Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Am J Sports Med ; 52(2): 503-515, 2024 02.
Article in English | MEDLINE | ID: mdl-38186352

ABSTRACT

BACKGROUND: The functional heterogeneity of culture-expanded mesenchymal stem cells (MSCs) has hindered the clinical application of MSCs. Previous studies have shown that MSC subpopulations with superior chondrogenic capacity can be isolated using a spiral microfluidic device based on the principle of inertial cell focusing. HYPOTHESIS: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function will overcome the challenge of the functional heterogeneity of expanded MSCs and will significantly improve MSC-based cartilage repair. STUDY DESIGN: Controlled laboratory study. METHODS: A next-generation, fully automated multidimensional double spiral microfluidic device was designed to provide more refined and efficient isolation of MSC subpopulations based on size. Analysis of in vitro chondrogenic potential and RNA sequencing was performed on size-sorted MSC subpopulations. In vivo cartilage repair efficacy was demonstrated in an osteochondral injury model in 12-week-old rats. Defects were implanted with MSC subpopulations (n = 6 per group) and compared with those implanted with unsegregated MSCs (n = 6). Osteochondral repair was assessed at 6 and 12 weeks after surgery by histological, micro-computed tomography, and mechanical analysis. RESULTS: A chondrogenic MSC subpopulation was efficiently isolated using the multidimensional double spiral device. RNA sequencing revealed distinct transcriptomic profiles and identified differential gene expression between subpopulations. The delivery of a chondrogenic MSC subpopulation resulted in improved cartilage repair, as indicated by histological scoring, the compression modulus, and micro-computed tomography of the subchondral bone. CONCLUSION: We have established a rapid, label-free, and reliable microfluidic protocol for more efficient size-based enrichment of a chondrogenic MSC subpopulation. Our proof-of-concept in vivo study demonstrates the enhanced cartilage repair efficacy of these enriched chondrogenic MSCs. CLINICAL RELEVANCE: The delivery of microfluidic-enriched chondrogenic MSCs that are consistent in size and function can overcome the challenge of the functional heterogeneity of expanded MSCs, resulting in significant improvement in MSC-based cartilage repair. The availability of such rapid, label-free enriched chondrogenic MSCs can enable better cell therapy products for cartilage repair with improved treatment outcomes.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Rats , Cartilage, Articular/surgery , Microfluidics , X-Ray Microtomography , Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis
2.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-36798312

ABSTRACT

Expansion microscopy (ExM) enables nanoscale imaging using a standard confocal microscope through the physical, isotropic expansion of fixed immunolabeled specimens. ExM is widely employed to image proteins, nucleic acids, and lipid membranes in single cells at nanoscale resolution; however, current methods cannot be performed in multi-well cell culture plates which limits the number of samples that can be processed simultaneously. We developed High-throughput Expansion Microscopy (HiExM), a robust platform that enables expansion microscopy of cells cultured in a standard 96-well plate. Our method enables consistent ~4.2x expansion within individual wells, across multiple wells, and between plates processed in parallel. We also demonstrate that HiExM can be combined with high-throughput confocal imaging platforms greatly improve the ease and scalability of image acquisition. As an example, we analyzed the effects of doxorubicin, a known cardiotoxic agent, in human cardiomyocytes (CMs) based on Hoechst signal intensity. We show a dose dependent effect on nuclear chromatin that is not observed in unexpanded CMs, suggesting that HiExM improves the detection of cellular phenotypes in response to drug treatment. Our method broadens the application of ExM as a tool for scalable super-resolution imaging in biological research applications.

3.
Stem Cell Res Ther ; 14(1): 259, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37726837

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) have broad potential as a cell therapy including for the treatment of drug-resistant inflammatory conditions with abnormal T cell proliferation such as graft-versus-host disease (GVHD). Clinical success, however, has been complicated by the heterogeneity of culture-expanded MSCs as well as donor variability. Here, we devise culture conditions that promote expansion of MSCs with enhanced immunomodulatory functions both in vitro and in animal models of GVHD. METHODS: Human bone marrow-derived MSCs were expanded at high-confluency (MSCHC) and low-confluency state (MSCLC). Their immunomodulatory properties were evaluated with in vitro co-culture assays based on suppression of activated T cell proliferation and secretion of pro-inflammatory cytokines from activated T cells. Metabolic state of these cells was determined, while RNA sequencing was performed to explore transcriptome of these MSCs. Ex vivo expanded MSCHC or MSCLC was injected into human peripheral blood mononuclear cells (PBMC)-induced GVHD mouse model to determine their in vivo therapeutic efficacy based on clinical grade scoring, human CD45+ blood count and histopathological examination. RESULTS: As compared to MSCLC, MSCHC significantly reduced both the proliferation of anti-CD3/CD28-activated T cells and secretion of pro-inflammatory cytokines upon MSCHC co-culture across several donors even in the absence of cytokine priming. Mechanistically, metabolic analysis of MSCHC prior to co-culture with activated T cells showed increased glycolytic metabolism and lactate secretion compared to MSCLC, consistent with their ability to inhibit T cell proliferation. Transcriptome analysis further revealed differential expression of immunomodulatory genes including TRIM29, BPIFB4, MMP3 and SPP1 in MSCHC as well as enriched pathways including cytokine-cytokine receptor interactions, cell adhesion and PI3K-AKT signalling. Lastly, we demonstrate in a human PBMC-induced GVHD mouse model that delivery of MSCHC showed greater suppression of inflammation and improved outcomes compared to MSCLC and saline controls. CONCLUSION: Our study provides evidence that ex vivo expansion of MSCs at high confluency alters the metabolic and transcriptomic states of these cells. Importantly, this approach maximizes the production of MSCs with enhanced immunomodulatory functions without priming, thus providing a non-invasive and generalizable strategy for improving the use of MSCs for the treatment of inflammatory diseases.


Subject(s)
Leukocytes, Mononuclear , Mesenchymal Stem Cells , Animals , Mice , Humans , Bone Marrow , Phosphatidylinositol 3-Kinases , Cytokines , Disease Models, Animal , DNA-Binding Proteins , Transcription Factors , Intercellular Signaling Peptides and Proteins
4.
Sci Adv ; 9(32): eadg9781, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37566656

ABSTRACT

Vascularization is driven by morphogen signals and mechanical cues that coordinately regulate cellular force generation, migration, and shape change to sculpt the developing vascular network. However, it remains unclear whether developing vasculature actively regulates its own mechanical properties to achieve effective vascularization. We engineered tissue constructs containing endothelial cells and fibroblasts to investigate the mechanics of vascularization. Tissue stiffness increases during vascular morphogenesis resulting from emergent interactions between endothelial cells, fibroblasts, and ECM and correlates with enhanced vascular function. Contractile cellular forces are key to emergent tissue stiffening and synergize with ECM mechanical properties to modulate the mechanics of vascularization. Emergent tissue stiffening and vascular function rely on mechanotransduction signaling within fibroblasts, mediated by YAP1. Mouse embryos lacking YAP1 in fibroblasts exhibit both reduced tissue stiffness and develop lethal vascular defects. Translating our findings through biology-inspired vascular tissue engineering approaches will have substantial implications in regenerative medicine.


Subject(s)
Endothelial Cells , Mechanotransduction, Cellular , Mice , Animals , Mechanotransduction, Cellular/physiology , Tissue Engineering/methods , Morphogenesis , Cell Differentiation , Extracellular Matrix
5.
Stem Cells Transl Med ; 12(5): 266-280, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36988042

ABSTRACT

Detection of cellular senescence is important quality analytics of cell therapy products, including mesenchymal stromal cells (MSCs). However, its detection is critically limited by the lack of specific markers and the destructive assays used to read out these markers. Here, we establish a rapid, live-cell assay for detecting senescent cells in heterogeneous mesenchymal stromal cell (MSC) cultures. We report that the T2 relaxation time measured by microscale Magnetic Resonance Relaxometry, which is related to intracellular iron accumulation, correlates strongly with senescence markers in MSC cultures under diverse conditions, including different passages and donors, size-sorted MSCs by inertial spiral microfluidic device, and drug-induced senescence. In addition, the live-cell and non-destructive method presented here has general applicability to other cells and tissues and can critically advance our understanding of cellular senescence.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells , Cell Proliferation , Cell- and Tissue-Based Therapy , Magnetic Resonance Spectroscopy , Cells, Cultured
6.
Sci Adv ; 7(46): eabk0271, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34767451

ABSTRACT

Stem cells are remarkably small. Whether small size is important for stem cell function is unknown. We find that hematopoietic stem cells (HSCs) enlarge under conditions known to decrease stem cell function. This decreased fitness of large HSCs is due to reduced proliferation and was accompanied by altered metabolism. Preventing HSC enlargement or reducing large HSCs in size averts the loss of stem cell potential under conditions causing stem cell exhaustion. Last, we show that murine and human HSCs enlarge during aging. Preventing this age-dependent enlargement improves HSC function. We conclude that small cell size is important for stem cell function in vivo and propose that stem cell enlargement contributes to their functional decline during aging.

7.
Nat Struct Mol Biol ; 28(5): 435-442, 2021 05.
Article in English | MEDLINE | ID: mdl-33972784

ABSTRACT

RNA polymerase II (RNAPII) pausing immediately downstream of the transcription start site is a critical rate-limiting step for the expression of most metazoan genes. During pause release, RNAPII encounters a highly conserved +1 H2A.Z nucleosome, yet how this histone variant contributes to transcription is poorly understood. Here, using an inducible protein degron system combined with genomic approaches and live cell super-resolution microscopy, we show that H2A.Z.1 modulates RNAPII dynamics across most genes in murine embryonic stem cells. Our quantitative analysis shows that H2A.Z.1 slows the rate of RNAPII pause release and consequently impacts negative elongation factor dynamics as well as nascent transcription. Consequently, H2A.Z.1 also impacts re-loading of the pre-initiation complex components TFIIB and TBP. Altogether, this work provides a critical mechanistic link between H2A.Z.1 and the proper induction of mammalian gene expression programs through the regulation of RNAPII dynamics and pause release.


Subject(s)
Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , RNA Polymerase II/metabolism , Animals , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Nucleosomes/metabolism , Transcription, Genetic
8.
Nature ; 585(7826): 603-608, 2020 09.
Article in English | MEDLINE | ID: mdl-32939090

ABSTRACT

Ferroptosis-an iron-dependent, non-apoptotic cell death process-is involved in various degenerative diseases and represents a targetable susceptibility in certain cancers1. The ferroptosis-susceptible cell state can either pre-exist in cells that arise from certain lineages or be acquired during cell-state transitions2-5. However, precisely how susceptibility to ferroptosis is dynamically regulated remains poorly understood. Here we use genome-wide CRISPR-Cas9 suppressor screens to identify the oxidative organelles peroxisomes as critical contributors to ferroptosis sensitivity in human renal and ovarian carcinoma cells. Using lipidomic profiling we show that peroxisomes contribute to ferroptosis by synthesizing polyunsaturated ether phospholipids (PUFA-ePLs), which act as substrates for lipid peroxidation that, in turn, results in the induction of ferroptosis. Carcinoma cells that are initially sensitive to ferroptosis can switch to a ferroptosis-resistant state in vivo in mice, which is associated with extensive downregulation of PUFA-ePLs. We further find that the pro-ferroptotic role of PUFA-ePLs can be extended beyond neoplastic cells to other cell types, including neurons and cardiomyocytes. Together, our work reveals roles for the peroxisome-ether-phospholipid axis in driving susceptibility to and evasion from ferroptosis, highlights PUFA-ePL as a distinct functional lipid class that is dynamically regulated during cell-state transitions, and suggests multiple regulatory nodes for therapeutic interventions in diseases that involve ferroptosis.


Subject(s)
Ethers/metabolism , Ferroptosis , Peroxisomes/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Ethers/chemistry , Female , Gene Editing , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lipid Peroxidation , Male , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peroxisomes/genetics
9.
Cell ; 178(5): 1115-1131.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442404

ABSTRACT

Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (ßOHB), distinguishes self-renewing Lgr5+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes ßOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous ßOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, ßOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through ßOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of ßOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury.


Subject(s)
Diet, High-Fat , Ketone Bodies/metabolism , Stem Cells/metabolism , 3-Hydroxybutyric Acid/blood , 3-Hydroxybutyric Acid/pharmacology , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Cell Self Renewal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxymethylglutaryl-CoA Synthase/deficiency , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Intestines/cytology , Intestines/pathology , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Young Adult
10.
Development ; 146(19)2019 10 09.
Article in English | MEDLINE | ID: mdl-31427288

ABSTRACT

Deciphering the genetic and epigenetic regulation of cardiomyocyte proliferation in organisms that are capable of robust cardiac renewal, such as zebrafish, represents an attractive inroad towards regenerating the human heart. Using integrated high-throughput transcriptional and chromatin analyses, we have identified a strong association between H3K27me3 deposition and reduced sarcomere and cytoskeletal gene expression in proliferative cardiomyocytes following cardiac injury in zebrafish. To move beyond an association, we generated an inducible transgenic strain expressing a mutant version of histone 3, H3.3K27M, that inhibits H3K27me3 catalysis in cardiomyocytes during the regenerative window. Hearts comprising H3.3K27M-expressing cardiomyocytes fail to regenerate, with wound edge cells showing heightened expression of structural genes and prominent sarcomeres. Although cell cycle re-entry was unperturbed, cytokinesis and wound invasion were significantly compromised. Collectively, our study identifies H3K27me3-mediated silencing of structural genes as requisite for zebrafish heart regeneration and suggests that repression of similar structural components in the border zone of an infarcted human heart might improve its regenerative capacity.


Subject(s)
Gene Silencing , Heart/physiology , Histones/metabolism , Lysine/metabolism , Regeneration/physiology , Zebrafish/genetics , Zebrafish/physiology , Animals , Cell Proliferation , Cytokinesis , Cytoskeleton/metabolism , Gene Expression Regulation, Developmental , Methylation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Sarcomeres/metabolism
11.
Cell Rep ; 24(5): 1342-1354.e5, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067987

ABSTRACT

Microdeletions involving TBX1 result in variable congenital malformations known collectively as 22q11.2 deletion syndrome (22q11.2DS). Tbx1-deficient mice and zebrafish recapitulate several disease phenotypes, including pharyngeal arch artery (PAA), head muscle (HM), and cardiac outflow tract (OFT) deficiencies. In zebrafish, these structures arise from nkx2.5+ progenitors in pharyngeal arches 2-6. Because pharyngeal arch morphogenesis is compromised in Tbx1-deficient animals, the malformations were considered secondary. Here, we report that the PAA, HM, and OFT phenotypes in tbx1 mutant zebrafish are primary and arise prior to pharyngeal arch morphogenesis from failed specification of the nkx2.5+ pharyngeal lineage. Through in situ analysis and lineage tracing, we reveal that nkx2.5 and tbx1 are co-expressed in this progenitor population. Furthermore, we present evidence suggesting that gdf3-ALK4 signaling is a downstream mediator of nkx2.5+ pharyngeal lineage specification. Collectively, these studies support a cellular mechanism potentially underlying the cardiovascular and craniofacial defects observed in the 22q11.2DS population.


Subject(s)
22q11 Deletion Syndrome/pathology , Cell Differentiation , Embryonic Stem Cells/cytology , Pharynx/embryology , 22q11 Deletion Syndrome/genetics , Animals , Cell Lineage , Embryonic Stem Cells/metabolism , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Pharynx/cytology , Phenotype , T-Box Domain Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Sci Rep ; 8(1): 4530, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540740

ABSTRACT

Microphysiological systems (MPSs) are in vitro models that capture facets of in vivo organ function through use of specialized culture microenvironments, including 3D matrices and microperfusion. Here, we report an approach to co-culture multiple different MPSs linked together physiologically on re-useable, open-system microfluidic platforms that are compatible with the quantitative study of a range of compounds, including lipophilic drugs. We describe three different platform designs - "4-way", "7-way", and "10-way" - each accommodating a mixing chamber and up to 4, 7, or 10 MPSs. Platforms accommodate multiple different MPS flow configurations, each with internal re-circulation to enhance molecular exchange, and feature on-board pneumatically-driven pumps with independently programmable flow rates to provide precise control over both intra- and inter-MPS flow partitioning and drug distribution. We first developed a 4-MPS system, showing accurate prediction of secreted liver protein distribution and 2-week maintenance of phenotypic markers. We then developed 7-MPS and 10-MPS platforms, demonstrating reliable, robust operation and maintenance of MPS phenotypic function for 3 weeks (7-way) and 4 weeks (10-way) of continuous interaction, as well as PK analysis of diclofenac metabolism. This study illustrates several generalizable design and operational principles for implementing multi-MPS "physiome-on-a-chip" approaches in drug discovery.


Subject(s)
Coculture Techniques/methods , Diclofenac/pharmacokinetics , Lab-On-A-Chip Devices , Liver/metabolism , Animals , Drug Evaluation, Preclinical , Humans , Microchip Analytical Procedures , Models, Biological , Phenotype , Rats
13.
PLoS One ; 12(3): e0172671, 2017.
Article in English | MEDLINE | ID: mdl-28333933

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are a promising platform for cardiac studies in vitro, and possibly for tissue repair in humans. However, hiPSC-CM cells tend to retain morphology, metabolism, patterns of gene expression, and electrophysiology similar to that of embryonic cardiomyocytes. We grew hiPSC-CM in patterned islands of different sizes and shapes, and measured the effect of island geometry on action potential waveform and calcium dynamics using optical recordings of voltage and calcium from 970 islands of different sizes. hiPSC-CM in larger islands showed electrical and calcium dynamics indicative of greater functional maturity. We then compared transcriptional signatures of the small and large islands against a developmental time course of cardiac differentiation. Although island size had little effect on expression of most genes whose levels differed between hiPSC-CM and adult primary CM, we identified a subset of genes for which island size drove the majority (58%) of the changes associated with functional maturation. Finally, we patterned hiPSC-CM on islands with a variety of shapes to probe the relative contributions of soluble factors, electrical coupling, and direct cell-cell contacts to the functional maturation. Collectively, our data show that optical electrophysiology is a powerful tool for assaying hiPSC-CM maturation, and that island size powerfully drives activation of a subset of genes involved in cardiac maturation.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Action Potentials/physiology , Calcium/metabolism , Cell Differentiation/genetics , Cells, Cultured , Electrophysiological Phenomena/physiology , Gene Expression/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Sequence Analysis, RNA/methods , Transcription, Genetic/genetics
14.
Mol Cell ; 64(1): 37-50, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27618485

ABSTRACT

Long non-coding RNAs (lncRNAs) are an emerging class of transcripts that can modulate gene expression; however, their mechanisms of action remain poorly understood. Here, we experimentally determine the secondary structure of Braveheart (Bvht) using chemical probing methods and show that this âˆ¼590 nt transcript has a modular fold. Using CRISPR/Cas9-mediated editing of mouse embryonic stem cells, we find that deletion of 11 nt in a 5' asymmetric G-rich internal loop (AGIL) of Bvht (bvhtdAGIL) dramatically impairs cardiomyocyte differentiation. We demonstrate a specific interaction between AGIL and cellular nucleic acid binding protein (CNBP/ZNF9), a zinc-finger protein known to bind single-stranded G-rich sequences. We further show that CNBP deletion partially rescues the bvhtdAGIL mutant phenotype by restoring differentiation capacity. Together, our work shows that Bvht functions with CNBP through a well-defined RNA motif to regulate cardiovascular lineage commitment, opening the door for exploring broader roles of RNA structure in development and disease.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Composition , Base Sequence , Binding Sites , CRISPR-Associated Protein 9 , Cell Differentiation , Cell Lineage/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/genetics , Endonucleases/metabolism , Gene Deletion , Gene Editing , Gene Expression Regulation , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Nucleic Acid Conformation , Phenotype , Protein Binding , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction
15.
Sci Adv ; 2(8): e1501429, 2016 08.
Article in English | MEDLINE | ID: mdl-27493991

ABSTRACT

Motor units are the fundamental elements responsible for muscle movement. They are formed by lower motor neurons and their muscle targets, synapsed via neuromuscular junctions (NMJs). The loss of NMJs in neurodegenerative disorders (such as amyotrophic lateral sclerosis or spinal muscle atrophy) or as a result of traumatic injuries affects millions of lives each year. Developing in vitro assays that closely recapitulate the physiology of neuromuscular tissues is crucial to understand the formation and maturation of NMJs, as well as to help unravel the mechanisms leading to their degeneration and repair. We present a microfluidic platform designed to coculture myoblast-derived muscle strips and motor neurons differentiated from mouse embryonic stem cells (ESCs) within a three-dimensional (3D) hydrogel. The device geometry mimics the spinal cord-limb physical separation by compartmentalizing the two cell types, which also facilitates the observation of 3D neurite outgrowth and remote muscle innervation. Moreover, the use of compliant pillars as anchors for muscle strips provides a quantitative functional readout of force generation. Finally, photosensitizing the ESC provides a pool of source cells that can be differentiated into optically excitable motor neurons, allowing for spatiodynamic, versatile, and noninvasive in vitro control of the motor units.


Subject(s)
Lab-On-A-Chip Devices , Motor Neurons/physiology , Muscle Fibers, Skeletal/physiology , Neuromuscular Junction/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Channelrhodopsins , Coculture Techniques , Gene Expression , Genes, Reporter , In Vitro Techniques , Mice , Motor Neurons/cytology , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Muscle Contraction/genetics , Muscle Fibers, Skeletal/cytology , Tissue Engineering
16.
PLoS Genet ; 12(5): e1006034, 2016 05.
Article in English | MEDLINE | ID: mdl-27149122

ABSTRACT

Failure of the human heart to maintain sufficient output of blood for the demands of the body, heart failure, is a common condition with high mortality even with modern therapeutic alternatives. To identify molecular determinants of mortality in patients with new-onset heart failure, we performed a meta-analysis of genome-wide association studies and follow-up genotyping in independent populations. We identified and replicated an association for a genetic variant on chromosome 5q22 with 36% increased risk of death in subjects with heart failure (rs9885413, P = 2.7x10-9). We provide evidence from reporter gene assays, computational predictions and epigenomic marks that this polymorphism increases activity of an enhancer region active in multiple human tissues. The polymorphism was further reproducibly associated with a DNA methylation signature in whole blood (P = 4.5x10-40) that also associated with allergic sensitization and expression in blood of the cytokine TSLP (P = 1.1x10-4). Knockdown of the transcription factor predicted to bind the enhancer region (NHLH1) in a human cell line (HEK293) expressing NHLH1 resulted in lower TSLP expression. In addition, we observed evidence of recent positive selection acting on the risk allele in populations of African descent. Our findings provide novel genetic leads to factors that influence mortality in patients with heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Methylation/genetics , Heart Failure/genetics , Receptors, Cytokine/genetics , Black or African American/genetics , Alleles , Basic Helix-Loop-Helix Transcription Factors/blood , Chromosomes, Human, Pair 5/genetics , Female , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genotype , HEK293 Cells , Heart Failure/blood , Heart Failure/mortality , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Cytokine/blood
17.
Elife ; 52016 05 10.
Article in English | MEDLINE | ID: mdl-27162171

ABSTRACT

Genetic variants identified by genome-wide association studies explain only a modest proportion of heritability, suggesting that meaningful associations lie 'hidden' below current thresholds. Here, we integrate information from association studies with epigenomic maps to demonstrate that enhancers significantly overlap known loci associated with the cardiac QT interval and QRS duration. We apply functional criteria to identify loci associated with QT interval that do not meet genome-wide significance and are missed by existing studies. We demonstrate that these 'sub-threshold' signals represent novel loci, and that epigenomic maps are effective at discriminating true biological signals from noise. We experimentally validate the molecular, gene-regulatory, cellular and organismal phenotypes of these sub-threshold loci, demonstrating that most sub-threshold loci have regulatory consequences and that genetic perturbation of nearby genes causes cardiac phenotypes in mouse. Our work provides a general approach for improving the detection of novel loci associated with complex human traits.


Subject(s)
Epigenomics , Genetic Loci , Genome-Wide Association Study , Heart Conduction System/physiology , Animals , Humans , Mice
18.
Hum Mol Genet ; 25(10): 2093-2103, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962151

ABSTRACT

The ST-segment and adjacent T-wave (ST-T wave) amplitudes of the electrocardiogram are quantitative characteristics of cardiac repolarization. Repolarization abnormalities have been linked to ventricular arrhythmias and sudden cardiac death. We performed the first genome-wide association meta-analysis of ST-T-wave amplitudes in up to 37 977 individuals identifying 71 robust genotype-phenotype associations clustered within 28 independent loci. Fifty-four genes were prioritized as candidates underlying the phenotypes, including genes with established roles in the cardiac repolarization phase (SCN5A/SCN10A, KCND3, KCNB1, NOS1AP and HEY2) and others with as yet undefined cardiac function. These associations may provide insights in the spatiotemporal contribution of genetic variation influencing cardiac repolarization and provide novel leads for future functional follow-up.


Subject(s)
Arrhythmias, Cardiac/genetics , Brugada Syndrome/genetics , Electrocardiography , Genetic Predisposition to Disease , Genome-Wide Association Study , Adaptor Proteins, Signal Transducing/genetics , Arrhythmias, Cardiac/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Brugada Syndrome/physiopathology , Cardiac Conduction System Disease , Death, Sudden, Cardiac/pathology , Female , Heart Conduction System/physiopathology , Humans , Male , NAV1.5 Voltage-Gated Sodium Channel/genetics , Polymorphism, Single Nucleotide/genetics , Repressor Proteins/genetics , Shab Potassium Channels/genetics , Shal Potassium Channels/genetics
19.
Cell Rep ; 14(5): 1142-1155, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26804911

ABSTRACT

Histone variant H2A.Z occupies the promoters of active and poised, bivalent genes in embryonic stem cells (ESCs) to regulate developmental programs, yet how it contributes to these contrasting states is poorly understood. Here, we investigate the function of H2A.Z.1 monoubiquitylation (H2A.Z.1ub) by mutation of the PRC1 target residues (H2A.Z.1(K3R3)). We show that H2A.Z.1(K3R3) is properly incorporated at target promoters in murine ESCs (mESCs), but loss of monoubiquitylation leads to de-repression of bivalent genes, loss of Polycomb binding, and faulty lineage commitment. Using quantitative proteomics, we find that tandem bromodomain proteins, including the BET family member BRD2, are enriched in H2A.Z.1 chromatin. We further show that BRD2 is gained at de-repressed promoters in H2A.Z.1(K3R3) mESCs, whereas BRD2 inhibition restores gene silencing at these sites. Together, our study reveals an antagonistic relationship between H2A.Z.1ub and BRD2 to regulate the transcriptional balance at bivalent genes to enable proper execution of developmental programs.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , Ubiquitination , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Self Renewal/genetics , Gene Expression Regulation, Developmental , Mice , Mouse Embryonic Stem Cells/cytology , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors
20.
Cell Rep ; 13(8): 1610-22, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586442

ABSTRACT

The histone variant H2A.Z is a hallmark of nucleosomes flanking promoters of protein-coding genes and is often found in nucleosomes that carry lysine 56-acetylated histone H3 (H3-K56Ac), a mark that promotes replication-independent nucleosome turnover. Here, we find that H3-K56Ac promotes RNA polymerase II occupancy at many protein-coding and noncoding loci, yet neither H3-K56Ac nor H2A.Z has a significant impact on steady-state mRNA levels in yeast. Instead, broad effects of H3-K56Ac or H2A.Z on RNA levels are revealed only in the absence of the nuclear RNA exosome. H2A.Z is also necessary for the expression of divergent, promoter-proximal noncoding RNAs (ncRNAs) in mouse embryonic stem cells. Finally, we show that H2A.Z functions with H3-K56Ac to facilitate formation of chromosome interaction domains (CIDs). Our study suggests that H2A.Z and H3-K56Ac work in concert with the RNA exosome to control mRNA and ncRNA expression, perhaps in part by regulating higher-order chromatin structures.


Subject(s)
Chromatin/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Homeostasis/genetics , Transcription, Genetic/genetics , Acetylation , Animals , Cells, Cultured , Chromosomes/genetics , DNA Replication/genetics , Embryonic Stem Cells/metabolism , Histones/genetics , Mice , Nucleosomes/genetics , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/genetics , RNA, Messenger/genetics , RNA, Untranslated/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...