Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 551
Filter
1.
Cell ; 181(6): 1307-1328.e15, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32502393

ABSTRACT

The view that sleep is essential for survival is supported by the ubiquity of this behavior, the apparent existence of sleep-like states in the earliest animals, and the fact that severe sleep loss can be lethal. The cause of this lethality is unknown. Here we show, using flies and mice, that sleep deprivation leads to accumulation of reactive oxygen species (ROS) and consequent oxidative stress, specifically in the gut. ROS are not just correlates of sleep deprivation but drivers of death: their neutralization prevents oxidative stress and allows flies to have a normal lifespan with little to no sleep. The rescue can be achieved with oral antioxidant compounds or with gut-targeted transgenic expression of antioxidant enzymes. We conclude that death upon severe sleep restriction can be caused by oxidative stress, that the gut is central in this process, and that survival without sleep is possible when ROS accumulation is prevented. VIDEO ABSTRACT.


Subject(s)
Gastrointestinal Tract/metabolism , Reactive Oxygen Species/metabolism , Sleep Deprivation/metabolism , Sleep/physiology , Animals , Antioxidants/metabolism , Drosophila , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Oxidative Stress/physiology
2.
Cell ; 171(5): 1151-1164.e16, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29056337

ABSTRACT

In mammals, the environment plays a critical role in promoting the final steps in neuronal development during the early postnatal period. While epigenetic factors are thought to contribute to this process, the underlying molecular mechanisms remain poorly understood. Here, we show that in the brain during early life, the DNA methyltransferase DNMT3A transiently binds across transcribed regions of lowly expressed genes, and its binding specifies the pattern of DNA methylation at CA sequences (mCA) within these genes. We find that DNMT3A occupancy and mCA deposition within the transcribed regions of genes is negatively regulated by gene transcription and may be modified by early-life experience. Once deposited, mCA is bound by the methyl-DNA-binding protein MECP2 and functions in a rheostat-like manner to fine-tune the cell-type-specific transcription of genes that are critical for brain function.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Epigenesis, Genetic , Neurons/metabolism , Animals , Brain/cytology , Brain/metabolism , DNA Methyltransferase 3A , Gene Expression Regulation, Developmental , Humans , Methyl-CpG-Binding Protein 2 , Mice , Transcription, Genetic , Transcriptional Activation
3.
Cell ; 165(7): 1570-1571, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27315474

ABSTRACT

Understanding how genes within cells, and cells within circuits, function together to produce the extraordinary repertoire of animal behaviors is arguably one of the most challenging undertakings in neuroscience. Two papers in this issue move toward this goal via 3D imaging of active neurons across the entire mouse brain.


Subject(s)
Brain , Neurons , Animals , Behavior, Animal , Neurosciences
4.
Immunity ; 54(9): 2072-2088.e7, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34320366

ABSTRACT

Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2+) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2-) macrophages. Herein, we identified an essential role for CCR2- macrophages in the chronically failing heart. Depletion of CCR2- macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2- macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4)-dependent pathway that controlled growth factor expression. These findings establish a role for tissue-resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Ventricular Remodeling/physiology , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Myocardium/metabolism , Troponin T/genetics
5.
Cell ; 161(7): 1496-8, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091031

ABSTRACT

Neuronal activity results in the rapid induction of gene transcription through a series of defined molecular events. Madabhushi et al. describe an unexpected role for the cutting of promoter DNA by topoisomerase IIB to facilitate transcription of activity-induced genes.


