthetases to charge tRNA are thus required for all three compartments. However, the Plasmodiumfalciparum genome encodes too few tRNA synthetases to supply a unique enzyme for each amino acid in all three compartments. We have investigated the subcellular localisation of three tRNA synthetases (AlaRS, GlyRS and ThrRS), which occur only once in the nuclear genome, and we show that each of these enzymes is dually localised to the P. falciparum cytosol and the apicoplast. No mitochondrial fraction is apparent for these three enzymes, which suggests that the Plasmodium mitochondrion lacks at least these three tRNA synthetases. The unique Plasmodium ThrRS is the presumed target of the antimalarial compound borrelidin. Borrelidin kills P. falciparum parasites quickly without the delayed death effect typical of apicoplast translation inhibitors and without an observable effect on apicoplast morphology. By contrast, mupirocin, an inhibitor of the apicoplast IleRS, kills with a delayed death effect that inhibits apicoplast growth and division. Because inhibition of dual targeted tRNA synthetases should arrest translation in all compartments of the parasite, these enzymes deserve further investigation as potential targets for antimalarial drug development.


Assuntos
Aminoacil-tRNA Sintetases/metabolismo , Citosol/enzimologia , Plasmodium falciparum/fisiologia , Plastídeos/enzimologia , Antimaláricos/farmacologia , Álcoois Graxos/farmacologia , Mupirocina/farmacologia , Plasmodium falciparum/metabolismo , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...