Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nature ; 632(8024): 390-400, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048830

ABSTRACT

Most cases of herpes simplex virus 1 (HSV-1) encephalitis (HSE) remain unexplained1,2. Here, we report on two unrelated people who had HSE as children and are homozygous for rare deleterious variants of TMEFF1, which encodes a cell membrane protein that is preferentially expressed by brain cortical neurons. TMEFF1 interacts with the cell-surface HSV-1 receptor NECTIN-1, impairing HSV-1 glycoprotein D- and NECTIN-1-mediated fusion of the virus and the cell membrane, blocking viral entry. Genetic TMEFF1 deficiency allows HSV-1 to rapidly enter cortical neurons that are either patient specific or derived from CRISPR-Cas9-engineered human pluripotent stem cells, thereby enhancing HSV-1 translocation to the nucleus and subsequent replication. This cellular phenotype can be rescued by pretreatment with type I interferon (IFN) or the expression of exogenous wild-type TMEFF1. Moreover, ectopic expression of full-length TMEFF1 or its amino-terminal extracellular domain, but not its carboxy-terminal intracellular domain, impairs HSV-1 entry into NECTIN-1-expressing cells other than neurons, increasing their resistance to HSV-1 infection. Human TMEFF1 is therefore a host restriction factor for HSV-1 entry into cortical neurons. Its constitutively high abundance in cortical neurons protects these cells from HSV-1 infection, whereas inherited TMEFF1 deficiency renders them susceptible to this virus and can therefore underlie HSE.


Subject(s)
Brain , Encephalitis, Herpes Simplex , Herpesvirus 1, Human , Membrane Proteins , Virus Internalization , Animals , Female , Humans , Male , Brain/cytology , Brain/metabolism , Brain/virology , Encephalitis, Herpes Simplex/virology , Encephalitis, Herpes Simplex/metabolism , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Homozygote , Interferon Type I/metabolism , Interferon Type I/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nectins/genetics , Nectins/metabolism , Neurons/cytology , Neurons/metabolism , Neurons/virology , Pluripotent Stem Cells/cytology , Virus Replication , Child, Preschool , Young Adult , Pedigree
2.
Cell Stem Cell ; 31(8): 1162-1174.e8, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38917806

ABSTRACT

Aging is the biggest risk factor for the development of Alzheimer's disease (AD). Here, we performed a whole-genome CRISPR screen to identify regulators of neuronal age and show that the neddylation pathway regulates both cellular age and AD neurodegeneration in a human stem cell model. Specifically, we demonstrate that blocking neddylation increased cellular hallmarks of aging and led to an increase in Tau aggregation and phosphorylation in neurons carrying the APPswe/swe mutation. Aged APPswe/swe but not isogenic control neurons also showed a progressive decrease in viability. Selective neuronal loss upon neddylation inhibition was similarly observed in other isogenic AD and in Parkinson's disease (PD) models, including PSENM146V/M146V cortical and LRRK2G2019S/G2019S midbrain dopamine neurons, respectively. This study indicates that cellular aging can reveal late-onset disease phenotypes, identifies new potential targets to modulate AD progression, and describes a strategy to program age-associated phenotypes into stem cell models of disease.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cellular Senescence/genetics , Neurons/metabolism , Neurons/pathology , NEDD8 Protein/metabolism , NEDD8 Protein/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , tau Proteins/metabolism , tau Proteins/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/metabolism , Aging/genetics , Aging/pathology , Aging/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , CRISPR-Cas Systems/genetics
4.
Cell Rep ; 43(4): 114031, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38583153

ABSTRACT

Outer radial glia (oRG) emerge as cortical progenitor cells that support the development of an enlarged outer subventricular zone (oSVZ) and the expansion of the neocortex. The in vitro generation of oRG is essential to investigate the underlying mechanisms of human neocortical development and expansion. By activating the STAT3 signaling pathway using leukemia inhibitory factor (LIF), which is not expressed in guided cortical organoids, we define a cortical organoid differentiation method from human pluripotent stem cells (hPSCs) that recapitulates the expansion of a progenitor pool into the oSVZ. The oSVZ comprises progenitor cells expressing specific oRG markers such as GFAP, LIFR, and HOPX, closely matching human fetal oRG. Finally, incorporating neural crest-derived LIF-producing cortical pericytes into cortical organoids recapitulates the effects of LIF treatment. These data indicate that increasing the cellular complexity of the organoid microenvironment promotes the emergence of oRG and supports a platform to study oRG in hPSC-derived brain organoids routinely.


Subject(s)
Cell Differentiation , Lateral Ventricles , Leukemia Inhibitory Factor , Organoids , Pluripotent Stem Cells , Humans , Organoids/metabolism , Organoids/cytology , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/pharmacology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , STAT3 Transcription Factor/metabolism , Neuroglia/metabolism , Neuroglia/cytology , Signal Transduction
5.
Curr Opin Genet Dev ; 85: 102164, 2024 04.
Article in English | MEDLINE | ID: mdl-38412562

ABSTRACT

During brain development, the sequence of developmental steps and the underlying transcriptional regulatory logic are largely conserved across species. However, the temporal unfolding of developmental programs varies dramatically across species and within a given species varies across brain regions and cell identities. The maturation of neurons in the human cerebral cortex is particularly slow and lasts for many years compared with only a few weeks for the corresponding mouse neurons. The mechanisms setting the 'schedule' of neuronal maturation remain unclear but appear to be linked to a cell-intrinsic 'clock'. Here, we discuss recent findings that highlight a role for epigenetic factors in the timing of neuronal maturation. Manipulations of those factors in stem cell-based models can override the intrinsic pace of neuronal maturation, including its protracted nature in human cortical neurons. We then contextualize the epigenetic regulation of maturation programs with findings from other model systems and propose potential interactions between epigenetic pathways and other drivers of developmental rates.


Subject(s)
Epigenesis, Genetic , Neurons , Mice , Humans , Animals , Neurons/metabolism , Gene Expression Regulation , Brain/physiology , Neurogenesis/genetics
6.
Nature ; 626(8000): 881-890, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297124

ABSTRACT

The pace of human brain development is highly protracted compared with most other species1-7. The maturation of cortical neurons is particularly slow, taking months to years to develop adult functions3-5. Remarkably, such protracted timing is retained in cortical neurons derived from human pluripotent stem cells (hPSCs) during in vitro differentiation or upon transplantation into the mouse brain4,8,9. Those findings suggest the presence of a cell-intrinsic clock setting the pace of neuronal maturation, although the molecular nature of this clock remains unknown. Here we identify an epigenetic developmental programme that sets the timing of human neuronal maturation. First, we developed a hPSC-based approach to synchronize the birth of cortical neurons in vitro which enabled us to define an atlas of morphological, functional and molecular maturation. We observed a slow unfolding of maturation programmes, limited by the retention of specific epigenetic factors. Loss of function of several of those factors in cortical neurons enables precocious maturation. Transient inhibition of EZH2, EHMT1 and EHMT2 or DOT1L, at progenitor stage primes newly born neurons to rapidly acquire mature properties upon differentiation. Thus our findings reveal that the rate at which human neurons mature is set well before neurogenesis through the establishment of an epigenetic barrier in progenitor cells. Mechanistically, this barrier holds transcriptional maturation programmes in a poised state that is gradually released to ensure the prolonged timeline of human cortical neuron maturation.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells , Neural Stem Cells , Neurogenesis , Neurons , Adult , Animals , Humans , Mice , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Time Factors , Transcription, Genetic
7.
Nat Biotechnol ; 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38168993

ABSTRACT

The maturation of human pluripotent stem cell (hPSC)-derived neurons mimics the protracted timing of human brain development, extending over months to years for reaching adult-like function. Prolonged in vitro maturation presents a major challenge to stem cell-based applications in modeling and treating neurological disease. Therefore, we designed a high-content imaging assay based on morphological and functional readouts in hPSC-derived cortical neurons which identified multiple compounds that drive neuronal maturation including inhibitors of lysine-specific demethylase 1 and disruptor of telomerase-like 1 and activators of calcium-dependent transcription. A cocktail of four factors, GSK2879552, EPZ-5676, N-methyl-D-aspartate and Bay K 8644, collectively termed GENtoniK, triggered maturation across all parameters tested, including synaptic density, electrophysiology and transcriptomics. Maturation effects were further validated in cortical organoids, spinal motoneurons and non-neural lineages including melanocytes and pancreatic ß-cells. The effects on maturation observed across a broad range of hPSC-derived cell types indicate that some of the mechanisms controlling the timing of human maturation might be shared across lineages.

SELECTION OF CITATIONS
SEARCH DETAIL