Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109.020
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 40: 1-14, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-34871102

RESUMEN

I've had serious misgivings about writing this article, because from living the experience day by day, it's hard to believe my accomplishments merit the attention. To skirt this roadblock, I forced myself to pretend I was in a conversation with my trainees, trying to distill the central driving forces of my career in science. The below chronicles my evolution from would-be astronaut/ballerina to budding developmental biologist to devoted T cell immunologist. It traces my work from a focus on intrathymic events that mold developing T cells into self-major histocompatibility complex (MHC)-restricted lymphocytes to extrathymic events that fine-tune the T cell receptor (TCR) repertoire and impose the finishing touches on T cell maturation. It is a story of a few personal attributes multiplied by generous mentors, good luck, hard work, perseverance, and knowing when to step down.


Asunto(s)
Complejo Mayor de Histocompatibilidad , Linfocitos T , Animales , Diferenciación Celular , Humanos , Timo
2.
Annu Rev Immunol ; 39: 251-277, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33556248

RESUMEN

The immune system of the central nervous system (CNS) consists primarily of innate immune cells. These are highly specialized macrophages found either in the parenchyma, called microglia, or at the CNS interfaces, such as leptomeningeal, perivascular, and choroid plexus macrophages. While they were primarily thought of as phagocytes, their function extends well beyond simple removal of cell debris during development and diseases. Brain-resident innate immune cells were found to be plastic, long-lived, and host to an outstanding number of risk genes for multiple pathologies. As a result, they are now considered the most suitable targets for modulating CNS diseases. Additionally, recent single-cell technologies enhanced our molecular understanding of their origins, fates, interactomes, and functional cell statesduring health and perturbation. Here, we review the current state of our understanding and challenges of the myeloid cell biology in the CNS and treatment options for related diseases.


Asunto(s)
Sistema Nervioso Central , Microglía , Animales , Encéfalo , Humanos , Macrófagos , Células Mieloides
3.
Annu Rev Immunol ; 38: 397-419, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31990620

RESUMEN

T cell development involves stepwise progression through defined stages that give rise to multiple T cell subtypes, and this is accompanied by the establishment of stage-specific gene expression. Changes in chromatin accessibility and chromatin modifications accompany changes in gene expression during T cell development. Chromatin-modifying enzymes that add or reverse covalent modifications to DNA and histones have a critical role in the dynamic regulation of gene expression throughout T cell development. As each chromatin-modifying enzyme has multiple family members that are typically all coexpressed during T cell development, their function is sometimes revealed only when two related enzymes are concurrently deleted. This work has also revealed that the biological effects of these enzymes often involve regulation of a limited set of targets. The growing diversity in the types and sites of modification, as well as the potential for a single enzyme to catalyze multiple modifications, is also highlighted.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Linfopoyesis , Linfocitos T/inmunología , Linfocitos T/metabolismo , Acetilación , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Histonas , Humanos , Linfopoyesis/genética , Linfopoyesis/inmunología , Metilación , Procesamiento Proteico-Postraduccional , Linfocitos T/citología , Linfocitos T/enzimología , Ubiquitinación
4.
Annu Rev Immunol ; 38: 229-247, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31928469

RESUMEN

Neonatal CD4+ and CD8+ T cells have historically been characterized as immature or defective. However, recent studies prompt a reinterpretation of the functions of neonatal T cells. Rather than a population of cells always falling short of expectations set by their adult counterparts, neonatal T cells are gaining recognition as a distinct population of lymphocytes well suited for the rapidly changing environment in early life. In this review, I will highlight new evidence indicating that neonatal T cells are not inert or less potent versions of adult T cells but instead are a broadly reactive layer of T cells poised to quickly develop into regulatory or effector cells, depending on the needs of the host. In this way, neonatal T cells are well adapted to provide fast-acting immune protection against foreign pathogens, while also sustaining tolerance to self-antigens.