Subject(s)
DNA Breaks, Double-Stranded , Neurons/metabolism , Animals
6.
Cell ; 157(5): 1216-29, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855953

ABSTRACT

The nervous system adapts to experience by inducing a transcriptional program that controls important aspects of synaptic plasticity. Although the molecular mechanisms of experience-dependent plasticity are well characterized in excitatory neurons, the mechanisms that regulate this process in inhibitory neurons are only poorly understood. Here, we describe a transcriptional program that is induced by neuronal activity in inhibitory neurons. We find that, while neuronal activity induces expression of early-response transcription factors such as Npas4 in both excitatory and inhibitory neurons, Npas4 activates distinct programs of late-response genes in inhibitory and excitatory neurons. These late-response genes differentially regulate synaptic input to these two types of neurons, promoting inhibition onto excitatory neurons while inducing excitation onto inhibitory neurons. These findings suggest that the functional outcomes of activity-induced transcriptional responses are adapted in a cell-type-specific manner to achieve a circuit-wide homeostatic response.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Neurons/metabolism , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Culture Techniques , Embryo, Mammalian/cytology , Mice , Mice, Knockout , Synapses/metabolism
7.
Nature ; 620(7973): 366-373, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37468637

ABSTRACT

Neurons in the posterior parietal cortex contribute to the execution of goal-directed navigation1 and other decision-making tasks2-4. Although molecular studies have catalogued more than 50 cortical cell types5, it remains unclear what distinct functions they have in this area. Here we identified a molecularly defined subset of somatostatin (Sst) inhibitory neurons that, in the mouse posterior parietal cortex, carry a cell-type-specific error-correction signal for navigation. We obtained repeatable experimental access to these cells using an adeno-associated virus in which gene expression is driven by an enhancer that functions specifically in a subset of Sst cells6. We found that during goal-directed navigation in a virtual environment, this subset of Sst neurons activates in a synchronous pattern that is distinct from the activity of surrounding neurons, including other Sst neurons. Using in vivo two-photon photostimulation and ex vivo paired patch-clamp recordings, we show that nearby cells of this Sst subtype excite each other through gap junctions, revealing a self-excitation circuit motif that contributes to the synchronous activity of this cell type. These cells selectively activate as mice execute course corrections for deviations in their virtual heading during navigation towards a reward location, for both self-induced and experimentally induced deviations. We propose that this subtype of Sst neurons provides a self-reinforcing and cell-type-specific error-correction signal in the posterior parietal cortex that may help with the execution and learning of accurate goal-directed navigation trajectories.


Subject(s)
Neurons , Parietal Lobe , Animals , Mice , Learning , Neurons/metabolism , Parietal Lobe/cytology , Parietal Lobe/metabolism , Goals , Somatostatin/metabolism , Neural Inhibition , Spatial Navigation , Patch-Clamp Techniques , Gap Junctions/metabolism
8.
Nature ; 614(7949): 732-741, 2023 02.
Article in English | MEDLINE | ID: mdl-36792830

ABSTRACT

Neuronal activity is crucial for adaptive circuit remodelling but poses an inherent risk to the stability of the genome across the long lifespan of postmitotic neurons1-5. Whether neurons have acquired specialized genome protection mechanisms that enable them to withstand decades of potentially damaging stimuli during periods of heightened activity is unknown. Here we identify an activity-dependent DNA repair mechanism in which a new form of the NuA4-TIP60 chromatin modifier assembles in activated neurons around the inducible, neuronal-specific transcription factor NPAS4. We purify this complex from the brain and demonstrate its functions in eliciting activity-dependent changes to neuronal transcriptomes and circuitry. By characterizing the landscape of activity-induced DNA double-strand breaks in the brain, we show that NPAS4-NuA4 binds to recurrently damaged regulatory elements and recruits additional DNA repair machinery to stimulate their repair. Gene regulatory elements bound by NPAS4-NuA4 are partially protected against age-dependent accumulation of somatic mutations. Impaired NPAS4-NuA4 signalling leads to a cascade of cellular defects, including dysregulated activity-dependent transcriptional responses, loss of control over neuronal inhibition and genome instability, which all culminate to reduce organismal lifespan. In addition, mutations in several components of the NuA4 complex are reported to lead to neurodevelopmental and autism spectrum disorders. Together, these findings identify a neuronal-specific complex that couples neuronal activity directly to genome preservation, the disruption of which may contribute to developmental disorders, neurodegeneration and ageing.


Subject(s)
Brain , DNA Repair , Multiprotein Complexes , Neurons , Synapses , Basic Helix-Loop-Helix Transcription Factors , Brain/metabolism , DNA Breaks, Double-Stranded , Gene Expression Regulation , Lysine Acetyltransferase 5/metabolism , Multiprotein Complexes/metabolism , Neurons/metabolism , Synapses/metabolism , Mutation , Longevity/genetics , Genome , Aging/genetics , Neurodegenerative Diseases
9.
Nature ; 609(7926): 327-334, 2022 09.
Article in English | MEDLINE | ID: mdl-36002569

ABSTRACT

In the hippocampus, spatial maps are formed by place cells while contextual memories are thought to be encoded as engrams1-6. Engrams are typically identified by expression of the immediate early gene Fos, but little is known about the neural activity patterns that drive, and are shaped by, Fos expression in behaving animals7-10. Thus, it is unclear whether Fos-expressing hippocampal neurons also encode spatial maps and whether Fos expression correlates with and affects specific features of the place code11. Here we measured the activity of CA1 neurons with calcium imaging while monitoring Fos induction in mice performing a hippocampus-dependent spatial learning task in virtual reality. We find that neurons with high Fos induction form ensembles of cells with highly correlated activity, exhibit reliable place fields that evenly tile the environment and have more stable tuning across days than nearby non-Fos-induced cells. Comparing neighbouring cells with and without Fos function using a sparse genetic loss-of-function approach, we find that neurons with disrupted Fos function have less reliable activity, decreased spatial selectivity and lower across-day stability. Our results demonstrate that Fos-induced cells contribute to hippocampal place codes by encoding accurate, stable and spatially uniform maps and that Fos itself has a causal role in shaping these place codes. Fos ensembles may therefore link two key aspects of hippocampal function: engrams for contextual memories and place codes that underlie cognitive maps.


Subject(s)
Hippocampus , Proto-Oncogene Proteins c-fos , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Calcium/metabolism , Hippocampus/cytology , Hippocampus/physiology , Mice , Neurons/physiology , Place Cells/physiology , Proto-Oncogene Proteins c-fos/metabolism
10.
Mol Cell ; 77(2): 294-309.e9, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31784358

ABSTRACT

Mutations in the methyl-DNA-binding repressor protein MeCP2 cause the devastating neurodevelopmental disorder Rett syndrome. It has been challenging to understand how MeCP2 regulates transcription because MeCP2 binds broadly across the genome and MeCP2 mutations are associated with widespread small-magnitude changes in neuronal gene expression. We demonstrate here that MeCP2 represses nascent RNA transcription of highly methylated long genes in the brain through its interaction with the NCoR co-repressor complex. By measuring the rates of transcriptional initiation and elongation directly in the brain, we find that MeCP2 has no measurable effect on transcriptional elongation, but instead represses the rate at which Pol II initiates transcription of highly methylated long genes. These findings suggest a new model of MeCP2 function in which MeCP2 binds broadly across highly methylated regions of DNA, but acts at transcription start sites to attenuate transcriptional initiation.


Subject(s)
DNA Methylation/genetics , Methyl-CpG-Binding Protein 2/genetics , Repressor Proteins/genetics , Transcription, Genetic/genetics , Animals , Brain/physiology , DNA/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Neurons/physiology , RNA/genetics , Rett Syndrome/genetics
11.
Nature ; 590(7844): 115-121, 2021 02.
Article in English | MEDLINE | ID: mdl-33299180

ABSTRACT

Behavioural experiences activate the FOS transcription factor in sparse populations of neurons that are critical for encoding and recalling specific events1-3. However, there is limited understanding of the mechanisms by which experience drives circuit reorganization to establish a network of Fos-activated cells. It is also not known whether FOS is required in this process beyond serving as a marker of recent neural activity and, if so, which of its many gene targets underlie circuit reorganization. Here we demonstrate that when mice engage in spatial exploration of novel environments, perisomatic inhibition of Fos-activated hippocampal CA1 pyramidal neurons by parvalbumin-expressing interneurons is enhanced, whereas perisomatic inhibition by cholecystokinin-expressing interneurons is weakened. This bidirectional modulation of inhibition is abolished when the function of the FOS transcription factor complex is disrupted. Single-cell RNA-sequencing, ribosome-associated mRNA profiling and chromatin analyses, combined with electrophysiology, reveal that FOS activates the transcription of Scg2, a gene that encodes multiple distinct neuropeptides, to coordinate these changes in inhibition. As parvalbumin- and cholecystokinin-expressing interneurons mediate distinct features of pyramidal cell activity4-6, the SCG2-dependent reorganization of inhibitory synaptic input might be predicted to affect network function in vivo. Consistent with this prediction, hippocampal gamma rhythms and pyramidal cell coupling to theta phase are significantly altered in the absence of Scg2. These findings reveal an instructive role for FOS and SCG2 in establishing a network of Fos-activated neurons via the rewiring of local inhibition to form a selectively modulated state. The opposing plasticity mechanisms acting on distinct inhibitory pathways may support the consolidation of memories over time.


Subject(s)
Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Cholecystokinin/metabolism , Exploratory Behavior/physiology , Female , Gamma Rhythm , Interneurons/metabolism , Male , Memory Consolidation , Mice , Parvalbumins/metabolism , Pyramidal Cells/metabolism , Secretogranin II/genetics , Secretogranin II/metabolism , Spatial Navigation/physiology , Theta Rhythm
12.
Nature ; 583(7814): 115-121, 2020 07.
Article in English | MEDLINE | ID: mdl-32528180

ABSTRACT

The advent of endothermy, which is achieved through the continuous homeostatic regulation of body temperature and metabolism1,2, is a defining feature of mammalian and avian evolution. However, when challenged by food deprivation or harsh environmental conditions, many mammalian species initiate adaptive energy-conserving survival strategies-including torpor and hibernation-during which their body temperature decreases far below its homeostatic set-point3-5. How homeothermic mammals initiate and regulate these hypothermic states remains largely unknown. Here we show that entry into mouse torpor, a fasting-induced state with a greatly decreased metabolic rate and a body temperature as low as 20 °C6, is regulated by neurons in the medial and lateral preoptic area of the hypothalamus. We show that restimulation of neurons that were activated during a previous bout of torpor is sufficient to initiate the key features of torpor, even in mice that are not calorically restricted. Among these neurons we identify a population of glutamatergic Adcyap1-positive cells, the activity of which accurately determines when mice naturally initiate and exit torpor, and the inhibition of which disrupts the natural process of torpor entry, maintenance and arousal. Taken together, our results reveal a specific neuronal population in the mouse hypothalamus that serves as a core regulator of torpor. This work forms a basis for the future exploration of mechanisms and circuitry that regulate extreme hypothermic and hypometabolic states, and enables genetic access to monitor, initiate, manipulate and study these ancient adaptations of homeotherm biology.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/cytology , Neural Pathways/physiology , Neurons/physiology , Torpor/physiology , Animals , Fasting , Female , Food Deprivation , Glutamine/metabolism , Hypothalamus/physiology , Male , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
13.
Proc Natl Acad Sci U S A ; 120(44): e2310344120, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37871205

ABSTRACT

Mutations in MECP2 give rise to Rett syndrome (RTT), an X-linked neurodevelopmental disorder that results in broad cognitive impairments in females. While the exact etiology of RTT symptoms remains unknown, one possible explanation for its clinical presentation is that loss of MECP2 causes miswiring of neural circuits due to defects in the brain's capacity to respond to changes in neuronal activity and sensory experience. Here, we show that MeCP2 is phosphorylated at four residues in the mouse brain (S86, S274, T308, and S421) in response to neuronal activity, and we generate a quadruple knock-in (QKI) mouse line in which all four activity-dependent sites are mutated to alanines to prevent phosphorylation. QKI mice do not display overt RTT phenotypes or detectable gene expression changes in two brain regions. However, electrophysiological recordings from the retinogeniculate synapse of QKI mice reveal that while synapse elimination is initially normal at P14, it is significantly compromised at P20. Notably, this phenotype is distinct from the synapse refinement defect previously reported for Mecp2 null mice, where synapses initially refine but then regress after the third postnatal week. We thus propose a model in which activity-induced phosphorylation of MeCP2 is critical for the proper timing of retinogeniculate synapse maturation specifically during the early postnatal period.


Subject(s)
Methyl-CpG-Binding Protein 2 , Rett Syndrome , Female , Mice , Animals , Phosphorylation , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Brain/metabolism , Synapses/metabolism , Neurons/metabolism , Mice, Knockout , Disease Models, Animal
14.
Circulation ; 149(16): 1285-1297, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38235591

ABSTRACT

BACKGROUND: TTN truncation variants (TTNtvs) are the most common genetic lesion identified in individuals with dilated cardiomyopathy, a disease with high morbidity and mortality rates. TTNtvs reduce normal TTN (titin) protein levels, produce truncated proteins, and impair sarcomere content and function. Therapeutics targeting TTNtvs have been elusive because of the immense size of TTN, the rarity of specific TTNtvs, and incomplete knowledge of TTNtv pathogenicity. METHODS: We adapted CRISPR activation using dCas9-VPR to functionally interrogate TTNtv pathogenicity and develop a therapeutic in human cardiomyocytes and 3-dimensional cardiac microtissues engineered from induced pluripotent stem cell models harboring a dilated cardiomyopathy-associated TTNtv. We performed guide RNA screening with custom TTN reporter assays, agarose gel electrophoresis to quantify TTN protein levels and isoforms, and RNA sequencing to identify molecular consequences of TTN activation. Cardiomyocyte epigenetic assays were also used to nominate DNA regulatory elements to enable cardiomyocyte-specific TTN activation. RESULTS: CRISPR activation of TTN using single guide RNAs targeting either the TTN promoter or regulatory elements in spatial proximity to the TTN promoter through 3-dimensional chromatin interactions rescued TTN protein deficits disturbed by TTNtvs. Increasing TTN protein levels normalized sarcomere content and contractile function despite increasing truncated TTN protein. In addition to TTN transcripts, CRISPR activation also increased levels of myofibril assembly-related and sarcomere-related transcripts. CONCLUSIONS: TTN CRISPR activation rescued TTNtv-related functional deficits despite increasing truncated TTN levels, which provides evidence to support haploinsufficiency as a relevant genetic mechanism underlying heterozygous TTNtvs. CRISPR activation could be developed as a therapeutic to treat a large proportion of TTNtvs.


Subject(s)
Cardiomyopathy, Dilated , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/therapy , Cardiomyopathy, Dilated/pathology , Connectin/genetics , Haploinsufficiency/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , RNA, Guide, CRISPR-Cas Systems , Myocytes, Cardiac/metabolism
15.
Cell ; 143(3): 442-55, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-21029865

ABSTRACT

The mechanisms that promote excitatory synapse formation and maturation have been extensively studied. However, the molecular events that limit excitatory synapse development so that synapses form at the right time and place and in the correct numbers are less well understood. We have identified a RhoA guanine nucleotide exchange factor, Ephexin5, which negatively regulates excitatory synapse development until EphrinB binding to the EphB receptor tyrosine kinase triggers Ephexin5 phosphorylation, ubiquitination, and degradation. The degradation of Ephexin5 promotes EphB-dependent excitatory synapse development and is mediated by Ube3A, a ubiquitin ligase that is mutated in the human cognitive disorder Angelman syndrome and duplicated in some forms of Autism Spectrum Disorders (ASDs). These findings suggest that aberrant EphB/Ephexin5 signaling during the development of synapses may contribute to the abnormal cognitive function that occurs in Angelman syndrome and, possibly, ASDs.


Subject(s)
Synapses/metabolism , rhoA GTP-Binding Protein/metabolism , Angelman Syndrome/metabolism , Animals , Child , Child Development Disorders, Pervasive/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Embryo, Mammalian/metabolism , Gene Knockout Techniques , Humans , Mice , Rats , Rats, Long-Evans , Receptors, Eph Family/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , rhoA GTP-Binding Protein/genetics
16.
Cell ; 140(5): 704-16, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211139

ABSTRACT

Angelman Syndrome is a debilitating neurological disorder caused by mutation of the E3 ubiquitin ligase Ube3A, a gene whose mutation has also recently been associated with autism spectrum disorders (ASDs). The function of Ube3A during nervous system development and how Ube3A mutations give rise to cognitive impairment in individuals with Angleman Syndrome and ASDs are not clear. We report here that experience-driven neuronal activity induces Ube3A transcription and that Ube3A then regulates excitatory synapse development by controlling the degradation of Arc, a synaptic protein that promotes the internalization of the AMPA subtype of glutamate receptors. We find that disruption of Ube3A function in neurons leads to an increase in Arc expression and a concomitant decrease in the number of AMPA receptors at excitatory synapses. We propose that this deregulation of AMPA receptor expression at synapses may contribute to the cognitive dysfunction that occurs in Angelman Syndrome and possibly other ASDs.


Subject(s)
Angelman Syndrome/physiopathology , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Cognition , Humans , Mice , Mice, Knockout , Receptors, AMPA/metabolism , Synapses/metabolism , Ubiquitination
17.
Mol Cell ; 68(6): 1067-1082.e12, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29272704

ABSTRACT

Enhancer elements are genomic regulatory sequences that direct the selective expression of genes so that genetically identical cells can differentiate and acquire the highly specialized forms and functions required to build a functioning animal. To differentiate, cells must select from among the ∼106 enhancers encoded in the genome the thousands of enhancers that drive the gene programs that impart their distinct features. We used a genetic approach to identify transcription factors (TFs) required for enhancer selection in fibroblasts. This revealed that the broadly expressed, growth-factor-inducible TFs FOS/JUN (AP-1) play a central role in enhancer selection. FOS/JUN selects enhancers together with cell-type-specific TFs by collaboratively binding to nucleosomal enhancers and recruiting the SWI/SNF (BAF) chromatin remodeling complex to establish accessible chromatin. These experiments demonstrate how environmental signals acting via FOS/JUN and BAF coordinate with cell-type-specific TFs to select enhancer repertoires that enable differentiation during development.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic , Proto-Oncogene Proteins c-fos/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Expression Regulation, Neoplastic , Male , Mice, Inbred C57BL , Mice, Knockout , Nucleosomes , Promoter Regions, Genetic , Transcription Factors/genetics
18.
J Biol Chem ; 299(5): 104631, 2023 05.
Article in English | MEDLINE | ID: mdl-36963494

ABSTRACT

For decades, sarcomeric myosin heavy chain proteins were assumed to be restricted to striated muscle where they function as molecular motors that contract muscle. However, MYH7b, an evolutionarily ancient member of this myosin family, has been detected in mammalian nonmuscle tissues, and mutations in MYH7b are linked to hereditary hearing loss in compound heterozygous patients. These mutations are the first associated with hearing loss rather than a muscle pathology, and because there are no homologous mutations in other myosin isoforms, their functional effects were unknown. We generated recombinant human MYH7b harboring the D515N or R1651Q hearing loss-associated mutation and studied their effects on motor activity and structural and assembly properties, respectively. The D515N mutation had no effect on steady-state actin-activated ATPase rate or load-dependent detachment kinetics but increased actin sliding velocity because of an increased displacement during the myosin working stroke. Furthermore, we found that the D515N mutation caused an increase in the proportion of myosin heads that occupy the disordered-relaxed state, meaning more myosin heads are available to interact with actin. Although we found no impact of the R1651Q mutation on myosin rod secondary structure or solubility, we observed a striking aggregation phenotype when this mutation was introduced into nonmuscle cells. Our results suggest that each mutation independently affects MYH7b function and structure. Together, these results provide the foundation for further study of a role for MYH7b outside the sarcomere.


Subject(s)
Hearing Loss , Myosin Heavy Chains , Animals , Humans , Mice , Actins/metabolism , Cell Line , Chlorocebus aethiops , COS Cells , Hearing Loss/genetics , Hearing Loss/physiopathology , Kinetics , Mutation , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Aggregates/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
J Biol Chem ; 299(1): 102657, 2023 01.
Article in English | MEDLINE | ID: mdl-36334627

ABSTRACT

Myosin heavy chain 7b (MYH7b) is an evolutionarily ancient member of the sarcomeric myosin family, which typically supports striated muscle function. However, in mammals, alternative splicing prevents MYH7b protein production in cardiac and most skeletal muscles and limits expression to a subset of specialized muscles and certain nonmuscle environments. In contrast, MYH7b protein is abundant in python cardiac and skeletal muscles. Although the MYH7b expression pattern diverges in mammals versus reptiles, MYH7b shares high sequence identity across species. So, it remains unclear how mammalian MYH7b function may differ from that of other sarcomeric myosins and whether human and python MYH7b motor functions diverge as their expression patterns suggest. Thus, we generated recombinant human and python MYH7b protein and measured their motor properties to investigate any species-specific differences in activity. Our results reveal that despite having similar working strokes, the MYH7b isoforms have slower actin-activated ATPase cycles and actin sliding velocities than human cardiac ß-MyHC. Furthermore, python MYH7b is tuned to have slower motor activity than human MYH7b because of slower kinetics of the chemomechanical cycle. We found that the MYH7b isoforms adopt a higher proportion of myosin heads in the ultraslow, super-relaxed state compared with human cardiac ß-MyHC. These findings are supported by molecular dynamics simulations that predict MYH7b preferentially occupies myosin active site conformations similar to those observed in the structurally inactive state. Together, these results suggest that MYH7b is specialized for slow and energy-conserving motor activity and that differential tuning of MYH7b orthologs contributes to species-specific biological roles.


Subject(s)
Cardiac Myosins , Muscle, Skeletal , Myosin Heavy Chains , Animals , Humans , Mammals/metabolism , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism
20.
PLoS Pathog ; 18(7): e1010716, 2022 07.
Article in English | MEDLINE | ID: mdl-35877772

ABSTRACT

Pseudomonas syringae is a genetically diverse bacterial species complex responsible for numerous agronomically important crop diseases. Individual P. syringae isolates are assigned pathovar designations based on their host of isolation and the associated disease symptoms, and these pathovar designations are often assumed to reflect host specificity although this assumption has rarely been rigorously tested. Here we developed a rapid seed infection assay to measure the virulence of 121 diverse P. syringae isolates on common bean (Phaseolus vulgaris). This collection includes P. syringae phylogroup 2 (PG2) bean isolates (pathovar syringae) that cause bacterial spot disease and P. syringae phylogroup 3 (PG3) bean isolates (pathovar phaseolicola) that cause the more serious halo blight disease. We found that bean isolates in general were significantly more virulent on bean than non-bean isolates and observed no significant virulence difference between the PG2 and PG3 bean isolates. However, when we compared virulence within PGs we found that PG3 bean isolates were significantly more virulent than PG3 non-bean isolates, while there was no significant difference in virulence between PG2 bean and non-bean isolates. These results indicate that PG3 strains have a higher level of host specificity than PG2 strains. We then used gradient boosting machine learning to predict each strain's virulence on bean based on whole genome k-mers, type III secreted effector k-mers, and the presence/absence of type III effectors and phytotoxins. Our model performed best using whole genome data and was able to predict virulence with high accuracy (mean absolute error = 0.05). Finally, we functionally validated the model by predicting virulence for 16 strains and found that 15 (94%) had virulence levels within the bounds of estimated predictions. This study strengthens the hypothesis that P. syringae PG2 strains have evolved a different lifestyle than other P. syringae strains as reflected in their lower level of host specificity. It also acts as a proof-of-principle to demonstrate the power of machine learning for predicting host specific adaptation.


Subject(s)
Phaseolus , Pseudomonas syringae , Decision Trees , Host Specificity , Phaseolus/microbiology , Plant Diseases/microbiology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL