Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Transfus Med Rev ; 37(3): 150748, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37827586

RESUMEN

Biphosphoglycerate mutase (BPGM) is a tri-functional enzyme expressed exclusively in erythroid cells and tissues that is responsible for the production of 2,3-biphosphoglycerate (2,3-BPG) through the Rapoport-Luebering shunt. The 2,3-BPG is required for efficient glycolysis and ATP production under anaerobic conditions, but is also a critical allosteric regulator of hemoglobin (Hb), acting to regulate oxygen release in peripheral tissues. In humans, BPGM deficiency is very rare, and is associated with reduced levels of erythrocytic 2,3-BPG and ATP, left shifted Hb-O2 dissociation curve, low P50, elevated Hb and constitutive erythrocytosis. BPGM deficiency in mice recapitulates the erythroid defects seen in human patients. A recent report has shown that BPGM deficiency in mice affords striking protection against both severe malaria anemia and cerebral malaria. These findings are reminiscent of studies of another erythrocyte specific glycolytic enzyme, Pyruvate Kinase (PKLR), which mutational inactivation protects humans and mice against malaria through impairment of glycolysis and ATP production in erythrocytes. BPGM, and PKLR join glucose-6-phosphate dehydrogenase (G6PD) and other erythrocyte variants as modulating response to malaria. Recent studies reviewed suggest glycolysis in general, and BPGM in particular, as a novel pharmacological target for therapeutic intervention in malaria.


Asunto(s)
Transferasas Intramoleculares , Malaria , Humanos , Ratones , Animales , Eritrocitos , Hemoglobinas , Oxígeno , Malaria/tratamiento farmacológico , Adenosina Trifosfato
2.
J Neurosurg Pediatr ; 31(6): 584-592, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36905673

RESUMEN

OBJECTIVE: The aim of this study was to characterize a novel pathogenic variant in the transient receptor potential vanilloid 4 (TRPV4) gene, causing familial nonsyndromic craniosynostosis (CS) with complete penetrance and variable expressivity. METHODS: Whole-exome sequencing was performed on germline DNA of a family with nonsyndromic CS to a mean depth coverage of 300× per sample, with greater than 98% of the targeted region covered at least 25×. In this study, the authors detected a novel variant, c.496C>A in TRPV4, exclusively in the four affected family members. The variant was modeled using the structure of the TRPV4 protein from Xenopus tropicalis. In vitro assays in HEK293 cells overexpressing wild-type TRPV4 or TRPV4 p.Leu166Met were used to assess the effect of the mutation on channel activity and downstream MAPK signaling. RESULTS: The authors identified a novel, highly penetrant heterozygous variant in TRPV4 (NM_021625.4:c.496C>A) causing nonsyndromic CS in a mother and all three of her children. This variant results in an amino acid change (p.Leu166Met) in the intracellular ankyrin repeat domain distant from the Ca2+-dependent membrane channel domain. In contrast to other TRPV4 mutations in channelopathies, this variant does not interfere with channel activity as identified by in silico modeling and in vitro overexpression assays in HEK293 cells. CONCLUSIONS: Based on these findings, the authors hypothesized that this novel variant causes CS by modulating the binding of allosteric regulatory factors to TRPV4 rather than directly modifying its channel activity. Overall, this study expands the genetic and functional spectrum of TRPV4 channelopathies and is particularly relevant for the genetic counseling of CS patients.


Asunto(s)
Canalopatías , Craneosinostosis , Humanos , Femenino , Niño , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/metabolismo , Penetrancia , Canalopatías/genética , Células HEK293 , Mutación/genética , Craneosinostosis/genética
3.
PLoS One ; 17(8): e0269684, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35921328

RESUMEN

Canonical aminoglycosides are a large group of antibiotics, where the part of chemical diversity stems from the substitution of the neamine ring system on positions 5 and 6. Certain aminoglycoside modifying enzymes can modify a broad range of 4,5- and 4,6-disubstituted aminoglycosides, with some as many as 15. This study presents the structural and kinetic results describing a promiscuous aminoglycoside acetyltransferase AAC(3)-IIIa. This enzyme has been crystallized in ternary complex with coenzyme A and 4,5- and 4,6-disubstituted aminoglycosides. We have followed up this work with kinetic characterization utilizing a panel of diverse aminoglycosides, including a next-generation aminoglycoside, plazomicin. Lastly, we observed an alternative binding mode of gentamicin in the aminoglycoside binding site, which was proven to be a crystallographic artifact based on mutagenesis.


Asunto(s)
Acetiltransferasas , Aminoglicósidos , Acetiltransferasas/metabolismo , Aminoglicósidos/química , Antibacterianos/química , Especificidad por Sustrato
4.
Cancer Res ; 82(19): 3499-3515, 2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-35913887

RESUMEN

CDC20 is a coactivator of the anaphase promoting complex/cyclosome (APC/C) and is essential for mitotic progression. APC/CCDC20 is inhibited by the spindle assembly checkpoint (SAC), which prevents premature separation of sister chromatids and aneuploidy in daughter cells. Although overexpression of CDC20 is common in many cancers, oncogenic mutations have never been identified in humans. Using whole-exome sequencing, we identified heterozygous missense CDC20 variants (L151R and N331K) that segregate with ovarian germ cell tumors in two families. Functional characterization showed these mutants retain APC/C activation activity but have impaired binding to BUBR1, a component of the SAC. Expression of L151R and N331K variants promoted mitotic slippage in HeLa cells and primary skin fibroblasts derived from carriers. Generation of mice carrying the N331K variant using CRISPR-Cas9 showed that, although homozygous N331K mice were nonviable, heterozygotes displayed accelerated oncogenicity of Myc-driven cancers. These findings highlight an unappreciated role for CDC20 variants as tumor-promoting genes. SIGNIFICANCE: Two germline CDC20 missense variants that segregate with cancer in two families compromise the spindle assembly checkpoint and lead to aberrant mitotic progression, which could predispose cells to transformation. See related commentary by Villarroya-Beltri and Malumbres, p. 3432.


Asunto(s)
Neoplasias , Huso Acromático , Ciclosoma-Complejo Promotor de la Anafase/genética , Animales , Proteínas Cdc20/genética , Proteínas Cdc20/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Germinativas/metabolismo , Células HeLa , Humanos , Ratones , Mitosis/genética , Neoplasias/metabolismo , Unión Proteica , Huso Acromático/metabolismo
5.
Front Microbiol ; 13: 933983, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35847101

RESUMEN

Since the end of 2019, the world has been challenged by the coronavirus disease 2019 (COVID-19) pandemic. With COVID-19 cases rising globally, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, resulting in the emergence of variants of interest (VOI) and of concern (VOC). Of the hundreds of millions infected, immunodeficient patients are one of the vulnerable cohorts that are most susceptible to this virus. These individuals include those with preexisting health conditions and/or those undergoing immunosuppressive treatment (secondary immunodeficiency). In these cases, several researchers have reported chronic infections in the presence of anti-COVID-19 treatments that may potentially lead to the evolution of the virus within the host. Such variations occurred in a variety of viral proteins, including key structural ones involved in pathogenesis such as spike proteins. Tracking and comparing such mutations with those arisen in the general population may provide information about functional sites within the SARS-CoV-2 genome. In this study, we reviewed the current literature regarding the specific features of SARS-CoV-2 evolution in immunocompromised patients and identified recurrent de novo amino acid changes in virus isolates of these patients that can potentially play an important role in SARS-CoV-2 pathogenesis and evolution.

6.
J Med Genet ; 59(3): 305-312, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33685999

RESUMEN

BACKGROUND: Pathogenic germline variants in Transient Receptor Potential Vanilloid 4 Cation Channel (TRPV4) lead to channelopathies, which are phenotypically diverse and heterogeneous disorders grossly divided in neuromuscular disorders and skeletal dysplasia. We recently reported in sporadic giant cell lesions of the jaws (GCLJs) novel, somatic, heterozygous, gain-of-function mutations in TRPV4, at Met713. METHODS: Here we report two unrelated women with a de novo germline p.Leu619Pro TRPV4 variant and an overlapping systemic disorder affecting all organs individually described in TRPV4 channelopathies. RESULTS: From an early age, both patients had several lesions of the nervous system including progressive polyneuropathy, and multiple aggressive giant cell-rich lesions of the jaws and craniofacial/skull bones, and other skeletal lesions. One patient had a relatively milder disease phenotype possibly due to postzygotic somatic mosaicism. Indeed, the TRPV4 p.Leu619Pro variant was present at a lower frequency (variant allele frequency (VAF)=21.6%) than expected for a heterozygous variant as seen in the other proband, and showed variable regional frequency in the GCLJ (VAF ranging from 42% to 10%). In silico structural analysis suggests that the gain-of-function p.Leu619Pro alters the ion channel activity leading to constitutive ion leakage. CONCLUSION: Our findings define a novel polysystemic syndrome due to germline TRPV4 p.Leu619Pro and further extend the spectrum of TRPV4 channelopathies. They further highlight the convergence of TRPV4 mutations on different organ systems leading to complex phenotypes which are further mitigated by possible post-zygotic mosaicism. Treatment of this disorder is challenging, and surgical intervention of the GCLJ worsens the lesions, suggesting the future use of MEK inhibitors and TRPV4 antagonists as therapeutic modalities for unmet clinical needs.


Asunto(s)
Canalopatías , Polineuropatías , Canales de Potencial de Receptor Transitorio , Femenino , Células Gigantes , Humanos , Maxilares , Mutación/genética , Cráneo , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/genética , Canales de Potencial de Receptor Transitorio/genética
7.
Commun Biol ; 4(1): 729, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117352

RESUMEN

The approval of plazomicin broadened the clinical library of aminoglycosides available for use against emerging bacterial pathogens. Contrarily to other aminoglycosides, resistance to plazomicin is limited; still, instances of resistance have been reported in clinical settings. Here, we present structural insights into the mechanism of plazomicin action and the mechanisms of clinical resistance. The structural data reveal that plazomicin exclusively binds to the 16S ribosomal A site, where it likely interferes with the fidelity of mRNA translation. The unique extensions to the core aminoglycoside scaffold incorporated into the structure of plazomicin do not interfere with ribosome binding, which is analogously seen in the binding of this antibiotic to the AAC(2')-Ia resistance enzyme. The data provides a structural rationale for resistance conferred by drug acetylation and ribosome methylation, i.e., the two mechanisms of resistance observed clinically. Finally, the crystal structures of plazomicin in complex with both its target and the clinically relevant resistance factor provide a roadmap for next-generation drug development that aims to ameliorate the impact of antibiotic resistance.


Asunto(s)
Antibacterianos/farmacología , Sisomicina/análogos & derivados , Antibacterianos/química , Antibacterianos/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Farmacorresistencia Bacteriana , Metilación , Providencia/efectos de los fármacos , Providencia/metabolismo , ARN Ribosómico 16S/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Sisomicina/química , Sisomicina/metabolismo , Sisomicina/farmacología , Relación Estructura-Actividad
8.
Sci Rep ; 11(1): 11614, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078922

RESUMEN

Plazomicin is currently the only next-generation aminoglycoside approved for clinical use that has the potential of evading the effects of widespread enzymatic resistance factors. However, plazomicin is still susceptible to the action of the resistance enzyme AAC(2')-Ia from Providencia stuartii. As the clinical use of plazomicin begins to increase, the spread of resistance factors will undoubtedly accelerate, rendering this aminoglycoside increasingly obsolete. Understanding resistance to plazomicin is an important step to ensure this aminoglycoside remains a viable treatment option for the foreseeable future. Here, we present three crystal structures of AAC(2')-Ia from P. stuartii, two in complex with acetylated aminoglycosides tobramycin and netilmicin, and one in complex with a non-substrate aminoglycoside, amikacin. Together, with our previously reported AAC(2')-Ia-acetylated plazomicin complex, these structures outline AAC(2')-Ia's specificity for a wide range of aminoglycosides. Additionally, our survey of AAC(2')-I homologues highlights the conservation of residues predicted to be involved in aminoglycoside binding, and identifies the presence of plasmid-encoded enzymes in environmental strains that confer resistance to the latest next-generation aminoglycoside. These results forecast the likely spread of plazomicin resistance and highlight the urgency for advancements in next-generation aminoglycoside design.


Asunto(s)
Acetiltransferasas/química , Antibacterianos/química , Proteínas Bacterianas/química , Farmacorresistencia Bacteriana/genética , Providencia/enzimología , Sisomicina/análogos & derivados , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Amicacina/química , Amicacina/metabolismo , Amicacina/farmacología , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Cinética , Modelos Moleculares , Netilmicina/química , Netilmicina/metabolismo , Netilmicina/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Providencia/química , Providencia/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sisomicina/química , Sisomicina/metabolismo , Sisomicina/farmacología , Especificidad por Sustrato , Tobramicina/química , Tobramicina/metabolismo , Tobramicina/farmacología
9.
Nat Commun ; 12(1): 1732, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741980

RESUMEN

Macrolides are a class of antibiotics widely used in both medicine and agriculture. Unsurprisingly, as a consequence of their exensive usage a plethora of resistance mechanisms have been encountered in pathogenic bacteria. One of these resistance mechanisms entails the enzymatic cleavage of the macrolides' macrolactone ring by erythromycin esterases (Eres). The most frequently identified Ere enzyme is EreA, which confers resistance to the majority of clinically used macrolides. Despite the role Eres play in macrolide resistance, research into this family enzymes has been sparse. Here, we report the first three-dimensional structures of an erythromycin esterase, EreC. EreC is an extremely close homologue of EreA, displaying more than 90% sequence identity. Two structures of this enzyme, in conjunction with in silico flexible docking studies and previously reported mutagenesis data allowed for the proposal of a detailed catalytic mechanism for the Ere family of enzymes, labeling them as metal-independent hydrolases. Also presented are substrate spectrum assays for different members of the Ere family. The results from these assays together with an examination of residue conservation for the macrolide binding site in Eres, suggests two distinct active site archetypes within the Ere enzyme family.


Asunto(s)
Antibacterianos/química , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Esterasas/química , Esterasas/genética , Macrólidos/química , Antibacterianos/farmacología , Bacterias/enzimología , Bacterias/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Hidrolasas de Éster Carboxílico/química , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Eritromicina/química , Genes Bacterianos , Macrólidos/farmacología , Simulación del Acoplamiento Molecular , Conformación Proteica , Difracción de Rayos X
10.
Cell ; 183(6): 1617-1633.e22, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33259802

RESUMEN

Histone H3.3 glycine 34 to arginine/valine (G34R/V) mutations drive deadly gliomas and show exquisite regional and temporal specificity, suggesting a developmental context permissive to their effects. Here we show that 50% of G34R/V tumors (n = 95) bear activating PDGFRA mutations that display strong selection pressure at recurrence. Although considered gliomas, G34R/V tumors actually arise in GSX2/DLX-expressing interneuron progenitors, where G34R/V mutations impair neuronal differentiation. The lineage of origin may facilitate PDGFRA co-option through a chromatin loop connecting PDGFRA to GSX2 regulatory elements, promoting PDGFRA overexpression and mutation. At the single-cell level, G34R/V tumors harbor dual neuronal/astroglial identity and lack oligodendroglial programs, actively repressed by GSX2/DLX-mediated cell fate specification. G34R/V may become dispensable for tumor maintenance, whereas mutant-PDGFRA is potently oncogenic. Collectively, our results open novel research avenues in deadly tumors. G34R/V gliomas are neuronal malignancies where interneuron progenitors are stalled in differentiation by G34R/V mutations and malignant gliogenesis is promoted by co-option of a potentially targetable pathway, PDGFRA signaling.


Asunto(s)
Neoplasias Encefálicas/genética , Carcinogénesis/genética , Glioma/genética , Histonas/genética , Interneuronas/metabolismo , Mutación/genética , Células-Madre Neurales/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Neoplasias Encefálicas/patología , Carcinogénesis/patología , Linaje de la Célula , Reprogramación Celular/genética , Cromatina/metabolismo , Embrión de Mamíferos/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioma/patología , Histonas/metabolismo , Lisina/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Clasificación del Tumor , Oligodendroglía/metabolismo , Regiones Promotoras Genéticas/genética , Prosencéfalo/embriología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transcripción Genética , Transcriptoma/genética
11.
Cell Rep ; 32(12): 108170, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32966787

RESUMEN

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Asunto(s)
Anemia/etiología , Anemia/prevención & control , Bisfosfoglicerato Mutasa/deficiencia , Malaria Cerebral/enzimología , Malaria Cerebral/prevención & control , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Bisfosfoglicerato Mutasa/química , Bisfosfoglicerato Mutasa/genética , Bisfosfoglicerato Mutasa/metabolismo , Estabilidad de Enzimas , Eritrocitos/enzimología , Eritrocitos/parasitología , Eritropoyesis , Matriz Extracelular/metabolismo , Femenino , Células HEK293 , Humanos , Malaria Cerebral/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación/genética , Parásitos/crecimiento & desarrollo , Plasmodium/crecimiento & desarrollo , Policitemia
12.
J Biol Chem ; 295(20): 7075-7095, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32277048

RESUMEN

Genetic screening has identified numerous variants of the endosomal solute carrier family 9 member A6 (SLC9A6)/(Na+,K+)/H+ exchanger 6 (NHE6) gene that cause Christianson syndrome, a debilitating X-linked developmental disorder associated with a range of neurological, somatic, and behavioral symptoms. Many of these variants cause complete loss of NHE6 expression, but how subtler missense substitutions or nonsense mutations that partially truncate its C-terminal cytoplasmic regulatory domain impair NHE6 activity and endosomal function are poorly understood. Here, we describe the molecular and cellular consequences of six unique mutations located in the N-terminal cytoplasmic segment (A9S), the membrane ion translocation domain (L188P and G383D), and the C-terminal regulatory domain (E547*, R568Q, and W570*) of human NHE6 that purportedly cause disease. Using a heterologous NHE6-deficient cell expression system, we show that the biochemical, catalytic, and cellular properties of the A9S and R568Q variants were largely indistinguishable from those of the WT transporter, which obscured their disease significance. By contrast, the L188P, G383D, E547*, and W570* mutants exhibited variable deficiencies in biosynthetic post-translational maturation, membrane sorting, pH homeostasis in recycling endosomes, and cargo trafficking, and they also triggered apoptosis. These findings broaden our understanding of the molecular dysfunctions of distinct NHE6 variants associated with Christianson syndrome.


Asunto(s)
Ataxia , Endosomas , Epilepsia , Enfermedades Genéticas Ligadas al Cromosoma X , Discapacidad Intelectual , Microcefalia , Mutación Missense , Trastornos de la Motilidad Ocular , Intercambiadores de Sodio-Hidrógeno , Sustitución de Aminoácidos , Animales , Ataxia/genética , Ataxia/metabolismo , Cricetinae , Endosomas/química , Endosomas/genética , Endosomas/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Microcefalia/genética , Microcefalia/metabolismo , Trastornos de la Motilidad Ocular/genética , Trastornos de la Motilidad Ocular/metabolismo , Dominios Proteicos , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo
13.
ACS Chem Biol ; 15(3): 686-694, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32100995

RESUMEN

Aminoglycoside antibiotics have lost much of their effectiveness due to widespread resistance, primarily via covalent modification. One of the most ubiquitous enzymes responsible for aminoglycoside resistance is aminoglycoside O-nucleotidyltransferase(2″), which catalyzes a nucleotidylation reaction. Due to its clinical importance, much research has focused on dissecting the mechanism of action, some of it dating back more than 30 years. Here, we present structural data for catalytically informative states of the enzyme, i.e., ANT(2″) in complex with adenosine monophosphate (AMP) and tobramycin (inactive-intermediate state) and in complex with adenylyl-2″-tobramycin, pyrophosphate, and Mn2+(product-bound state). These two structures in conjunction with our previously reported structure of ANT(2″)'s substrate-bound complex capture clinical states along ANT(2″)'s reaction coordinate. Additionally, isothermal titration calorimetry (ITC)-based studies are presented that assess the order of substrate binding and product release. Combined, these results outline a kinetic mechanism for ANT(2″) that contradicts what has been previously reported. Specifically, we show that the release of adenylated aminoglycoside precedes pyrophosphate. Furthermore, the ternary complex structures provide additional details on the catalytic mechanism, which reveals extensive similarities to the evolutionarily related DNA polymerase-ß superfamily.


Asunto(s)
Aminoglicósidos/farmacología , Antibacterianos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Nucleotidiltransferasas/metabolismo , Adenosina Monofosfato/química , Secuencia de Aminoácidos , Catálisis , Difosfatos/química , Cinética , Manganeso/química , Modelos Moleculares , Conformación Molecular , Unión Proteica , Transducción de Señal , Relación Estructura-Actividad , Especificidad por Sustrato , Tobramicina/química
14.
J Clin Invest ; 130(3): 1479-1490, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31805011

RESUMEN

BACKGROUNDDICER1 is the only miRNA biogenesis component associated with an inherited tumor syndrome, featuring multinodular goiter (MNG) and rare pediatric-onset lesions. Other susceptibility genes for familial forms of MNG likely exist.METHODSWhole-exome sequencing of a kindred with early-onset MNG and schwannomatosis was followed by investigation of germline pathogenic variants that fully segregated with the disease. Genome-wide analyses were performed on 13 tissue samples from familial and nonfamilial DGCR8-E518K-positive tumors, including MNG, schwannomas, papillary thyroid cancers (PTCs), and Wilms tumors. miRNA profiles of 4 tissue types were compared, and sequencing of miRNA, pre-miRNA, and mRNA was performed in a subset of 9 schwannomas, 4 of which harbor DGCR8-E518K.RESULTSWe identified c.1552G>A;p.E518K in DGCR8, a microprocessor component located in 22q, in the kindred. The variant identified is a somatic hotspot in Wilms tumors and has been identified in 2 PTCs. Copy number loss of chromosome 22q, leading to loss of heterozygosity at the DGCR8 locus, was found in all 13 samples harboring c.1552G>A;p.E518K. miRNA profiling of PTCs, MNG, schwannomas, and Wilms tumors revealed a common profile among E518K hemizygous tumors. In vitro cleavage demonstrated improper processing of pre-miRNA by DGCR8-E518K. MicroRNA and RNA profiling show that this variant disrupts precursor microRNA production, impacting populations of canonical microRNAs and mirtrons.CONCLUSIONWe identified DGCR8 as the cause of an unreported autosomal dominant mendelian tumor susceptibility syndrome: familial multinodular goiter with schwannomatosis.FUNDINGCanadian Institutes of Health Research, Compute Canada, Alex's Lemonade Stand Foundation, the Mia Neri Foundation for Childhood Cancer, Cassa di Sovvenzioni e Risparmio fra il Personale della Banca d'Italia, and the KinderKrebsInitiative Buchholz/Holm-Seppensen.


Asunto(s)
Predisposición Genética a la Enfermedad , Bocio Nodular/genética , Mutación Missense , Proteínas de Neoplasias/genética , Neurilemoma/genética , Neurofibromatosis/genética , Proteínas de Unión al ARN/genética , Neoplasias Cutáneas/genética , Sustitución de Aminoácidos , Niño , Cromosomas Humanos Par 22/genética , Femenino , Dosificación de Gen , Estudio de Asociación del Genoma Completo , Bocio Nodular/patología , Células HEK293 , Humanos , Masculino , Neurilemoma/patología , Neurofibromatosis/patología , Neoplasias Cutáneas/patología , Secuenciación del Exoma
15.
Infect Immun ; 88(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31792077

RESUMEN

We used a genome-wide screen in N-ethyl-N-nitrosourea (ENU)-mutagenized mice to identify genes in which recessive loss-of-function mutations protect against pathological neuroinflammation. We identified an R367Q mutation in the ZBTB7B (ThPOK) protein in which homozygosity causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Zbtb7bR367Q homozygous mice show a defect in the lymphoid compartment expressed as severe reduction in the number of single-positive CD4 T cells in the thymus and in the periphery, reduced brain infiltration of proinflammatory leukocytes in P. berghei ANKA-infected mice, and reduced production of proinflammatory cytokines by primary T cells ex vivo and in vivo Dampening of proinflammatory immune responses in Zbtb7bR367Q mice is concomitant to increased susceptibility to infection with avirulent (Mycobacterium bovis BCG) and virulent (Mycobacterium tuberculosis H37Rv) mycobacteria. The R367Q mutation maps to the first DNA-binding zinc finger domain of ThPOK and causes loss of base contact by R367 in the major groove of the DNA, which is predicted to impair DNA binding. Global immunoprecipitation of ThPOK-containing chromatin complexes coupled to DNA sequencing (ChIP-seq) identified transcriptional networks and candidate genes likely to play key roles in CD4+ CD8+ T cell development and in the expression of lineage-specific functions of these cells. This study highlights ThPOK as a global regulator of immune function in which alterations may affect normal responses to infectious and inflammatory stimuli.


Asunto(s)
Proteínas de Unión al ADN/genética , Malaria Cerebral/genética , Factores de Transcripción/genética , Tuberculosis Pulmonar/genética , Animales , Encéfalo/microbiología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD8-positivos/microbiología , Citocinas/genética , Femenino , Inflamación/genética , Inflamación/microbiología , Malaria Cerebral/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/patogenicidad , Plasmodium berghei/patogenicidad , Tuberculosis Pulmonar/microbiología , Virulencia/genética
16.
Front Chem ; 8: 612728, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33490038

RESUMEN

Phosphonates and bisphosphonates have proven their pharmacological utility as inhibitors of enzymes that metabolize phosphate and pyrophosphate substrates. The blockbuster class of drugs nitrogen-containing bisphosphonates represent one of the best-known examples. Widely used to treat bone-resorption disorders, these drugs work by inhibiting the enzyme farnesyl pyrophosphate synthase. Playing a key role in the isoprenoid biosynthetic pathway, this enzyme is also a potential anticancer target. Here, we provide a comprehensive overview of the research efforts to identify new inhibitors of farnesyl pyrophosphate synthase for various therapeutic applications. While the majority of these efforts have been directed against the human enzyme, some have been targeted on its homologs from other organisms, such as protozoan parasites and insects. Our particular focus is on the structures of the target enzymes and how the structural information has guided the drug discovery efforts.

17.
J Med Chem ; 62(21): 9691-9702, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31577901

RESUMEN

Thienopyrimidine-based allosteric inhibitors of the human farnesyl pyrophosphate synthase (hFPPS), characterized by a chiral α-aminophosphonic acid moiety, were synthesized as enantiomerically enriched pairs, and their binding mode was investigated by X-ray crystallography. A general consensus in the binding orientation of all (R)- and (S)-enantiomers was revealed. This finding is a prerequisite for establishing a reliable structure-activity relationship (SAR) model.


Asunto(s)
Ácido Aminoetilfosfónico/química , Ácido Aminoetilfosfónico/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ligasas/antagonistas & inhibidores , Ligasas/química , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos/metabolismo , Regulación Alostérica/efectos de los fármacos , Humanos , Ligasas/metabolismo , Modelos Moleculares , Conformación Proteica , Estereoisomerismo , Relación Estructura-Actividad
18.
ACS Omega ; 4(6): 10056-10069, 2019 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-31460098

RESUMEN

The worldwide use of the broad-spectrum antimicrobial trimethoprim (TMP) has induced the rise of TMP-resistant microorganisms. In addition to resistance-causing mutations of the microbial chromosomal dihydrofolate reductase (Dfr), the evolutionarily and structurally unrelated type II Dfrs (DfrBs) have been identified in TMP-resistant microorganisms. DfrBs are intrinsically TMP-resistant and allow bacterial proliferation when the microbial chromosomal Dfr is TMP-inhibited, making these enzymes important targets for inhibitor development. Furthermore, DfrBs occur in multiresistance plasmids, potentially accelerating their dissemination. We previously reported symmetrical bisbenzimidazoles that are the first selective inhibitors of the only well-characterized DfrB, DfrB1. Here, their diversification provides a new series of inhibitors (K i = 1.7-12.0 µM). Our results reveal two prominent features: terminal carboxylates and inhibitor length allow the establishment of essential interactions with DfrB1. Two crystal structures demonstrate the simultaneous binding of two inhibitor molecules in the symmetrical active site. Observations of those dimeric inhibitors inspired the design of monomeric analogues, binding in a single copy yet offering similar inhibition potency (K i = 1.1 and 7.4 µM). Inhibition of a second member of the DfrB family, DfrB4, suggests the generality of these inhibitors. These results provide key insights into inhibition of the highly TMP-resistant DfrBs, opening avenues to downstream development of antibiotics for combatting this emergent source of resistance.

19.
Sci Rep ; 9(1): 6656, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040324

RESUMEN

Understanding the principles of protein dynamics will help guide engineering of protein function: altering protein motions may be a barrier to success or may be an enabling tool for protein engineering. The impact of dynamics on protein function is typically reported over a fraction of the full scope of motional timescales. If motional patterns vary significantly at different timescales, then only by monitoring motions broadly will we understand the impact of protein dynamics on engineering functional proteins. Using an integrative approach combining experimental and in silico methodologies, we elucidate protein dynamics over the entire span of fast to slow timescales (ps to ms) for a laboratory-engineered system composed of five interrelated ß-lactamases: two natural homologs and three laboratory-recombined variants. Fast (ps-ns) and intermediate (ns-µs) dynamics were mostly conserved. However, slow motions (µs-ms) were few and conserved in the natural homologs yet were numerous and widely dispersed in their recombinants. Nonetheless, modified slow dynamics were functionally tolerated. Crystallographic B-factors from high-resolution X-ray structures were partly predictive of the conserved motions but not of the new slow motions captured in our solution studies. Our inspection of protein dynamics over a continuous range of timescales vividly illustrates the complexity of dynamic impacts of protein engineering as well as the functional tolerance of an engineered enzyme system to new slow motions.


Asunto(s)
Modelos Moleculares , Conformación Proteica , Ingeniería de Proteínas , beta-Lactamasas/química , Sitios de Unión , Cristalografía por Rayos X , Cinética , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , beta-Lactamasas/metabolismo
20.
Neurobiol Dis ; 121: 187-204, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30296617

RESUMEN

Loss-of-function mutations in the recycling endosomal (Na+,K+)/H+ exchanger gene SLC9A6/NHE6 result in overacidification and dysfunction of endosomal-lysosomal compartments, and cause a neurodevelopmental and degenerative form of X-linked intellectual disability called Christianson Syndrome (CS). However, knowledge of the disease heterogeneity of CS is limited. Here, we describe the clinical features and underlying molecular and cellular mechanisms associated with a CS patient carrying a de novo missense variant (p.Gly218Arg; G218R) of a conserved residue in its ion translocation domain that results in a potential gain-of-function. The patient manifested several core symptoms typical of CS, including pronounced cognitive impairment, mutism, epilepsy, ataxia and microcephaly; however, deterioration of motor function often observed after the first decade of life in CS children with total loss of SLC9A6/NHE6 function was not evident. In transfected non-neuronal cells, complex glycosylation and half-life of the G218R were significantly decreased compared to the wild-type transporter. This correlated with elevated ubiquitination and partial proteasomal-mediated proteolysis of G218R. However, a major fraction was delivered to the plasma membrane and endocytic pathways. Compared to wild-type, G218R-containing endosomes were atypically alkaline and showed impaired uptake of recycling endosomal cargo. Moreover, instead of accumulating in recycling endosomes, G218R was redirected to multivesicular bodies/late endosomes and ejected extracellularly in exosomes rather than progressing to lysosomes for degradation. Attenuated acidification and trafficking of G218R-containing endosomes were also observed in transfected hippocampal neurons, and correlated with diminished dendritic branching and density of mature mushroom-shaped spines and increased appearance of filopodia-like protrusions. Collectively, these findings expand our understanding of the genetic diversity of CS and further elucidate a critical role for SLC9A6/NHE6 in fine-tuning recycling endosomal pH and cargo trafficking, processes crucial for the maintenance of neuronal polarity and mature synaptic structures.


Asunto(s)
Ataxia/genética , Ataxia/patología , Endosomas/metabolismo , Epilepsia/genética , Epilepsia/patología , Mutación con Ganancia de Función , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Microcefalia/genética , Microcefalia/patología , Neuronas/patología , Trastornos de la Motilidad Ocular/genética , Trastornos de la Motilidad Ocular/patología , Intercambiadores de Sodio-Hidrógeno/genética , Adulto , Animales , Atrofia , Cricetulus , Dendritas/patología , Vesículas Extracelulares/metabolismo , Células HeLa , Hipocampo/patología , Humanos , Masculino , Mutación Missense , Intercambiadores de Sodio-Hidrógeno/química , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...