Asunto(s)
Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Biomarcadores , Diferenciación Celular/inmunología , Interacciones Huésped-Patógeno , Humanos , Memoria Inmunológica , Activación de Linfocitos/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/citología
5.
Cell ; 187(1): 204-215.e14, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38070508

RESUMEN

Mounting evidence suggests metabolism instructs stem cell fate decisions. However, how fetal metabolism changes during development and how altered maternal metabolism shapes fetal metabolism remain unexplored. We present a descriptive atlas of in vivo fetal murine metabolism during mid-to-late gestation in normal and diabetic pregnancy. Using 13C-glucose and liquid chromatography-mass spectrometry (LC-MS), we profiled the metabolism of fetal brains, hearts, livers, and placentas harvested from pregnant dams between embryonic days (E)10.5 and 18.5. Our analysis revealed metabolic features specific to a hyperglycemic environment and signatures that may denote developmental transitions during euglycemic development. We observed sorbitol accumulation in fetal tissues and altered neurotransmitter levels in fetal brains isolated from hyperglycemic dams. Tracing 13C-glucose revealed disparate fetal nutrient sourcing depending on maternal glycemic states. Regardless of glycemic state, histidine-derived metabolites accumulated in late-stage fetal tissues. Our rich dataset presents a comprehensive overview of in vivo fetal tissue metabolism and alterations due to maternal hyperglycemia.


Asunto(s)
Diabetes Mellitus , Diabetes Gestacional , Feto , Animales , Femenino , Ratones , Embarazo , Diabetes Mellitus/metabolismo , Feto/metabolismo , Glucosa/metabolismo , Placenta/metabolismo , Diabetes Gestacional/metabolismo
6.
Cell ; 187(11): 2838-2854.e17, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38744282

RESUMEN

Retrospective lineage reconstruction of humans predicts that dramatic clonal imbalances in the body can be traced to the 2-cell stage embryo. However, whether and how such clonal asymmetries arise in the embryo is unclear. Here, we performed prospective lineage tracing of human embryos using live imaging, non-invasive cell labeling, and computational predictions to determine the contribution of each 2-cell stage blastomere to the epiblast (body), hypoblast (yolk sac), and trophectoderm (placenta). We show that the majority of epiblast cells originate from only one blastomere of the 2-cell stage embryo. We observe that only one to three cells become internalized at the 8-to-16-cell stage transition. Moreover, these internalized cells are more frequently derived from the first cell to divide at the 2-cell stage. We propose that cell division dynamics and a cell internalization bottleneck in the early embryo establish asymmetry in the clonal composition of the future human body.


Asunto(s)
Blastómeros , Linaje de la Célula , Embrión de Mamíferos , Femenino , Humanos , Blastómeros/citología , Blastómeros/metabolismo , División Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Masculino , Animales , Ratones
7.
Cell ; 187(19): 5431-5452.e20, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39303691

RESUMEN

Breastfeeding and microbial colonization during infancy occur within a critical time window for development, and both are thought to influence the risk of respiratory illness. However, the mechanisms underlying the protective effects of breastfeeding and the regulation of microbial colonization are poorly understood. Here, we profiled the nasal and gut microbiomes, breastfeeding characteristics, and maternal milk composition of 2,227 children from the CHILD Cohort Study. We identified robust colonization patterns that, together with milk components, predict preschool asthma and mediate the protective effects of breastfeeding. We found that early cessation of breastfeeding (before 3 months) leads to the premature acquisition of microbial species and functions, including Ruminococcus gnavus and tryptophan biosynthesis, which were previously linked to immune modulation and asthma. Conversely, longer exclusive breastfeeding supports a paced microbial development, protecting against asthma. These findings underscore the importance of extended breastfeeding for respiratory health and highlight potential microbial targets for intervention.


Asunto(s)
Lactancia Materna , Leche Humana , Humanos , Femenino , Leche Humana/microbiología , Lactante , Preescolar , Asma/microbiología , Asma/prevención & control , Asma/inmunología , Microbiota , Microbioma Gastrointestinal , Masculino , Estudios de Cohortes , Recién Nacido
8.
Cell ; 187(13): 3236-3248.e21, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38772369

RESUMEN

Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.


Asunto(s)
Redes Reguladoras de Genes , Análisis de la Célula Individual , Animales , Femenino , Humanos , Ratones , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Vectores Genéticos/metabolismo , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/citología , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Línea Celular , Transcripción Genética
9.
Cell ; 187(19): 5195-5216, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39303686

RESUMEN

Microorganisms, including bacteria, archaea, viruses, fungi, and protists, are essential to life on Earth and the functioning of the biosphere. Here, we discuss the key roles of microorganisms in achieving the United Nations Sustainable Development Goals (SDGs), highlighting recent and emerging advances in microbial research and technology that can facilitate our transition toward a sustainable future. Given the central role of microorganisms in the biochemical processing of elements, synthesizing new materials, supporting human health, and facilitating life in managed and natural landscapes, microbial research and technologies are directly or indirectly relevant for achieving each of the SDGs. More importantly, the ubiquitous and global role of microbes means that they present new opportunities for synergistically accelerating progress toward multiple sustainability goals. By effectively managing microbial health, we can achieve solutions that address multiple sustainability targets ranging from climate and human health to food and energy production. Emerging international policy frameworks should reflect the vital importance of microorganisms in achieving a sustainable future.


