Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Hum Mol Genet ; 33(3): 211-223, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-37819629

RESUMEN

Duchenne muscular dystrophy (DMD) is a progressive disabling X-linked recessive disorder that causes gradual and irreversible loss of muscle, resulting in early death. The corticosteroids prednisone/prednisolone and deflazacort are used to treat DMD as the standard of care; however, only deflazacort is FDA approved for DMD. The novel atypical corticosteroid vamorolone is being investigated for treatment of DMD. We compared the pharmaceutical properties as well as the efficacy and safety of the three corticosteroids across multiple doses in the B10-mdx DMD mouse model. Pharmacokinetic studies in the mouse and evaluation of p-glycoprotein (P-gP) efflux in a cellular system demonstrated that vamorolone is not a strong P-gp substrate resulting in measurable central nervous system (CNS) exposure in the mouse. In contrast, deflazacort and prednisolone are strong P-gp substrates. All three corticosteroids showed efficacy, but also side effects at efficacious doses. After dosing mdx mice for two weeks, all three corticosteroids induced changes in gene expression in the liver and the muscle, but prednisolone and vamorolone induced more changes in the brain than did deflazacort. Both prednisolone and vamorolone induced depression-like behavior. All three corticosteroids reduced endogenous corticosterone levels, increased glucose levels, and reduced osteocalcin levels. Using micro-computed tomography, femur bone density was decreased, reaching significance with prednisolone. The results of these studies indicate that efficacious doses of vamorolone, are associated with similar side effects as seen with other corticosteroids. Further, because vamorolone is not a strong P-gp substrate, vamorolone distributes into the CNS increasing the potential CNS side-effects.


Asunto(s)
Distrofia Muscular de Duchenne , Prednisolona , Pregnadienodioles , Pregnenodionas , Animales , Ratones , Prednisolona/uso terapéutico , Microtomografía por Rayos X , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Corticosterona/uso terapéutico , Preparaciones Farmacéuticas
2.
Am J Hum Genet ; 110(3): 531-547, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36809767

RESUMEN

Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the central and peripheral nervous systems. FD is a complex neurological disorder accompanied by severe gait ataxia and retinal degeneration. There is currently no effective treatment to restore ELP1 production in individuals with FD, and the disease is ultimately fatal. After identifying kinetin as a small molecule able to correct the ELP1 splicing defect, we worked on its optimization to generate novel splicing modulator compounds (SMCs) that can be used in individuals with FD. Here, we optimize the potency, efficacy, and bio-distribution of second-generation kinetin derivatives to develop an oral treatment for FD that can efficiently pass the blood-brain barrier and correct the ELP1 splicing defect in the nervous system. We demonstrate that the novel compound PTC258 efficiently restores correct ELP1 splicing in mouse tissues, including brain, and most importantly, prevents the progressive neuronal degeneration that is characteristic of FD. Postnatal oral administration of PTC258 to the phenotypic mouse model TgFD9;Elp1Δ20/flox increases full-length ELP1 transcript in a dose-dependent manner and leads to a 2-fold increase in functional ELP1 in the brain. Remarkably, PTC258 treatment improves survival, gait ataxia, and retinal degeneration in the phenotypic FD mice. Our findings highlight the great therapeutic potential of this novel class of small molecules as an oral treatment for FD.


Asunto(s)
Disautonomía Familiar , Enfermedades Neurodegenerativas , Degeneración Retiniana , Ratones , Animales , Disautonomía Familiar/genética , Cinetina , Ataxia de la Marcha , Administración Oral
3.
Hum Mol Genet ; 31(1): 82-96, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34368854

RESUMEN

Spinal muscular atrophy (SMA) is caused by the loss of the survival motor neuron 1 (SMN1) gene function. The related SMN2 gene partially compensates but produces insufficient levels of SMN protein due to alternative splicing of exon 7. Evrysdi™ (risdiplam), recently approved for the treatment of SMA, and related compounds promote exon 7 inclusion to generate full-length SMN2 mRNA and increase SMN protein levels. SMNΔ7 type I SMA mice survive without treatment for ~17 days. SMN2 mRNA splicing modulators increase survival of SMN∆7 mice with treatment initiated at postnatal day 3 (PND3). To define SMN requirements for adult mice, SMNΔ7 mice were dosed with an SMN2 mRNA splicing modifier from PND3 to PND40, then dosing was stopped. Mice not treated after PND40 showed progressive weight loss, necrosis, and muscle atrophy after ~20 days. Male mice presented a more severe phenotype than female mice. Mice dosed continuously did not show disease symptoms. The estimated half-life of SMN protein is 2 days indicating that the SMA phenotype reappeared after SMN protein levels returned to baseline. Although SMN protein levels decreased with age in mice and SMN protein levels were higher in brain than in muscle, our studies suggest that SMN protein is required throughout the life of the mouse and is especially essential in adult peripheral tissues including muscle. These studies indicate that drugs such as risdiplam will be optimally therapeutic when given as early as possible after diagnosis and potentially will be required for the life of an SMA patient.


Asunto(s)
Atrofia Muscular Espinal , Empalme Alternativo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Exones , Femenino , Humanos , Masculino , Ratones , Atrofia Muscular Espinal/metabolismo , Empalme del ARN , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora
4.
Am J Hum Genet ; 104(4): 638-650, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905397

RESUMEN

Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;IkbkapΔ20/flox recapitulates the proprioceptive impairment observed in individuals with FD, and we provide the in vivo evidence that postnatal correction, promoted by the small molecule kinetin, of the mutant ELP1 splicing can rescue neurological phenotypes in FD. Daily administration of kinetin starting at birth improves sensory-motor coordination and prevents the onset of spinal abnormalities by stopping the loss of proprioceptive neurons. These phenotypic improvements correlate with increased amounts of full-length ELP1 mRNA and protein in multiple tissues, including in the peripheral nervous system (PNS). Our results show that postnatal correction of the underlying ELP1 splicing defect can rescue devastating disease phenotypes and is therefore a viable therapeutic approach for persons with FD.


Asunto(s)
Disautonomía Familiar/terapia , Cinetina/uso terapéutico , Propiocepción , Empalme del ARN , Factores de Elongación Transcripcional/genética , Alelos , Animales , Conducta Animal , Línea Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Exones , Fibroblastos , Genotipo , Humanos , Intrones , Cinetina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neuronas/metabolismo , Fenotipo
5.
Anal Biochem ; 656: 114876, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36058293

RESUMEN

Paired box protein Pax-6 (oculothrombin) is a transcription factor that plays an important regulatory role in ocular, brain, and pancreatic development. Mutations of the PAX6 gene cause aniridia and Peters anomaly. Reduction in Pax-6 protein is also associated with ocular diseases such as dry eye. An electrochemiluminescence immunoassay method using the Meso Scale Discovery platform was developed to measure Pax-6 protein levels in corneal epithelial cells obtained by impression cytology. Impression cytology involves harvesting ocular epithelial cells by applying a polyethersulfone membrane patch briefly to the ocular surface using a commercially available EYEPRIM™ device. The epithelial cells that adhere to the membrane patch of the EYEPRIM™ device provide a biological sample which can be assayed for Pax-6 protein levels. Assay development identified an antibody pair capable of detecting purified recombinant Pax-6 protein produced in mammalian cells. The optimized assay has a dynamic range of 24 pg mL-1 to 100,000 pg mL-1 and a lower limit of quantification of 24 pg mL-1. Assay selectivity was demonstrated using either HeLa or HEK293 cells transfected with inhibitory RNA. Finally, the method was validated by measuring Pax-6 protein levels in impression cytology acquired samples obtained using the EYEPRIM™ device from rabbit cornea.


Asunto(s)
Proteínas de Homeodominio , Factores de Transcripción Paired Box , Animales , Proteínas del Ojo/genética , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Inmunoensayo , Mamíferos/genética , Mamíferos/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , ARN , Conejos , Proteínas Represoras/genética
6.
Hum Mol Genet ; 25(10): 1885-1899, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26931466

RESUMEN

Spinal muscular atrophy (SMA) is caused by the loss or mutation of both copies of the survival motor neuron 1 (SMN1) gene. The related SMN2 gene is retained, but due to alternative splicing of exon 7, produces insufficient levels of the SMN protein. Here, we systematically characterize the pharmacokinetic and pharmacodynamics properties of the SMN splicing modifier SMN-C1. SMN-C1 is a low-molecular weight compound that promotes the inclusion of exon 7 and increases production of SMN protein in human cells and in two transgenic mouse models of SMA. Furthermore, increases in SMN protein levels in peripheral blood mononuclear cells and skin correlate with those in the central nervous system (CNS), indicating that a change of these levels in blood or skin can be used as a non-invasive surrogate to monitor increases of SMN protein levels in the CNS. Consistent with restored SMN function, SMN-C1 treatment increases the levels of spliceosomal and U7 small-nuclear RNAs and corrects RNA processing defects induced by SMN deficiency in the spinal cord of SMNΔ7 SMA mice. A 100% or greater increase in SMN protein in the CNS of SMNΔ7 SMA mice robustly improves the phenotype. Importantly, a ∼50% increase in SMN leads to long-term survival, but the SMA phenotype is only partially corrected, indicating that certain SMA disease manifestations may respond to treatment at lower doses. Overall, we provide important insights for the translation of pre-clinical data to the clinic and further therapeutic development of this series of molecules for SMA treatment.


Asunto(s)
Isocumarinas/administración & dosificación , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Piperazinas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacocinética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Animales , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Exones/genética , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/sangre , Atrofia Muscular Espinal/patología , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Piel/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Proteína 2 para la Supervivencia de la Neurona Motora/sangre
7.
Bioorg Med Chem Lett ; 27(22): 5014-5021, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29032026

RESUMEN

The continued emergence of bacteria resistant to current standard of care antibiotics presents a rapidly growing threat to public health. New chemical entities (NCEs) to treat these serious infections are desperately needed. Herein we report the discovery, synthesis, SAR and in vivo efficacy of a novel series of 4-hydroxy-2-pyridones exhibiting activity against Gram-negative pathogens. Compound 1c, derived from the N-debenzylation of 1b, preferentially inhibits bacterial DNA synthesis as determined by standard macromolecular synthesis assays. The structural features of the 4-hydroxy-2-pyridone scaffold required for antibacterial activity were explored and compound 6q, identified through further optimization of the series, had an MIC90 value of 8 µg/mL against a panel of highly resistant strains of E. coli. In a murine septicemia model, compound 6q exhibited a PD50 of 8 mg/kg in mice infected with a lethal dose of E. coli. This novel series of 4-hydroxy-2-pyridones serves as an excellent starting point for the identification of NCEs treating Gram-negative infections.


Asunto(s)
Antibacterianos/metabolismo , Compuestos de Azabiciclo/química , ADN/metabolismo , Niacina/análogos & derivados , Piridinas/química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/metabolismo , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , ADN/química , Evaluación Preclínica de Medicamentos , Escherichia coli/efectos de los fármacos , Escherichia coli/patogenicidad , Bacterias Gramnegativas/efectos de los fármacos , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/veterinaria , Semivida , Ratones , Pruebas de Sensibilidad Microbiana , Niacina/metabolismo , Niacina/farmacología , Niacina/uso terapéutico , Piridinas/metabolismo , Piridinas/farmacología , Piridinas/uso terapéutico , Relación Estructura-Actividad
8.
Sci Rep ; 13(1): 18600, 2023 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-37903840

RESUMEN

Familial dysautonomia (FD) is a rare neurodevelopmental and neurodegenerative disease caused by a splicing mutation in the Elongator Acetyltransferase Complex Subunit 1 (ELP1) gene. The reduction in ELP1 mRNA and protein leads to the death of retinal ganglion cells (RGCs) and visual impairment in all FD patients. Currently patient symptoms are managed, but there is no treatment for the disease. We sought to test the hypothesis that restoring levels of Elp1 would thwart the death of RGCs in FD. To this end, we tested the effectiveness of two therapeutic strategies for rescuing RGCs. Here we provide proof-of-concept data that gene replacement therapy and small molecule splicing modifiers effectively reduce the death of RGCs in mouse models for FD and provide pre-clinical foundational data for translation to FD patients.


Asunto(s)
Disautonomía Familiar , Enfermedades Neurodegenerativas , Ratones , Animales , Humanos , Células Ganglionares de la Retina/metabolismo , Disautonomía Familiar/genética , Disautonomía Familiar/terapia , Disautonomía Familiar/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Empalme del ARN , Terapia Genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
9.
bioRxiv ; 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37293016

RESUMEN

Familial dysautonomia (FD) is a rare neurodevelopmental and neurodegenerative disease caused by a splicing mutation in the Elongator Acetyltransferase Complex Subunit 1 ( ELP1 ) gene. The reduction in ELP1 mRNA and protein leads to the death of retinal ganglion cells (RGCs) and visual impairment in all FD patients. Currently, patient symptoms are managed, but there is no treatment for the disease. We sought to test the hypothesis that restoring levels of Elp1 would thwart the death of RGCs in FD. To this end, we tested the effectiveness of two therapeutic strategies for rescuing RGCs. Here we provide proof-of-concept data that gene replacement therapy and small molecule splicing modifiers effectively reduce the death of RGCs in mouse models for FD and provide pre-clinical data foundation for translation to FD patients.

10.
Elife ; 112022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35137690

RESUMEN

Antibiotic-resistant Neisseria gonorrhoeae (Ng) are an emerging public health threat due to increasing numbers of multidrug resistant (MDR) organisms. We identified two novel orally active inhibitors, PTC-847 and PTC-672, that exhibit a narrow spectrum of activity against Ng including MDR isolates. By selecting organisms resistant to the novel inhibitors and sequencing their genomes, we identified a new therapeutic target, the class Ia ribonucleotide reductase (RNR). Resistance mutations in Ng map to the N-terminal cone domain of the α subunit, which we show here is involved in forming an inhibited α4ß4 state in the presence of the ß subunit and allosteric effector dATP. Enzyme assays confirm that PTC-847 and PTC-672 inhibit Ng RNR and reveal that allosteric effector dATP potentiates the inhibitory effect. Oral administration of PTC-672 reduces Ng infection in a mouse model and may have therapeutic potential for treatment of Ng that is resistant to current drugs.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Gonorrea/tratamiento farmacológico , Piridinas/farmacología , Ribonucleótido Reductasas/metabolismo , Regulación Alostérica , Animales , Nucleótidos de Desoxiadenina/metabolismo , Modelos Animales de Enfermedad , Escherichia coli/efectos de los fármacos , Femenino , Gonorrea/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana/métodos , Neisseria gonorrhoeae/efectos de los fármacos
11.
Nat Commun ; 12(1): 3332, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099697

RESUMEN

Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.


Asunto(s)
Aprendizaje Profundo , Marcación de Gen/métodos , Empalme del ARN , Animales , Biología Computacional , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Exones , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Homólogo 1 de la Proteína MutL/genética , Mutación , Fenetilaminas/administración & dosificación , Piridazinas/administración & dosificación , Esterol Esterasa/genética , Transcriptoma , Proteínas tau/genética
12.
Nat Commun ; 12(1): 7299, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34911927

RESUMEN

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.


Asunto(s)
Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Empalme del ARN , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Animales , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/metabolismo , Ratones , Empalme del ARN/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , Expansión de Repetición de Trinucleótido/efectos de los fármacos
13.
J Biol Chem ; 284(37): 25254-67, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19546227

RESUMEN

When cells are subjected to nutritional stress, uncharged tRNAs accumulate and activate Gcn2p phosphorylation of eukaryotic initiation factor-2 (eIF2) and the general amino acid control pathway. The Gcn2p regulatory domain homologous to histidyl-tRNA synthetases is proposed to bind to uncharged tRNA, directly contributing to activation of Gcn2p. Here we apply a microarray technology to analyze genome-wide changes in tRNA charging in yeast upon activation of Gcn2p in response to amino acid starvation and high salinity, a stress not directly linked to nutritional deficiency. This microarray technology is applicable for all eukaryotic cells. Strains were starved for histidine, leucine, or tryptophan and shown to rapidly induce Gcn2p phosphorylation of eIF2. The relative charging level of all tRNAs was measured before and after starvation, and Gcn2p activation and the intracellular levels of the starved amino acid correlate with the observed decrease in tRNA charging. Interestingly, in some cases, tRNAs not charged with the starved amino acid became deacylated more rapidly than tRNAs charged with the starved amino acid. This increase in uncharged tRNA levels occurred although the intracellular levels for these non-starved amino acids remained unchanged. Additionally, treatment of a wild-type strain with high salinity stress showed transient changes in the charging of several different tRNAs. These results suggest that Gcn2p can be activated by many different tRNA species in the cell. These results also depict a complex cellular relationship between tRNA charging, amino acid availability, and non-nutrient stress. These relationships are best revealed by simultaneous monitoring of the charging level of all tRNAs.


Asunto(s)
Genoma Fúngico , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , eIF-2 Quinasa/metabolismo , Aminoácidos/química , Northern Blotting , Factor 2 Eucariótico de Iniciación/metabolismo , Immunoblotting , Leucina/química , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , ARN de Transferencia/química , Proteínas de Saccharomyces cerevisiae/genética , Cloruro de Sodio/farmacología , Factores de Tiempo , Triptófano/química
14.
J Med Chem ; 61(10): 4456-4475, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29727185

RESUMEN

There exists an urgent medical need to identify new chemical entities (NCEs) targeting multidrug resistant (MDR) bacterial infections, particularly those caused by Gram-negative pathogens. 4-Hydroxy-2-pyridones represent a novel class of nonfluoroquinolone inhibitors of bacterial type II topoisomerases active against MDR Gram-negative bacteria. Herein, we report on the discovery and structure-activity relationships of a series of fused indolyl-containing 4-hydroxy-2-pyridones with improved in vitro antibacterial activity against fluoroquinolone resistant strains. Compounds 6o and 6v are representative of this class, targeting both bacterial DNA gyrase and topoisomerase IV (Topo IV). In an abbreviated susceptibility screen, compounds 6o and 6v showed improved MIC90 values against Escherichia coli (0.5-1 µg/mL) and Acinetobacter baumannii (8-16 µg/mL) compared to the precursor compounds. In a murine septicemia model, both compounds showed complete protection in mice infected with a lethal dose of E. coli.


Asunto(s)
Antibacterianos/farmacología , ADN-Topoisomerasas de Tipo II/química , Descubrimiento de Drogas , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Sepsis/tratamiento farmacológico , Inhibidores de Topoisomerasa II/farmacología , Animales , Antibacterianos/química , Femenino , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Piridinas/química , Sepsis/microbiología , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/química
15.
Acta Crystallogr D Struct Biol ; 72(Pt 4): 488-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27050128

RESUMEN

Klebsiella pneumoniae is a Gram-negative bacterium that is responsible for a range of common infections, including pulmonary pneumonia, bloodstream infections and meningitis. Certain strains of Klebsiella have become highly resistant to antibiotics. Despite the vast amount of research carried out on this class of bacteria, the molecular structure of its topoisomerase IV, a type II topoisomerase essential for catalysing chromosomal segregation, had remained unknown. In this paper, the structure of its DNA-cleavage complex is reported at 3.35 Å resolution. The complex is comprised of ParC breakage-reunion and ParE TOPRIM domains of K. pneumoniae topoisomerase IV with DNA stabilized by levofloxacin, a broad-spectrum fluoroquinolone antimicrobial agent. This complex is compared with a similar complex from Streptococcus pneumoniae, which has recently been solved.


Asunto(s)
Proteínas Bacterianas/química , Topoisomerasa de ADN IV/química , Klebsiella pneumoniae/enzimología , Quinolonas/química , Streptococcus pneumoniae/enzimología , ADN Bacteriano/química
16.
J Med Chem ; 59(13): 6070-85, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27299569

RESUMEN

The underlying cause of spinal muscular atrophy (SMA) is a deficiency of the survival motor neuron (SMN) protein. Starting from hits identified in a high-throughput screening campaign and through structure-activity relationship investigations, we have developed small molecules that potently shift the alternative splicing of the SMN2 exon 7, resulting in increased production of the full-length SMN mRNA and protein. Three novel chemical series, represented by compounds 9, 14, and 20, have been optimized to increase the level of SMN protein by >50% in SMA patient-derived fibroblasts at concentrations of <160 nM. Daily administration of these compounds to severe SMA Δ7 mice results in an increased production of SMN protein in disease-relevant tissues and a significant increase in median survival time in a dose-dependent manner. Our work supports the development of an orally administered small molecule for the treatment of patients with SMA.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Atrofia Muscular Espinal/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Descubrimiento de Drogas , Exones/efectos de los fármacos , Células HEK293 , Humanos , Ratones Noqueados , Atrofia Muscular Espinal/genética , ARN Mensajero/genética , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Relación Estructura-Actividad , Proteína 2 para la Supervivencia de la Neurona Motora/genética
17.
Genetics ; 168(4): 1867-75, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15611163

RESUMEN

Phosphorylation of eukaryotic initiation factor-2 (eIF2) is an important mechanism mitigating cellular injury in response to diverse environmental stresses. While all eukaryotic organisms characterized to date contain an eIF2 kinase stress response pathway, the composition of eIF2 kinases differs, with mammals containing four distinct family members and the well-studied lower eukaryote Saccharomyces cerevisiae expressing only a single eIF2 kinase. We are interested in the mechanisms by which multiple eIF2 kinases interface with complex stress signals and elicit response pathways. In this report we find that in addition to two previously described eIF2 kinases related to mammalian HRI, designated Hri1p and Hri2p, the yeast Schizosaccharomyces pombe expresses a third eIF2 kinase, a Gcn2p ortholog. To delineate the roles of each eIF2 kinase, we constructed S. pombe strains expressing only a single eIF2 kinase gene or deleted for the entire eIF2 kinase family. We find that Hri2p is the primary activated eIF2 kinase in response to exposure to heat shock, arsenite, or cadmium. Gcn2p serves as the primary eIF2 kinase induced during a nutrient downshift, treatment with the amino acid biosynthetic inhibitor 3-aminotriazole, or upon exposure to high concentrations of sodium chloride. In one stress example, exposure to H(2)O(2), there is early tandem activation of both Hri2p and Gcn2p. Interestingly, with extended stress conditions there is activation of alternative secondary eIF2 kinases, suggesting that eukaryotes have mechanisms of coordinate activation of eIF2 kinase in their stress remediation responses. Deletion of these eIF2 kinases renders S. pombe more sensitive to many of these stress conditions.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Schizosaccharomyces/enzimología , eIF-2 Quinasa/metabolismo , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas de Saccharomyces cerevisiae , Schizosaccharomyces/genética , Cloruro de Sodio/metabolismo , eIF-2 Quinasa/genética
18.
Biochem J ; 380(Pt 2): 523-31, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-14989696

RESUMEN

The ubiquitous intracellular parasite Toxoplasma gondii (phylum Apicomplexa) differentiates into an encysted form (bradyzoite) that can repeatedly re-emerge as a life-threatening acute infection (tachyzoite) upon impairment of immunity. Since the switch from tachyzoite to bradyzoite is a stress-induced response, we sought to identify components related to the phosphorylation of the alpha subunit of eIF2 (eukaryotic initiation factor-2), a well-characterized event associated with stress remediation in other eukaryotic systems. In addition to characterizing Toxoplasma eIF2alpha (TgIF2alpha), we have discovered a novel eIF2 protein kinase, designated TgIF2K-A (Toxoplasma gondii initiation factor-2kinase). Although the catalytic domain of TgIF2K-A contains sequence and structural features that are conserved among members of the eIF2 kinase family, TgIF2K-A has an extended N-terminal region that is highly divergent from other eIF2 kinases. TgIF2K-A specifically phosphorylates the regulatory serine residue of yeast eIF2alpha in vitro and in vivo, and can modulate translation when expressed in the yeast model system. We also demonstrate that TgIF2K-A phosphorylates the analogous regulatory serine residue of recombinant TgIF2alpha in vitro. Finally, we demonstrate that TgIF2alpha phosphorylation in tachyzoites is enhanced in response to heat shock or alkaline stress, conditions known to induce parasite differentiation in vitro. Collectively, this study suggests that eIF2 kinase-mediated stress responses are conserved in Apicomplexa, and a novel family member exists that may control parasite-specific events, including the clinically relevant conversion into bradyzoite cysts.


Asunto(s)
Biosíntesis de Proteínas/fisiología , Toxoplasma/enzimología , eIF-2 Quinasa/fisiología , Secuencia de Aminoácidos/genética , Animales , Clonación Molecular/métodos , Factor 2 Eucariótico de Iniciación/química , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/fisiología , Metamorfosis Biológica/genética , Metamorfosis Biológica/fisiología , Datos de Secuencia Molecular , Péptido Sintasas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Protozoarias/química , Proteínas Protozoarias/fisiología , Alineación de Secuencia/métodos , Especificidad de la Especie , Toxoplasma/crecimiento & desarrollo , Toxoplasma/patogenicidad , eIF-2 Quinasa/química , eIF-2 Quinasa/genética
19.
Science ; 345(6197): 688-93, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25104390

RESUMEN

Spinal muscular atrophy (SMA) is a genetic disease caused by mutation or deletion of the survival of motor neuron 1 (SMN1) gene. A paralogous gene in humans, SMN2, produces low, insufficient levels of functional SMN protein due to alternative splicing that truncates the transcript. The decreased levels of SMN protein lead to progressive neuromuscular degeneration and high rates of mortality. Through chemical screening and optimization, we identified orally available small molecules that shift the balance of SMN2 splicing toward the production of full-length SMN2 messenger RNA with high selectivity. Administration of these compounds to Δ7 mice, a model of severe SMA, led to an increase in SMN protein levels, improvement of motor function, and protection of the neuromuscular circuit. These compounds also extended the life span of the mice. Selective SMN2 splicing modifiers may have therapeutic potential for patients with SMA.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Cumarinas/administración & dosificación , Isocumarinas/administración & dosificación , Longevidad/efectos de los fármacos , Atrofia Muscular Espinal/tratamiento farmacológico , Pirimidinonas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Administración Oral , Animales , Células Cultivadas , Cumarinas/química , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Isocumarinas/química , Ratones , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Pirimidinonas/química , ARN Mensajero/genética , Eliminación de Secuencia , Bibliotecas de Moléculas Pequeñas/química , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA