Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38701782

ABSTRACT

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Subject(s)
Epigenesis, Genetic , Myelin Sheath , Oligodendroglia , Remyelination , Animals , Myelin Sheath/metabolism , Humans , Mice , Remyelination/drug effects , Oligodendroglia/metabolism , Central Nervous System/metabolism , Mice, Inbred C57BL , Rejuvenation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Organoids/metabolism , Organoids/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/genetics , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Male , Regeneration/drug effects , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
2.
bioRxiv ; 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38370768

ABSTRACT

To investigate the co-development of vasculature, mesenchyme, and epithelium crucial for organogenesis and the acquisition of organ-specific characteristics, we constructed a human pluripotent stem cell-derived organoid system comprising lung or intestinal epithelium surrounded by organotypic mesenchyme and vasculature. We demonstrated the pivotal role of co-differentiating mesoderm and endoderm via precise BMP regulation in generating multilineage organoids and gut tube patterning. Single-cell RNA-seq analysis revealed organ specificity in endothelium and mesenchyme, and uncovered key ligands driving endothelial specification in the lung (e.g., WNT2B and Semaphorins) or intestine (e.g., GDF15). Upon transplantation under the kidney capsule in mice, these organoids further matured and developed perfusable human-specific sub-epithelial capillaries. Additionally, our model recapitulated the abnormal endothelial-epithelial crosstalk in patients with FOXF1 deletion or mutations. Multilineage organoids provide a unique platform to study developmental cues guiding endothelial and mesenchymal cell fate determination, and investigate intricate cell-cell communications in human organogenesis and disease. Highlights: BMP signaling fine-tunes the co-differentiation of mesoderm and endoderm.The cellular composition in multilineage organoids resembles that of human fetal organs.Mesenchyme and endothelium co-developed within the organoids adopt organ-specific characteristics.Multilineage organoids recapitulate abnormal endothelial-epithelial crosstalk in FOXF1-associated disorders.

3.
Transl Pediatr ; 12(4): 768-786, 2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37181016

ABSTRACT

Neurodevelopmental disability (NDD) is recognised as one of the most common comorbidities in children with congenital heart disease (CHD) and is associated with altered brain structure and growth throughout the life course. Causes and contributors underpinning the CHD and NDD paradigm are not fully understood, and likely include innate patient factors, such as genetic and epigenetic factors, prenatal haemodynamic consequences as a result of the heart defect, and factors affecting the fetal-placental-maternal environment, such as placental pathology, maternal diet, psychological stress and autoimmune disease. Additional postnatal factors, including the type and complexity of disease and other clinical factors such as prematurity, peri-operative factors and socioeconomic factors are also expected to play a role in determining the final presentation of the NDD. Despite significant advances in knowledge and strategies to optimise outcomes, the extent to which adverse neurodevelopment can be modified remains unknown. Understanding biological and structural phenotypes associated with NDD in CHD are vital for understanding disease mechanisms, which in turn will advance the development of effective intervention strategies for those at risk. This review article summarises our current knowledge surrounding biological, structural, and genetic contributors to NDD in CHD and describes avenues for future research; highlighting the need for translational studies that bridge the gap between basic science and clinical practice.

4.
Cell Stem Cell ; 30(5): 632-647.e10, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37146583

ABSTRACT

Schwann cells (SCs) are the primary glia of the peripheral nervous system. SCs are involved in many debilitating disorders, including diabetic peripheral neuropathy (DPN). Here, we present a strategy for deriving SCs from human pluripotent stem cells (hPSCs) that enables comprehensive studies of SC development, physiology, and disease. hPSC-derived SCs recapitulate the molecular features of primary SCs and are capable of in vitro and in vivo myelination. We established a model of DPN that revealed the selective vulnerability of SCs to high glucose. We performed a high-throughput screen and found that an antidepressant drug, bupropion, counteracts glucotoxicity in SCs. Treatment of hyperglycemic mice with bupropion prevents their sensory dysfunction, SC death, and myelin damage. Further, our retrospective analysis of health records revealed that bupropion treatment is associated with a lower incidence of neuropathy among diabetic patients. These results highlight the power of this approach for identifying therapeutic candidates for DPN.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Mice , Animals , Humans , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/etiology , Bupropion/therapeutic use , Retrospective Studies , Sciatic Nerve , Schwann Cells , Drug Discovery
5.
bioRxiv ; 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36909615

ABSTRACT

Brain-inspired hardware emulates the structure and working principles of a biological brain and may address the hardware bottleneck for fast-growing artificial intelligence (AI). Current brain-inspired silicon chips are promising but still limit their power to fully mimic brain function for AI computing. Here, we develop Brainoware , living AI hardware that harnesses the computation power of 3D biological neural networks in a brain organoid. Brain-like 3D in vitro cultures compute by receiving and sending information via a multielectrode array. Applying spatiotemporal electrical stimulation, this approach not only exhibits nonlinear dynamics and fading memory properties but also learns from training data. Further experiments demonstrate real-world applications in solving non-linear equations. This approach may provide new insights into AI hardware.

6.
Cells ; 11(3)2022 01 28.
Article in English | MEDLINE | ID: mdl-35159270

ABSTRACT

Advances in human pluripotent stem cell (hPSC) technology allow one to deconstruct the human body into specific disease-relevant cell types or create functional units representing various organs. hPSC-based models present a unique opportunity for the study of co-occurring disorders where "cause and effect" can be addressed. Poor neurodevelopmental outcomes have been reported in children with congenital heart diseases (CHD). Intuitively, abnormal cardiac function or surgical intervention may stunt the developing brain, leading to neurodevelopmental disorders (NDD). However, recent work has uncovered several genetic variants within genes associated with the development of both the heart and brain that could also explain this co-occurrence. Given the scalability of hPSCs, straightforward genetic modification, and established differentiation strategies, it is now possible to investigate both CHD and NDD as independent events. We will first overview the potential for shared genetics in both heart and brain development. We will then summarize methods to differentiate both cardiac & neural cells and organoids from hPSCs that represent the developmental process of the heart and forebrain. Finally, we will highlight strategies to rapidly screen several genetic variants together to uncover potential phenotypes and how therapeutic advances could be achieved by hPSC-based models.


Subject(s)
Heart Defects, Congenital , Neurodevelopmental Disorders , Pluripotent Stem Cells , Cell Differentiation/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Organoids/metabolism , Pluripotent Stem Cells/metabolism
8.
Cell Stem Cell ; 28(9): 1566-1581.e8, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33951478

ABSTRACT

The biological function and disease association of human endogenous retroviruses (HERVs) are largely elusive. HERV-K(HML-2) has been associated with neurotoxicity, but there is no clear understanding of its role or mechanistic basis. We addressed the physiological functions of HERV-K(HML-2) in neuronal differentiation using CRISPR engineering to activate or repress its expression levels in a human-pluripotent-stem-cell-based system. We found that elevated HERV-K(HML-2) transcription is detrimental for the development and function of cortical neurons. These effects are cell-type-specific, as dopaminergic neurons are unaffected. Moreover, high HERV-K(HML-2) transcription alters cortical layer formation in forebrain organoids. HERV-K(HML-2) transcriptional activation leads to hyperactivation of NTRK3 expression and other neurodegeneration-related genes. Direct activation of NTRK3 phenotypically resembles HERV-K(HML-2) induction, and reducing NTRK3 levels in context of HERV-K(HML-2) induction restores cortical neuron differentiation. Hence, these findings unravel a cell-type-specific role for HERV-K(HML-2) in cortical neuron development.


Subject(s)
Endogenous Retroviruses , Cell Differentiation , Humans , Transcriptional Activation
9.
Cancer Res ; 81(13): 3706-3716, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33941615

ABSTRACT

Fanconi anemia is an inherited genome instability syndrome characterized by interstrand cross-link hypersensitivity, congenital defects, bone marrow failure, and cancer predisposition. Although DNA repair mediated by Fanconi anemia genes has been extensively studied, how inactivation of these genes leads to specific cellular phenotypic consequences associated with Fanconi anemia is not well understood. Here we report that Fanconi anemia stem cells in the C. elegans germline and in murine embryos display marked nonhomologous end joining (NHEJ)-dependent radiation resistance, leading to survival of progeny cells carrying genetic lesions. In contrast, DNA cross-linking does not induce generational genomic instability in Fanconi anemia stem cells, as widely accepted, but rather drives NHEJ-dependent apoptosis in both species. These findings suggest that Fanconi anemia is a stem cell disease reflecting inappropriate NHEJ, which is mutagenic and carcinogenic as a result of DNA misrepair, while marrow failure represents hematopoietic stem cell apoptosis. SIGNIFICANCE: This study finds that Fanconi anemia stem cells preferentially activate error-prone NHEJ-dependent DNA repair to survive irradiation, thereby conferring generational genomic instability that is instrumental in carcinogenesis.


Subject(s)
Cesium Radioisotopes/adverse effects , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Embryonic Stem Cells/pathology , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/pathology , Genomic Instability , Animals , Apoptosis , Caenorhabditis elegans , DNA Repair , Embryonic Stem Cells/radiation effects , Fanconi Anemia/genetics , Fanconi Anemia/radiotherapy , Fanconi Anemia Complementation Group Proteins/genetics , Mice
10.
Nat Neurosci ; 24(3): 343-354, 2021 03.
Article in English | MEDLINE | ID: mdl-33558694

ABSTRACT

Aberrant inflammation in the CNS has been implicated as a major player in the pathogenesis of human neurodegenerative disease. We developed a new approach to derive microglia from human pluripotent stem cells (hPSCs) and built a defined hPSC-derived tri-culture system containing pure populations of hPSC-derived microglia, astrocytes, and neurons to dissect cellular cross-talk along the neuroinflammatory axis in vitro. We used the tri-culture system to model neuroinflammation in Alzheimer's disease with hPSCs harboring the APPSWE+/+ mutation and their isogenic control. We found that complement C3, a protein that is increased under inflammatory conditions and implicated in synaptic loss, is potentiated in tri-culture and further enhanced in APPSWE+/+ tri-cultures due to microglia initiating reciprocal signaling with astrocytes to produce excess C3. Our study defines the major cellular players contributing to increased C3 in Alzheimer's disease and presents a broadly applicable platform to study neuroinflammation in human disease.


Subject(s)
Alzheimer Disease/metabolism , Complement C3/metabolism , Microglia/metabolism , Pluripotent Stem Cells/pathology , Alzheimer Disease/pathology , Astrocytes/metabolism , Astrocytes/pathology , Hematopoiesis/physiology , Humans , Inflammation/metabolism , Inflammation/pathology , Microglia/pathology , Models, Biological , Neurons/metabolism , Neurons/pathology
11.
Cell Stem Cell ; 27(4): 508-510, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33007233

ABSTRACT

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.

12.
Cell Stem Cell ; 27(1): 35-49.e6, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619517

ABSTRACT

Autism is a clinically heterogeneous neurodevelopmental disorder characterized by impaired social interactions, restricted interests, and repetitive behaviors. Despite significant advances in the genetics of autism, understanding how genetic changes perturb brain development and affect clinical symptoms remains elusive. Here, we present a multiplex human pluripotent stem cell (hPSC) platform, in which 30 isogenic disease lines are pooled in a single dish and differentiated into prefrontal cortex (PFC) lineages to efficiently test early-developmental hypotheses of autism. We define subgroups of autism mutations that perturb PFC neurogenesis and are correlated to abnormal WNT/ßcatenin responses. Class 1 mutations (8 of 27) inhibit while class 2 mutations (5 of 27) enhance PFC neurogenesis. Remarkably, autism patient data reveal that individuals carrying subclass-specific mutations differ clinically in their corresponding language acquisition profiles. Our study provides a framework to disentangle genetic heterogeneity associated with autism and points toward converging molecular and developmental pathways of diverse autism-associated mutations.


Subject(s)
Autistic Disorder , Neurodevelopmental Disorders , Pluripotent Stem Cells , Autistic Disorder/genetics , Cell Differentiation/genetics , Humans , Neurogenesis
13.
Cell Stem Cell ; 25(1): 120-136.e10, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31155483

ABSTRACT

Current challenges in capturing naive human pluripotent stem cells (hPSCs) suggest that the factors regulating human naive versus primed pluripotency remain incompletely defined. Here we demonstrate that the widely used Essential 8 minimal medium (E8) captures hPSCs at a naive-to-primed intermediate state of pluripotency expressing several naive-like developmental, bioenergetic, and epigenomic features despite providing primed-state-sustaining growth factor conditions. Transcriptionally, E8 hPSCs are marked by activated lipid biosynthesis and suppressed MAPK/TGF-ß gene expression, resulting in endogenous ERK inhibition. These features are dependent on lipid-free culture conditions and are lost upon lipid exposure, whereas short-term pharmacological ERK inhibition restores naive-to-primed intermediate traits even in the presence of lipids. Finally, we identify de novo lipogenesis as a common transcriptional signature of E8 hPSCs and the pre-implantation human epiblast in vivo. These findings implicate exogenous lipid availability in regulating human pluripotency and define E8 hPSCs as a stable, naive-to-primed intermediate (NPI) pluripotent state.


Subject(s)
Blastocyst/cytology , Germ Layers/cytology , Pluripotent Stem Cells/physiology , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , Embryonic Stem Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lipid Metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
14.
Nat Biotechnol ; 37(4): 436-444, 2019 04.
Article in English | MEDLINE | ID: mdl-30936566

ABSTRACT

Human brain organoids generated with current technologies recapitulate histological features of the human brain, but they lack a reproducible topographic organization. During development, spatial topography is determined by gradients of signaling molecules released from discrete signaling centers. We hypothesized that introduction of a signaling center into forebrain organoids would specify the positional identity of neural tissue in a distance-dependent manner. Here, we present a system to trigger a Sonic Hedgehog (SHH) protein gradient in developing forebrain organoids that enables ordered self-organization along dorso-ventral and antero-posterior positional axes. SHH-patterned forebrain organoids establish major forebrain subdivisions that are positioned with in vivo-like topography. Consistent with its behavior in vivo, SHH exhibits long-range signaling activity in organoids. Finally, we use SHH-patterned cerebral organoids as a tool to study the role of cholesterol metabolism in SHH signaling. Together, this work identifies inductive signaling as an effective organizing strategy to recapitulate in vivo-like topography in human brain organoids.


Subject(s)
Hedgehog Proteins/metabolism , Organoids/growth & development , Organoids/metabolism , Prosencephalon/growth & development , Prosencephalon/metabolism , Animals , Biotechnology , Body Patterning , Cell Differentiation , Cholesterol/metabolism , Humans , Mice , Models, Neurological , Neural Stem Cells/metabolism , Neurogenesis , Organoids/cytology , Pluripotent Stem Cells/metabolism , Prosencephalon/cytology , Signal Transduction
15.
Nat Biotechnol ; 37(3): 267-275, 2019 03.
Article in English | MEDLINE | ID: mdl-30804533

ABSTRACT

The mechanistic basis of gliogenesis, which occurs late in human development, is poorly understood. Here we identify nuclear factor IA (NFIA) as a molecular switch inducing human glial competency. Transient expression of NFIA is sufficient to trigger glial competency of human pluripotent stem cell-derived neural stem cells within 5 days and to convert these cells into astrocytes in the presence of glial-promoting factors, as compared to 3-6 months using current protocols. NFIA-induced astrocytes promote synaptogenesis, exhibit neuroprotective properties, display calcium transients in response to appropriate stimuli and engraft in the adult mouse brain. Differentiation involves rapid but reversible chromatin remodeling, glial fibrillary acidic protein (GFAP) promoter demethylation and a striking lengthening of the G1 cell cycle phase. Genetic or pharmacological manipulation of G1 length partially mimics NFIA function. We used the approach to generate astrocytes with region-specific or reactive features. Our study defines key mechanisms of the gliogenic switch and enables the rapid production of human astrocytes for disease modeling and regenerative medicine.


Subject(s)
Cell Differentiation/genetics , NFI Transcription Factors/genetics , Neurogenesis/genetics , Pluripotent Stem Cells/metabolism , Animals , Astrocytes/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , NFI Transcription Factors/metabolism , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Promoter Regions, Genetic
16.
Proc Natl Acad Sci U S A ; 115(37): E8775-E8782, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30154162

ABSTRACT

Herpes simplex virus type 1 (HSV-1) encephalitis (HSE) is the most common sporadic viral encephalitis in Western countries. Some HSE children carry inborn errors of the Toll-like receptor 3 (TLR3)-dependent IFN-α/ß- and -λ-inducing pathway. Induced pluripotent stem cell (iPSC)-derived cortical neurons with TLR3 pathway mutations are highly susceptible to HSV-1, due to impairment of cell-intrinsic TLR3-IFN immunity. In contrast, the contribution of cell-intrinsic immunity of human trigeminal ganglion (TG) neurons remains unclear. Here, we describe efficient in vitro derivation and purification of TG neurons from human iPSCs via a cranial placode intermediate. The resulting TG neurons are of sensory identity and exhibit robust responses to heat (capsaicin), cold (icilin), and inflammatory pain (ATP). Unlike control cortical neurons, both control and TLR3-deficient TG neurons were highly susceptible to HSV-1. However, pretreatment of control TG neurons with poly(I:C) induced the cells into an anti-HSV-1 state. Moreover, both control and TLR3-deficient TG neurons developed resistance to HSV-1 following pretreatment with IFN-ß but not IFN-λ. These data indicate that TG neurons are vulnerable to HSV-1 because they require preemptive stimulation of the TLR3 or IFN-α/ß receptors to induce antiviral immunity, whereas cortical neurons possess a TLR3-dependent constitutive resistance that is sufficient to block incoming HSV-1 in the absence of prior antiviral signals. The lack of constitutive resistance in TG neurons in vitro is consistent with their exploitation as a latent virus reservoir in vivo. Our results incriminate deficiencies in the constitutive TLR3-dependent response of cortical neurons in the pathogenesis of HSE.


Subject(s)
Immunity/immunology , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Toll-Like Receptor 3/metabolism , Antiviral Agents/pharmacology , Cell Differentiation/genetics , Cells, Cultured , Cerebral Cortex/cytology , Child , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Humans , Immunity/genetics , Induced Pluripotent Stem Cells/cytology , Interferon-beta/pharmacology , Mutation , Neurons/drug effects , Neurons/virology , Poly I-C/pharmacology , Toll-Like Receptor 3/genetics , Trigeminal Ganglion/cytology
17.
Cell Stem Cell ; 21(3): 399-410.e7, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28886367

ABSTRACT

Directing the fate of human pluripotent stem cells (hPSCs) into different lineages requires variable starting conditions and components with undefined activities, introducing inconsistencies that confound reproducibility and assessment of specific perturbations. Here we introduce a simple, modular protocol for deriving the four main ectodermal lineages from hPSCs. By precisely varying FGF, BMP, WNT, and TGFß pathway activity in a minimal, chemically defined medium, we show parallel, robust, and reproducible derivation of neuroectoderm, neural crest (NC), cranial placode (CP), and non-neural ectoderm in multiple hPSC lines, on different substrates independently of cell density. We highlight the utility of this system by interrogating the role of TFAP2 transcription factors in ectodermal differentiation, revealing the importance of TFAP2A in NC and CP specification, and performing a small-molecule screen that identified compounds that further enhance CP differentiation. This platform provides a simple stage for systematic derivation of the entire range of ectodermal cell types.


Subject(s)
Cell Differentiation , Cell Lineage , Ectoderm/cytology , Pluripotent Stem Cells/cytology , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Cell Lineage/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Neural Crest/cytology , Neural Plate/cytology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Phenanthrolines/pharmacology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Transcription Factor AP-2/metabolism
18.
Nat Biotechnol ; 35(2): 154-163, 2017 02.
Article in English | MEDLINE | ID: mdl-28112759

ABSTRACT

Considerable progress has been made in converting human pluripotent stem cells (hPSCs) into functional neurons. However, the protracted timing of human neuron specification and functional maturation remains a key challenge that hampers the routine application of hPSC-derived lineages in disease modeling and regenerative medicine. Using a combinatorial small-molecule screen, we previously identified conditions to rapidly differentiate hPSCs into peripheral sensory neurons. Here we generalize the approach to central nervous system (CNS) fates by developing a small-molecule approach for accelerated induction of early-born cortical neurons. Combinatorial application of six pathway inhibitors induces post-mitotic cortical neurons with functional electrophysiological properties by day 16 of differentiation, in the absence of glial cell co-culture. The resulting neurons, transplanted at 8 d of differentiation into the postnatal mouse cortex, are functional and establish long-distance projections, as shown using iDISCO whole-brain imaging. Accelerated differentiation into cortical neuron fates should facilitate hPSC-based strategies for disease modeling and cell therapy in CNS disorders.


Subject(s)
Cell Differentiation/physiology , Central Nervous System Agents/administration & dosage , Neurons/cytology , Neurons/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Batch Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Pluripotent Stem Cells/drug effects
19.
Nat Med ; 22(12): 1421-1427, 2016 12.
Article in English | MEDLINE | ID: mdl-27841875

ABSTRACT

Familial dysautonomia (FD) is a debilitating disorder that affects derivatives of the neural crest (NC). For unknown reasons, people with FD show marked differences in disease severity despite carrying an identical, homozygous point mutation in IKBKAP, encoding IκB kinase complex-associated protein. Here we present disease-related phenotypes in human pluripotent stem cells (PSCs) that capture FD severity. Cells from individuals with severe but not mild disease show impaired specification of NC derivatives, including autonomic and sensory neurons. In contrast, cells from individuals with severe and mild FD show defects in peripheral neuron survival, indicating that neurodegeneration is the main culprit for cases of mild FD. Although genetic repair of the FD-associated mutation reversed early developmental NC defects, sensory neuron specification was not restored, indicating that other factors may contribute to disease severity. Whole-exome sequencing identified candidate modifier genes for individuals with severe FD. Our study demonstrates that PSC-based modeling is sensitive in recapitulating disease severity, which presents an important step toward personalized medicine.


Subject(s)
Autonomic Nervous System/physiopathology , Dysautonomia, Familial/physiopathology , Induced Pluripotent Stem Cells , Sensory Receptor Cells/cytology , Adolescent , Adult , Autonomic Nervous System/cytology , Autonomic Nervous System/growth & development , Carrier Proteins/genetics , Case-Control Studies , Cell Survival/genetics , Child , Dysautonomia, Familial/genetics , Female , Genotype , Humans , Male , Models, Neurological , Mutation , Neural Crest/cytology , Neurons/cytology , Phenotype , Sequence Analysis, DNA , Severity of Illness Index , Transcriptional Elongation Factors , Young Adult
20.
Nature ; 531(7592): 105-9, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26863197

ABSTRACT

The enteric nervous system (ENS) is the largest component of the autonomic nervous system, with neuron numbers surpassing those present in the spinal cord. The ENS has been called the 'second brain' given its autonomy, remarkable neurotransmitter diversity and complex cytoarchitecture. Defects in ENS development are responsible for many human disorders including Hirschsprung disease (HSCR). HSCR is caused by the developmental failure of ENS progenitors to migrate into the gastrointestinal tract, particularly the distal colon. Human ENS development remains poorly understood owing to the lack of an easily accessible model system. Here we demonstrate the efficient derivation and isolation of ENS progenitors from human pluripotent stem (PS) cells, and their further differentiation into functional enteric neurons. ENS precursors derived in vitro are capable of targeted migration in the developing chick embryo and extensive colonization of the adult mouse colon. The in vivo engraftment and migration of human PS-cell-derived ENS precursors rescue disease-related mortality in HSCR mice (Ednrb(s-l/s-l)), although the mechanism of action remains unclear. Finally, EDNRB-null mutant ENS precursors enable modelling of HSCR-related migration defects, and the identification of pepstatin A as a candidate therapeutic target. Our study establishes the first, to our knowledge, human PS-cell-based platform for the study of human ENS development, and presents cell- and drug-based strategies for the treatment of HSCR.


Subject(s)
Cell Lineage , Cell- and Tissue-Based Therapy , Drug Discovery/methods , Enteric Nervous System/pathology , Hirschsprung Disease/drug therapy , Hirschsprung Disease/pathology , Neurons/pathology , Aging , Animals , Cell Differentiation , Cell Line , Cell Movement , Cell Separation , Cell- and Tissue-Based Therapy/methods , Chick Embryo , Colon/drug effects , Colon/pathology , Disease Models, Animal , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Hirschsprung Disease/therapy , Humans , Male , Mice , Neurons/drug effects , Pepstatins/metabolism , Pluripotent Stem Cells/pathology , Receptor, Endothelin B/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...