Asunto(s)
Desarrollo Sostenible , Humanos , Naciones Unidas , Objetivos , Bacterias/metabolismo , Salud Global , Hongos/metabolismo
10.
Cell ; 187(16): 4272-4288.e20, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39013469

RESUMEN

Vesicle trafficking is a fundamental process that allows for the sorting and transport of specific proteins (i.e., "cargoes") to different compartments of eukaryotic cells. Cargo recognition primarily occurs through coats and the associated proteins at the donor membrane. However, it remains unclear whether cargoes can also be selected at other stages of vesicle trafficking to further enhance the fidelity of the process. The WDR11-FAM91A1 complex functions downstream of the clathrin-associated AP-1 complex to facilitate protein transport from endosomes to the TGN. Here, we report the cryo-EM structure of human WDR11-FAM91A1 complex. WDR11 directly and specifically recognizes a subset of acidic clusters, which we term super acidic clusters (SACs). WDR11 complex assembly and its binding to SAC-containing proteins are indispensable for the trafficking of SAC-containing proteins and proper neuronal development in zebrafish. Our studies thus uncover that cargo proteins could be recognized in a sequence-specific manner downstream of a protein coat.


Asunto(s)
Microscopía por Crioelectrón , Transporte de Proteínas , Pez Cebra , Humanos , Animales , Endosomas/metabolismo , Células HEK293 , Células HeLa , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/química , Unión Proteica
11.
Cell ; 187(17): 4733-4750.e26, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38971152

RESUMEN

We identify a population of Protogenin-positive (PRTG+ve) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ, a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB.


Asunto(s)
Meduloblastoma , Células Madre Neoplásicas , Humanos , Meduloblastoma/patología , Meduloblastoma/metabolismo , Animales , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Ratones , Rombencéfalo/metabolismo , Rombencéfalo/embriología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Células Endoteliales/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Técnicas de Cocultivo , Estructuras Embrionarias , Metencéfalo/embriología
12.
Cell ; 187(8): 1936-1954.e24, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38490196

RESUMEN

Microglia are brain-resident macrophages that shape neural circuit development and are implicated in neurodevelopmental diseases. Multiple microglial transcriptional states have been defined, but their functional significance is unclear. Here, we identify a type I interferon (IFN-I)-responsive microglial state in the developing somatosensory cortex (postnatal day 5) that is actively engulfing whole neurons. This population expands during cortical remodeling induced by partial whisker deprivation. Global or microglial-specific loss of the IFN-I receptor resulted in microglia with phagolysosomal dysfunction and an accumulation of neurons with nuclear DNA damage. IFN-I gain of function increased neuronal engulfment by microglia in both mouse and zebrafish and restricted the accumulation of DNA-damaged neurons. Finally, IFN-I deficiency resulted in excess cortical excitatory neurons and tactile hypersensitivity. These data define a role for neuron-engulfing microglia during a critical window of brain development and reveal homeostatic functions of a canonical antiviral signaling pathway in the brain.


Asunto(s)
Encéfalo , Interferón Tipo I , Microglía , Animales , Ratones , Interferón Tipo I/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Pez Cebra , Encéfalo/citología , Encéfalo/crecimiento & desarrollo
13.
Cell ; 187(4): 962-980.e19, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38309258

RESUMEN

Microglia (MG), the brain-resident macrophages, play major roles in health and disease via a diversity of cellular states. While embryonic MG display a large heterogeneity of cellular distribution and transcriptomic states, their functions remain poorly characterized. Here, we uncovered a role for MG in the maintenance of structural integrity at two fetal cortical boundaries. At these boundaries between structures that grow in distinct directions, embryonic MG accumulate, display a state resembling post-natal axon-tract-associated microglia (ATM) and prevent the progression of microcavities into large cavitary lesions, in part via a mechanism involving the ATM-factor Spp1. MG and Spp1 furthermore contribute to the rapid repair of lesions, collectively highlighting protective functions that preserve the fetal brain from physiological morphogenetic stress and injury. Our study thus highlights key major roles for embryonic MG and Spp1 in maintaining structural integrity during morphogenesis, with major implications for our understanding of MG functions and brain development.


Asunto(s)
Encéfalo , Microglía , Axones , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Macrófagos/fisiología , Microglía/patología , Morfogénesis
14.
Cell ; 187(11): 2855-2874.e19, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38657603

RESUMEN

Progress in understanding early human development has been impeded by the scarcity of reference datasets from natural embryos, particularly those with spatial information during crucial stages like gastrulation. We conducted high-resolution spatial transcriptomics profiling on 38,562 spots from 62 transverse sections of an intact Carnegie stage (CS) 8 human embryo. From this spatial transcriptomic dataset, we constructed a 3D model of the CS8 embryo, in which a range of cell subtypes are identified, based on gene expression patterns and positional register, along the anterior-posterior, medial-lateral, and dorsal-ventral axis in the embryo. We further characterized the lineage trajectories of embryonic and extra-embryonic tissues and associated regulons and the regionalization of signaling centers and signaling activities that underpin lineage progression and tissue patterning during gastrulation. Collectively, the findings of this study provide insights into gastrulation and post-gastrulation development of the human embryo.


Asunto(s)
Embrión de Mamíferos , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Imagenología Tridimensional , Humanos , Embrión de Mamíferos/metabolismo , Transcriptoma/genética , Gástrula/metabolismo , Gástrula/embriología , Transducción de Señal , Linaje de la Célula , Perfilación de la Expresión Génica , Tipificación del Cuerpo/genética
15.
Cell ; 187(12): 3072-3089.e20, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38781967

RESUMEN

Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.


Asunto(s)
Matriz Extracelular , Mucosa Intestinal , Animales , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citología , Matriz Extracelular/metabolismo , Miosina Tipo II/metabolismo , Mesodermo/metabolismo , Mesodermo/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Morfogénesis , Metaloproteinasas de la Matriz/metabolismo
16.
Cell ; 187(13): 3303-3318.e18, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906101

RESUMEN

Gamete formation and subsequent offspring development often involve extended phases of suspended cellular development or even dormancy. How cells adapt to recover and resume growth remains poorly understood. Here, we visualized budding yeast cells undergoing meiosis by cryo-electron tomography (cryoET) and discovered elaborate filamentous assemblies decorating the nucleus, cytoplasm, and mitochondria. To determine filament composition, we developed a "filament identification" (FilamentID) workflow that combines multiscale cryoET/cryo-electron microscopy (cryoEM) analyses of partially lysed cells or organelles. FilamentID identified the mitochondrial filaments as being composed of the conserved aldehyde dehydrogenase Ald4ALDH2 and the nucleoplasmic/cytoplasmic filaments as consisting of acetyl-coenzyme A (CoA) synthetase Acs1ACSS2. Structural characterization further revealed the mechanism underlying polymerization and enabled us to genetically perturb filament formation. Acs1 polymerization facilitates the recovery of chronologically aged spores and, more generally, the cell cycle re-entry of starved cells. FilamentID is broadly applicable to characterize filaments of unknown identity in diverse cellular contexts.


Asunto(s)
Gametogénesis , Mitocondrias , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa/química , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Coenzima A Ligasas/metabolismo , Microscopía por Crioelectrón , Citoplasma/metabolismo , Tomografía con Microscopio Electrónico , Meiosis , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Esporas Fúngicas/metabolismo , Modelos Moleculares , Estructura Cuaternaria de Proteína
17.
Cell ; 187(14): 3726-3740.e43, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861993

RESUMEN

Many growth factors and cytokines signal by binding to the extracellular domains of their receptors and driving association and transphosphorylation of the receptor intracellular tyrosine kinase domains, initiating downstream signaling cascades. To enable systematic exploration of how receptor valency and geometry affect signaling outcomes, we designed cyclic homo-oligomers with up to 8 subunits using repeat protein building blocks that can be modularly extended. By incorporating a de novo-designed fibroblast growth factor receptor (FGFR)-binding module into these scaffolds, we generated a series of synthetic signaling ligands that exhibit potent valency- and geometry-dependent Ca2+ release and mitogen-activated protein kinase (MAPK) pathway activation. The high specificity of the designed agonists reveals distinct roles for two FGFR splice variants in driving arterial endothelium and perivascular cell fates during early vascular development. Our designed modular assemblies should be broadly useful for unraveling the complexities of signaling in key developmental transitions and for developing future therapeutic applications.


Asunto(s)
Diferenciación Celular , Factores de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos , Transducción de Señal , Animales , Humanos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Ratones , Ligandos , Calcio/metabolismo , Sistema de Señalización de MAP Quinasas
18.
Cell ; 187(7): 1666-1684.e26, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38490194

RESUMEN

Diminished hepatocyte regeneration is a key feature of acute and chronic liver diseases and after extended liver resections, resulting in the inability to maintain or restore a sufficient functional liver mass. Therapies to restore hepatocyte regeneration are lacking, making liver transplantation the only curative option for end-stage liver disease. Here, we report on the structure-based development and characterization (nuclear magnetic resonance [NMR] spectroscopy) of first-in-class small molecule inhibitors of the dual-specificity kinase MKK4 (MKK4i). MKK4i increased liver regeneration upon hepatectomy in murine and porcine models, allowed for survival of pigs in a lethal 85% hepatectomy model, and showed antisteatotic and antifibrotic effects in liver disease mouse models. A first-in-human phase I trial (European Union Drug Regulating Authorities Clinical Trials [EudraCT] 2021-000193-28) with the clinical candidate HRX215 was conducted and revealed excellent safety and pharmacokinetics. Clinical trials to probe HRX215 for prevention/treatment of liver failure after extensive oncological liver resections or after transplantation of small grafts are warranted.


Asunto(s)
Inhibidores Enzimáticos , Fallo Hepático , MAP Quinasa Quinasa 4 , Animales , Humanos , Ratones , Hepatectomía/métodos , Hepatocitos , Hígado , Hepatopatías/tratamiento farmacológico , Fallo Hepático/tratamiento farmacológico , Fallo Hepático/prevención & control , Regeneración Hepática , Porcinos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico
19.
Cell ; 187(17): 4690-4712.e30, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39142281

RESUMEN

Electrical excitability-the ability to fire and propagate action potentials-is a signature feature of neurons. How neurons become excitable during development and whether excitability is an intrinsic property of neurons remain unclear. Here, we demonstrate that Schwann cells, the most abundant glia in the peripheral nervous system, promote somatosensory neuron excitability during development. We find that Schwann cells secrete prostaglandin E2, which is necessary and sufficient to induce developing somatosensory neurons to express normal levels of genes required for neuronal function, including voltage-gated sodium channels, and to fire action potential trains. Inactivating this signaling pathway in Schwann cells impairs somatosensory neuron maturation, causing multimodal sensory defects that persist into adulthood. Collectively, our studies uncover a neurodevelopmental role for prostaglandin E2 distinct from its established role in inflammation, revealing a cell non-autonomous mechanism by which glia regulate neuronal excitability to enable the development of normal sensory functions.


Asunto(s)
Potenciales de Acción , Dinoprostona , Células de Schwann , Células Receptoras Sensoriales , Animales , Células de Schwann/metabolismo , Dinoprostona/metabolismo , Ratones , Células Receptoras Sensoriales/metabolismo , Transducción de Señal
20.
Cell ; 187(9): 2143-2157.e15, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38670072

RESUMEN

A central question for regenerative neuroscience is whether synthetic neural circuits, such as those built from two species, can function in an intact brain. Here, we apply blastocyst complementation to selectively build and test interspecies neural circuits. Despite approximately 10-20 million years of evolution, and prominent species differences in brain size, rat pluripotent stem cells injected into mouse blastocysts develop and persist throughout the mouse brain. Unexpectedly, the mouse niche reprograms the birth dates of rat neurons in the cortex and hippocampus, supporting rat-mouse synaptic activity. When mouse olfactory neurons are genetically silenced or killed, rat neurons restore information flow to odor processing circuits. Moreover, they rescue the primal behavior of food seeking, although less well than mouse neurons. By revealing that a mouse can sense the world using neurons from another species, we establish neural blastocyst complementation as a powerful tool to identify conserved mechanisms of brain development, plasticity, and repair.


Asunto(s)
Neuronas , Animales , Ratones , Ratas , Neuronas/metabolismo , Neuronas/citología , Neuronas/fisiología , Blastocisto/metabolismo , Blastocisto/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Encéfalo/citología , Encéfalo/fisiología , Femenino , Hipocampo/citología , Hipocampo/fisiología , Especificidad de la Especie , Ratones Endogámicos C57BL , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA