Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Front Cell Neurosci ; 17: 1286552, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38145283

RESUMEN

Introduction: Social behavioral changes are a hallmark of several neurodevelopmental and neuropsychiatric conditions, nevertheless the underlying neural substrates of such dysfunction remain poorly understood. Building evidence points to the prefrontal cortex (PFC) as one of the key brain regions that orchestrates social behavior. We used this concept with the aim to develop a translational rat model of social-circuit dysfunction, the chronic PFC activation model (CPA). Methods: Chemogenetic designer receptor hM3Dq was used to induce chronic activation of the PFC over 10 days, and the behavioral and electrophysiological signatures of prolonged PFC hyperactivity were evaluated. To test the sensitivity of this model to pharmacological interventions on longer timescales, and validate its translational potential, the rats were treated with our novel highly selective oxytocin receptor (OXTR) agonist RO6958375, which is not activating the related vasopressin V1a receptor. Results: CPA rats showed reduced sociability in the three-chamber sociability test, and a concomitant decrease in neuronal excitability and synaptic transmission within the PFC as measured by electrophysiological recordings in acute slice preparation. Sub-chronic treatment with a low dose of the novel OXTR agonist following CPA interferes with the emergence of PFC circuit dysfunction, abnormal social behavior and specific transcriptomic changes. Discussion: These results demonstrate that sustained PFC hyperactivity modifies circuit characteristics and social behaviors in ways that can be modulated by selective OXTR activation and that this model may be used to understand the circuit recruitment of prosocial therapies in drug discovery.

2.
Neuron ; 111(17): 2660-2674.e9, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37385246

RESUMEN

Many RNA-binding proteins (RBPs), particularly those associated with RNA granules, promote pathological protein aggregation in neurodegenerative diseases. Here, we demonstrate that G3BP2, a core component of stress granules, directly interacts with Tau and inhibits Tau aggregation. In the human brain, the interaction of G3BP2 and Tau is dramatically increased in multiple tauopathies, and it is independent of neurofibrillary tangle (NFT) formation in Alzheimer's disease (AD). Surprisingly, Tau pathology is significantly elevated upon loss of G3BP2 in human neurons and brain organoids. Moreover, we found that G3BP2 masks the microtubule-binding region (MTBR) of Tau, thereby inhibiting Tau aggregation. Our study defines a novel role for RBPs as a line of defense against Tau aggregation in tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Humanos , Proteínas tau/metabolismo , Tauopatías/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
3.
Nat Commun ; 14(1): 2057, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045813

RESUMEN

Mutations in glucocerebrosidase cause the lysosomal storage disorder Gaucher's disease and are the most common risk factor for Parkinson's disease. Therapies to restore the enzyme's function in the brain hold great promise for treating the neurological implications. Thus, we developed blood-brain barrier penetrant therapeutic molecules by fusing transferrin receptor-binding moieties to ß-glucocerebrosidase (referred to as GCase-BS). We demonstrate that these fusion proteins show significantly increased uptake and lysosomal efficiency compared to the enzyme alone. In a cellular disease model, GCase-BS rapidly rescues the lysosomal proteome and lipid accumulations beyond known substrates. In a mouse disease model, intravenous injection of GCase-BS leads to a sustained reduction of glucosylsphingosine and can lower neurofilament-light chain plasma levels. Collectively, these findings demonstrate the potential of GCase-BS for treating GBA1-associated lysosomal dysfunction, provide insight into candidate biomarkers, and may ultimately open a promising treatment paradigm for lysosomal storage diseases extending beyond the central nervous system.


Asunto(s)
Enfermedad de Gaucher , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Lisosomas/metabolismo , Mutación , alfa-Sinucleína/metabolismo
4.
Reprod Toxicol ; 118: 108360, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36894038

RESUMEN

Risdiplam is a daily, orally dosed, survival of motor neuron 2 (SMN2) mRNA splicing-modifying agent approved for the treatment of spinal muscular atrophy (SMA). RG7800 is a closely related SMN2 mRNA-splicing compound. Effects on secondary mRNA splice targets such as Forkhead Box M1 (FOXM1) and MAP kinase-activating death domain protein (MADD), which have been implicated in cell-cycle regulation, were observed in non-clinical studies with both risdiplam and RG7800. Potential effects of risdiplam on male fertility via FOXM1 and MADD are important as these secondary splice targets exist in humans. This publication reports the findings from 14 in vivo studies that investigated the reproductive tissues of male animals in various stages of development. Exposure to risdiplam or RG7800 induced changes within the germ cells in the testes of male cynomolgus monkeys and rats. Germ-cell changes included both cell-cycle gene changes (alteration of mRNA-splicing variants) and seminiferous tubule degeneration. In monkeys treated with RG7800, there was no evidence of damage to spermatogonia. Observed testicular changes were stage-specific with spermatocytes in the pachytene stage of meiosis and were fully reversible in monkeys following a sufficient recovery period of eight weeks following cessation of RG7800. In rats, seminiferous tubule degeneration was present, and full reversibility of germ-cell degeneration in the testes was observed among half of the rats that were exposed to risdiplam or RG7800 and then allowed to recover. With these results, coupled with histopathological findings, the effects on the male reproductive system are expected to be reversible in humans for these types of SMN2 mRNA-splicing modifiers.


Asunto(s)
Compuestos Azo , Empalme del ARN , Animales , Masculino , Ratas , Compuestos Azo/farmacología , Compuestos Azo/uso terapéutico , Neuronas Motoras , ARN Mensajero/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética
5.
Nat Biomed Eng ; 6(11): 1248-1256, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36138193

RESUMEN

The safety of most human recombinant proteins can be evaluated in transgenic mice tolerant to specific human proteins. However, owing to insufficient genetic diversity and to fundamental differences in immune mechanisms, small-animal models of human diseases are often unsuitable for immunogenicity testing and for predicting adverse outcomes in human patients. Most human therapeutic antibodies trigger xenogeneic responses in wild-type animals and thus rapid clearance of the drugs, which makes in vivo toxicological testing of human antibodies challenging. Here we report the generation of Göttingen minipigs carrying a mini-repertoire of human genes for the immunoglobulin heavy chains γ1 and γ4 and the immunoglobulin light chain κ. In line with observations in human patients, the genetically modified minipigs tolerated the clinically non-immunogenic IgG1κ-isotype monoclonal antibodies daratumumab and bevacizumab, and elicited antibodies against the checkpoint inhibitor atezolizumab and the engineered interleukin cergutuzumab amunaleukin. The humanized minipigs can facilitate the safety and efficacy testing of therapeutic antibodies.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina , Ratones , Humanos , Animales , Porcinos , Porcinos Enanos , Cadenas Pesadas de Inmunoglobulina/genética , Proteínas Recombinantes , Ratones Transgénicos
6.
NAR Genom Bioinform ; 3(4): lqab102, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34761219

RESUMEN

Single-cell RNA sequencing (scRNA-seq) revolutionized our understanding of disease biology. The promise it presents to also transform translational research requires highly standardized and robust software workflows. Here, we present the toolkit Besca, which streamlines scRNA-seq analyses and their use to deconvolute bulk RNA-seq data according to current best practices. Beyond a standard workflow covering quality control, filtering, and clustering, two complementary Besca modules, utilizing hierarchical cell signatures and supervised machine learning, automate cell annotation and provide harmonized nomenclatures. Subsequently, the gene expression profiles can be employed to estimate cell type proportions in bulk transcriptomics data. Using multiple, diverse scRNA-seq datasets, some stemming from highly heterogeneous tumor tissue, we show how Besca aids acceleration, interoperability, reusability and interpretability of scRNA-seq data analyses, meeting crucial demands in translational research and beyond.

7.
Cell Rep Med ; 2(8): 100360, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34467244

RESUMEN

Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of maternal UBE3A, a ubiquitin protein ligase E3A. Here, we study neurons derived from patients with AS and neurotypical individuals, and reciprocally modulate UBE3A using antisense oligonucleotides. Unbiased proteomics reveal proteins that are regulated by UBE3A in a disease-specific manner, including PEG10, a retrotransposon-derived GAG protein. PEG10 protein increase, but not RNA, is dependent on UBE3A and proteasome function. PEG10 binds to both RNA and ataxia-associated proteins (ATXN2 and ATXN10), localizes to stress granules, and is secreted in extracellular vesicles, modulating vesicle content. Rescue of AS patient-derived neurons by UBE3A reinstatement or PEG10 reduction reveals similarity in transcriptome changes. Overexpression of PEG10 during mouse brain development alters neuronal migration, suggesting that it can affect brain development. These findings imply that PEG10 is a secreted human UBE3A target involved in AS pathophysiology.


Asunto(s)
Síndrome de Angelman/metabolismo , Síndrome de Angelman/fisiopatología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al ADN/metabolismo , Productos del Gen gag/química , Proteínas de Unión al ARN/metabolismo , Retroviridae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Movimiento Celular , Preescolar , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios Proteicos , Retroelementos/genética , Gránulos de Estrés/metabolismo , Gránulos de Estrés/ultraestructura , Transcriptoma/genética
8.
Front Immunol ; 12: 684430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34140954

RESUMEN

Microglia, the innate immune cells of the brain, are essential for maintaining homeostasis by their ramified, highly motile processes and for orchestrating the immune response to pathological stimuli. They are implicated in several neurodegenerative diseases like Alzheimer's and Parkinson's disease. One commonality of these diseases is their strong correlation with aging as the highest risk factor and studying age-related alterations in microglia physiology and associated signaling mechanism is indispensable for a better understanding of age-related pathomechanisms. CD22 has been identified as a modifier of microglia phagocytosis in a recent study, but not much is known about the function of CD22 in microglia. Here we show that CD22 surface levels are upregulated in aged versus adult microglia. Furthermore, in the amyloid mouse model PS2APP, Aß-containing microglia also exhibit increased CD22 signal. To assess the impact of CD22 blockage on microglia morphology and dynamics, we have established a protocol to image microglia process motility in acutely prepared brain slices from CX3CR1-GFP reporter mice. We observed a significant reduction of microglial ramification and surveillance capacity in brain slices from aged versus adult mice. The age-related decrease in surveillance can be restored by antibody-mediated CD22 blockage in aged mice, whereas surveillance in adult mice is not affected by CD22 inhibition. Moreover to complement the results obtained in mice, we show that human iPSC-derived macrophages exhibit an increased phagocytic capacity upon CD22 blockage. Downstream analysis of antibody-mediated CD22 inhibition revealed an influence on BMP and TGFß associated gene networks. Our results demonstrate CD22 as a broad age-associated modulator of microglia functionality with potential implications for neurodegenerative disorders.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Recuento de Células , Modelos Animales de Enfermedad , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Microglía/citología , Fagocitosis/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal
9.
Microb Genom ; 7(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33945454

RESUMEN

Understanding the dynamics and mechanisms of acquired drug resistance across major classes of antibiotics and bacterial pathogens is of critical importance for the optimization of current anti-infective therapies and the development of novel ones. To systematically address this challenge, we developed a workflow combining experimental evolution in a morbidostat continuous culturing device with deep genomic sequencing of population samples collected in time series. This approach was applied to the experimental evolution of six populations of Escherichia coli BW25113 towards acquiring resistance to triclosan (TCS), an antibacterial agent in various consumer products. This study revealed the rapid emergence and expansion (up to 100% in each culture within 4 days) of missense mutations in the fabI gene, encoding enoyl-acyl carrier protein reductase, the known TCS molecular target. A follow-up analysis of isolated clones showed that distinct amino acid substitutions increased the drug IC90 in a 3-16-fold range, reflecting their proximity to the TCS-binding site. In contrast to other antibiotics, efflux-upregulating mutations occurred only rarely and with low abundance. Mutations in several other genes were detected at an earlier stage of evolution. Most notably, three distinct amino acid substitutions were mapped in the C-terminal periplasmic domain of CadC protein, an acid stress-responsive transcriptional regulator. While these mutations do not confer robust TCS resistance, they appear to play a certain, yet unknown, role in adaptation to relatively low drug pressure. Overall, the observed evolutionary trajectories suggest that the FabI enzyme is the sole target of TCS (at least up to the ~50 µm level), and amino acid substitutions in the TCS-binding site represent the main mechanism of robust TCS resistance in E. coli. This model study illustrates the potential utility of the established morbidostat-based approach for uncovering resistance mechanisms and target identification for novel drug candidates with yet unknown mechanisms of action.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/genética , Farmacorresistencia Bacteriana/genética , Genómica , Triclosán/farmacología , Sustitución de Aminoácidos , Escherichia coli/genética , Genes Bacterianos/genética , Mutación , Secuenciación Completa del Genoma
10.
Reprod Toxicol ; 98: 286-298, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33147516

RESUMEN

Human induced pluripotent stem cells (hiPSC) were used to develop an assay format that may deliver information on teratogenicity of drugs. A human pluripotent stem cell scorecard panel was used to monitor the expression of 96 marker genes that are indicative of the stem cell state or differentiation into the ectoderm, mesoderm and endoderm lineages. We selected a human episomal iPS cell line for the assay based on karyotype stability, initial pluripotency, differentiation capacity and overall gene expression variability. The assay is based on embryoid body formation and was developed to be simply automated. In this proof of concept study, we used eight reference compounds (valproic acid, all-trans-retinoic acid, thalidomide, methotrexate, hydroxyurea, ascorbic acid, penicillin G and ibuprofen) to test the technical performance of the assay (readout stability) in concentration-response and time-course experiments. We also found that each compound affected marker gene expression in a different way. Various forms of data analysis identified 19 out of 96 early developmental genes as potential predictive markers for teratogenicity. Machine-learning models were run to exemplify how the assay will be developed further. The preliminary results from these analyses suggest that the assay could be suitable for the pre-screening of candidate pharmaceutical compounds. The approach presented here points a way towards development of a human cell-based assay that could replace the murine EST currently used to screen for early indications of potential teratogenicity of drug candidates.


Asunto(s)
Bioensayo/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Teratógenos/toxicidad , Pruebas de Toxicidad/métodos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Teratogénesis
11.
Drug Discov Today ; 25(3): 519-534, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31899257

RESUMEN

Here, we introduce models at three levels-molecular level, cellular and omics level, and organ and system level-that study drug mechanism and safety in preclinical drug discovery. The models differ in both their scope of study and technical details, but are all rooted in mathematical descriptions of complex biological systems, and all require informatics tools that handle large-volume, heterogeneous, and noisy data. We present principles and recent developments with examples at each level and highlight the synergy by a case study. We proffer a multiscale modelling view of drug discovery, call for a seamless flow of information in the form of models, and examine potential impacts.


Asunto(s)
Descubrimiento de Drogas/métodos , Modelos Biológicos , Modelos Teóricos , Animales , Simulación por Computador , Evaluación Preclínica de Medicamentos/métodos , Humanos , Modelos Moleculares
12.
Front Immunol ; 11: 617860, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613545

RESUMEN

Microglia are key in the homeostatic well-being of the brain and microglial dysfunction has been implicated in neurodegenerative disorders such as Alzheimer's disease (AD). Due to the many limitations to study microglia in situ or isolated for large scale drug discovery applications, there is a high need to develop robust and scalable human cellular models of microglia with reliable translatability to the disease. Here, we describe the generation of microglia-like cells from human induced pluripotent stem cells (iPSC) with distinct phenotypes for mechanistic studies in AD. We started out from an established differentiation protocol to generate primitive macrophage precursors mimicking the yolk sac ontogeny of microglia. Subsequently, we tested 36 differentiation conditions for the cells in monoculture where we exposed them to various combinations of media, morphogens, and extracellular matrices. The optimized protocol generated robustly ramified cells expressing key microglial markers. Bulk mRNA sequencing expression profiles revealed that compared to cells obtained in co-culture with neurons, microglia-like cells derived from a monoculture condition upregulate mRNA levels for Triggering Receptor Expressed On Myeloid Cells 2 (TREM2), which is reminiscent to the previously described disease-associated microglia. TREM2 is a risk gene for AD and an important regulator of microglia. The regulatory function of TREM2 in these cells was confirmed by comparing wild type with isogenic TREM2 knock-out iPSC microglia. The TREM2-deficient cells presented with stronger increase in free cytosolic calcium upon stimulation with ATP and ADP, as well as stronger migration towards complement C5a, compared to TREM2 expressing cells. The functional differences were associated with gene expression modulation of key regulators of microglia. In conclusion, we have established and validated a work stream to generate functional human iPSC-derived microglia-like cells by applying a directed and neuronal co-culture independent differentiation towards functional phenotypes in the context of AD. These cells can now be applied to study AD-related disease settings and to perform compound screening and testing for drug discovery.


Asunto(s)
Enfermedad de Alzheimer , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Receptores Inmunológicos/metabolismo , Diferenciación Celular , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
13.
Int J Mol Sci ; 20(24)2019 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-31835296

RESUMEN

The kinase AKT2 (PKB) is an important mediator of insulin signaling, for which loss-of-function knockout (KO) mutants lead to early onset diabetes mellitus, and dominant active mutations lead to early development of obesity and endothelial cell (EC) dysfunction. To model EC dysfunction, we used edited human pluripotent stem cells (hPSCs) that carried either a homozygous deletion of AKT2 (AKT2 KO) or a dominant active mutation (AKT2 E17K), which, along with the parental wild type (WT), were differentiated into ECs. Profiling of EC lines indicated an increase in proinflammatory and a reduction in anti-inflammatory fatty acids, an increase in inflammatory chemokines in cell supernatants, increased expression of proinflammatory genes, and increased binding to the EC monolayer in a functional leukocyte adhesion assay for both AKT2 KO and AKT2 E17K. Collectively, these findings suggest that vascular endothelial inflammation that results from dysregulated insulin signaling (homeostasis) may contribute to coronary artery disease, and that either downregulation or upregulation of the insulin pathway may lead to inflammation of endothelial cells. This suggests that the standard of care for patients must be expanded from control of metabolic parameters to include control of inflammation, such that endothelial dysfunction and cardiovascular disorders can ultimately be prevented.


Asunto(s)
Células Endoteliales/metabolismo , Edición Génica , Síndrome Metabólico , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/genética , Inflamación/metabolismo , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo
14.
Prog Med Chem ; 58: 119-156, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30879473

RESUMEN

Targeting RNA drastically expands our target space to therapeutically modulate numerous cellular processes implicated in human diseases. Of particular interest, drugging pre-mRNA splicing appears a very viable strategy; to control levels of splicing product by promoting the inclusion or exclusion of exons. After describing the concept of "splicing modulation", this chapter will cover the outstanding progress achieved in this field, by highlighting the breakthrough accomplished recently for the treatment of spinal muscular atrophy using two therapeutic modalities: splice switching oligonucleotides and small molecules. This review discusses the vital but feasible requirement for such drugs to deliver selectivity, and critical safety aspects are highlighted. Transformational medicines such as those developed to treat SMA are likely just the beginning of this story.


Asunto(s)
Atrofia Muscular Espinal/patología , Compuestos Azo/química , Compuestos Azo/uso terapéutico , Descubrimiento de Drogas , Fluorobencenos/química , Fluorobencenos/uso terapéutico , Humanos , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Oligonucleótidos/metabolismo , Oligonucleótidos/uso terapéutico , Pirimidinas/química , Pirimidinas/uso terapéutico , Empalme del ARN , Proteínas del Complejo SMN/genética , Proteínas del Complejo SMN/metabolismo
15.
Immunogenetics ; 71(2): 123-136, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30564855

RESUMEN

Safety and efficacy of therapeutic antibodies are often dependent on their interaction with Fc receptors for IgG (FcγRs). The Göttingen minipig represents a valuable species for biomedical research but its use in preclinical studies with therapeutic antibodies is hampered by the lack of knowledge about the porcine FcγRs. Genome analysis and sequencing now enabled the localization of the previously described FcγRIIIa in the orthologous location to human FCGR3A. In addition, we identified nearby the gene coding for the hitherto undescribed putative porcine FcγRIIa. The 1'241 bp long FCGR2A cDNA translates to a 274aa transmembrane protein containing an extracellular region with high similarity to human and cattle FcγRIIa. Like in cattle, the intracellular part does not contain an immunoreceptor tyrosine-based activation motif (ITAM) as in human FcγRIIa. Flow cytometry of the whole blood and single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) of Göttingen minipigs revealed the expression profile of all porcine FcγRs which is compared to human and mouse. The new FcγRIIa is mainly expressed on platelets making the minipig a good model to study IgG-mediated platelet activation and aggregation. In contrast to humans, minipig blood monocytes were found to express inhibitory FcγRIIb that could lead to the underestimation of FcγR-mediated effects of monocytes observed in minipig studies with therapeutic antibodies.


Asunto(s)
Receptores de IgG/genética , Porcinos Enanos/inmunología , Secuencia de Aminoácidos , Animales , Bovinos , Humanos , Ratones , Receptores de IgG/análisis , Receptores de IgG/química , Porcinos
16.
PLoS One ; 13(10): e0205589, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30304024

RESUMEN

Spinal muscular atrophy (SMA) is a severe genetic disorder that manifests in progressive neuromuscular degeneration. SMA originates from loss-of-function mutations of the SMN1 (Survival of Motor Neuron 1) gene. Recent evidence has implicated peripheral deficits, especially in skeletal muscle, as key contributors to disease progression in SMA. In this study we generated myogenic cells from two SMA-affected human embryonic stem cell (hESC) lines with deletion of SMN1 bearing two copies of the SMN2 gene and recapitulating the molecular phenotype of Type 1 SMA. We characterized myoblasts and myotubes by comparing them to two unaffected, control hESC lines and demonstrate that SMA myoblasts and myotubes showed altered expression of various myogenic markers, which translated into an impaired in vitro myogenic maturation and development process. Additionally, we provide evidence that these SMN1 deficient cells display functional deficits in cholinergic calcium signaling response, glycolysis and oxidative phosphorylation. Our data describe a novel human myogenic SMA model that might be used for interrogating the effect of SMN depletion during skeletal muscle development, and as model to investigate biological mechanisms targeting myogenic differentiation, mitochondrial respiration and calcium signaling processes in SMA muscle cells.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular Espinal/metabolismo , Mioblastos/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Línea Celular , Expresión Génica , Células Madre Embrionarias Humanas/patología , Humanos , Fibras Musculares Esqueléticas/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Mioblastos/patología , Receptores Colinérgicos/metabolismo , 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/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
17.
BMC Genomics ; 19(1): 558, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30060733

RESUMEN

After the publication of this work [1], a mistake was noticed in the Eq. 1. Given an m × n expression matrix with m genes and samples of n tissues, the correct definition of the Gini index for gene i is.

18.
J Med Chem ; 61(15): 6501-6517, 2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30044619

RESUMEN

SMA is an inherited disease that leads to loss of motor function and ambulation and a reduced life expectancy. We have been working to develop orally administrated, systemically distributed small molecules to increase levels of functional SMN protein. Compound 2 was the first SMN2 splicing modifier tested in clinical trials in healthy volunteers and SMA patients. It was safe and well tolerated and increased SMN protein levels up to 2-fold in patients. Nevertheless, its development was stopped as a precautionary measure because retinal toxicity was observed in cynomolgus monkeys after chronic daily oral dosing (39 weeks) at exposures in excess of those investigated in patients. Herein, we describe the discovery of 1 (risdiplam, RG7916, RO7034067) that focused on thorough pharmacology, DMPK and safety characterization and optimization. This compound is undergoing pivotal clinical trials and is a promising medicine for the treatment of patients in all ages and stages with SMA.


Asunto(s)
Compuestos Azo/farmacología , Descubrimiento de Drogas , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Pirimidinas/farmacología , Empalme del ARN/efectos de los fármacos , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Animales , Compuestos Azo/efectos adversos , Compuestos Azo/uso terapéutico , Humanos , Pirimidinas/efectos adversos , Pirimidinas/uso terapéutico , Seguridad
19.
Pharm Res ; 35(9): 169, 2018 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-29951887

RESUMEN

PURPOSE: Administration of therapeutic monoclonal antibodies (mAbs) is frequently accompanied by severe first infusion reactions (FIR). The mechanism driving FIR is still unclear. This study aimed to investigate the cellular and molecular mechanisms causing FIR in humanized mouse models and their potential for evaluating FIR risk in patients. METHODS: Mice humanized for Fc gamma receptors (FcγRs) were generated by recombination-mediated genomic replacement. Body temperature, cytokine release and reactive oxygen species (ROS) were measured to assess FIR to mAbs. RESULTS: Infusion of human mAb specific for mouse transferrin receptor (HamTfR) into FcγR-humanized mice, produced marked transient hypothermia accompanied by an increase in inflammatory cytokines KC and MIP-2, and ROS. FIR were dependent on administration route and Fc-triggered effector functions mediated by neutrophils. Human neutrophils also induced FIR in wild type mice infused with HamTfR. Specific knock-in mice demonstrated that human FcγRIIIb on neutrophils was both necessary and sufficient to cause FIR. FcγRIIIb-mediated FIR was abolished by depleting neutrophils or blocking FcγRIIIb with CD11b antibodies. CONCLUSIONS: Human FcγRIIIb and neutrophils are primarily responsible for triggering FIR. Clinical strategies to prevent FIR in patients should focus on this pathway and may include transient depletion of neutrophils or blocking FcγRIIIb with specific mAbs.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Hipotermia/inducido químicamente , Inflamación/inducido químicamente , Neutrófilos/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Humanos , Hipotermia/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/efectos de los fármacos , Receptores de IgG/genética , Receptores de Transferrina/inmunología
20.
Nat Commun ; 9(1): 2032, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29795225

RESUMEN

Modification of SMN2 exon 7 (E7) splicing is a validated therapeutic strategy against spinal muscular atrophy (SMA). However, a target-based approach to identify small-molecule E7 splicing modifiers has not been attempted, which could reveal novel therapies with improved mechanistic insight. Here, we chose as a target the stem-loop RNA structure TSL2, which overlaps with the 5' splicing site of E7. A small-molecule TSL2-binding compound, homocarbonyltopsentin (PK4C9), was identified that increases E7 splicing to therapeutic levels and rescues downstream molecular alterations in SMA cells. High-resolution NMR combined with molecular modelling revealed that PK4C9 binds to pentaloop conformations of TSL2 and promotes a shift to triloop conformations that display enhanced E7 splicing. Collectively, our study validates TSL2 as a target for small-molecule drug discovery in SMA, identifies a novel mechanism of action for an E7 splicing modifier, and sets a precedent for other splicing-mediated diseases where RNA structure could be similarly targeted.


Asunto(s)
Imidazoles/farmacología , Indoles/farmacología , Atrofia Muscular Espinal/tratamiento farmacológico , ARN Mensajero/metabolismo , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Drosophila , Evaluación Preclínica de Medicamentos , Exones/genética , Células HeLa , Humanos , Imidazoles/química , Imidazoles/uso terapéutico , Indoles/química , Indoles/uso terapéutico , Terapia Molecular Dirigida/métodos , Atrofia Muscular Espinal/genética , Fenotipo , Sitios de Empalme de ARN , ARN Mensajero/química , ARN Mensajero/genética , Elementos Reguladores de la Transcripción/efectos de los fármacos , Proteína 2 para la Supervivencia de la Neurona Motora/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA