Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
1.
Science ; 376(6599): eabh3104, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35549308

ABSTRACT

A hallmark of meiosis is chromosomal pairing, which requires telomere tethering and rotation on the nuclear envelope through microtubules, driving chromosome homology searches. Telomere pulling toward the centrosome forms the "zygotene chromosomal bouquet." Here, we identified the "zygotene cilium" in oocytes. This cilium provides a cable system for the bouquet machinery and extends throughout the germline cyst. Using zebrafish mutants and live manipulations, we demonstrate that the cilium anchors the centrosome to counterbalance telomere pulling. The cilium is essential for bouquet and synaptonemal complex formation, oogenesis, ovarian development, and fertility. Thus, a cilium represents a conserved player in zebrafish and mouse meiosis, which sheds light on reproductive aspects in ciliopathies and suggests that cilia can control chromosomal dynamics.


Subject(s)
Chromosome Pairing , Cilia , Oocytes , Oogenesis , Ovary , Animals , Centromere/genetics , Centromere/physiology , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Cilia/physiology , Female , Fertility/physiology , Mice , Morphogenesis , Oocytes/growth & development , Oogenesis/genetics , Oogenesis/physiology , Ovary/growth & development , Telomere/genetics , Telomere/physiology , Zebrafish/genetics , Zebrafish/physiology
2.
PLoS Genet ; 18(2): e1010040, 2022 02.
Article in English | MEDLINE | ID: mdl-35130272

ABSTRACT

During meiotic prophase I, homologous chromosomes pair, synapse and recombine in a tightly regulated process that ensures the generation of genetically variable haploid gametes. Although the mechanisms underlying meiotic cell division have been well studied in model species, our understanding of the dynamics of meiotic prophase I in non-traditional model mammals remains in its infancy. Here, we reveal key meiotic features in previously uncharacterised marsupial species (the tammar wallaby and the fat-tailed dunnart), plus the fat-tailed mouse opossum, with a focus on sex chromosome pairing strategies, recombination and meiotic telomere homeostasis. We uncovered differences between phylogroups with important functional and evolutionary implications. First, sex chromosomes, which lack a pseudo-autosomal region in marsupials, had species specific pairing and silencing strategies, with implications for sex chromosome evolution. Second, we detected two waves of γH2AX accumulation during prophase I. The first wave was accompanied by low γH2AX levels on autosomes, which correlated with the low recombination rates that distinguish marsupials from eutherian mammals. In the second wave, γH2AX was restricted to sex chromosomes in all three species, which correlated with transcription from the X in tammar wallaby. This suggests non-canonical functions of γH2AX on meiotic sex chromosomes. Finally, we uncover evidence for telomere elongation in primary spermatocytes of the fat-tailed dunnart, a unique strategy within mammals. Our results provide new insights into meiotic progression and telomere homeostasis in marsupials, highlighting the importance of capturing the diversity of meiotic strategies within mammals.


Subject(s)
Chromosome Pairing/physiology , Sex Chromosomes/physiology , Telomere/physiology , Animals , Macropodidae/genetics , Marsupialia/genetics , Meiosis/genetics , Meiosis/physiology , Meiotic Prophase I/physiology , Opossums/genetics , Sex Chromosomes/genetics , Telomere/genetics , X Chromosome/genetics , Y Chromosome/genetics
3.
Plant Physiol ; 188(2): 1210-1228, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34927688

ABSTRACT

Changes in environmental temperature affect multiple meiotic processes in flowering plants. Polyploid plants derived from whole-genome duplication (WGD) have enhanced genetic plasticity and tolerance to environmental stress but face challenges in organizing and segregating doubled chromosome sets. In this study, we investigated the impact of increased environmental temperature on male meiosis in autotetraploid Arabidopsis (Arabidopsis thaliana). Under low to mildly increased temperatures (5°C-28°C), irregular chromosome segregation universally occurred in synthetic autotetraploid Columbia-0 (Col-0). Similar meiotic lesions occurred in autotetraploid rice (Oryza sativa L.) and allotetraploid canola (Brassica napus cv Westar), but not in evolutionarily derived hexaploid wheat (Triticum aestivum). At extremely high temperatures, chromosome separation and tetrad formation became severely disordered due to univalent formation caused by the suppression of crossing-over. We found a strong correlation between tetravalent formation and successful chromosome pairing, both of which were negatively correlated with temperature elevation, suggesting that increased temperature interferes with crossing-over predominantly by impacting homolog pairing. We also showed that loading irregularities of axis proteins ASY1 and ASY4 co-localize on the chromosomes of the syn1 mutant and the heat-stressed diploid and autotetraploid Col-0, revealing that heat stress affects the lateral region of synaptonemal complex (SC) by impacting the stability of the chromosome axis. Moreover, we showed that chromosome axis and SC in autotetraploid Col-0 are more sensitive to increased temperature than those in diploid Arabidopsis. Taken together, our data provide evidence suggesting that WGD negatively affects the stability and thermal tolerance of meiotic recombination in newly synthetic autotetraploid Arabidopsis.


Subject(s)
Arabidopsis/genetics , Chromosome Pairing/physiology , Homologous Recombination/physiology , Hot Temperature/adverse effects , Meiosis/physiology , Oryza/genetics , Polyploidy , Arabidopsis/physiology , Crops, Agricultural/genetics , Crops, Agricultural/physiology , Genetic Variation , Genotype , Heat-Shock Response/genetics , Heat-Shock Response/physiology , Oryza/physiology
4.
Front Endocrinol (Lausanne) ; 12: 761249, 2021.
Article in English | MEDLINE | ID: mdl-34721307

ABSTRACT

To achieve spermatogenesis in vitro, one of the most challenging processes to mimic is meiosis. Meiotic problems, like incomplete synapsis of the homologous chromosomes, or impaired homologous recombination, can cause failure of crossover formation and subsequent chromosome nondisjunction, eventually leading to aneuploid sperm. These meiotic events are therefore strictly monitored by meiotic checkpoints that initiate apoptosis of aberrant spermatocytes and lead to spermatogenic arrest. However, we recently found that, in vitro derived meiotic cells proceeded to the first meiotic division (MI) stage, despite displaying incomplete chromosome synapsis, no discernible XY-body and lack of crossover formation. We therefore optimized our in vitro culture system of meiosis from male germline stem cells (mGSCs) in order to achieve full chromosome synapsis, XY-body formation and meiotic crossovers. In comparison to previous culture system, the in vitro-generated spermatocytes were transferred after meiotic initiation to a second culture dish. This dish already contained a freshly plated monolayer of proliferatively inactivated immortalized Sertoli cells supporting undifferentiated mGSCs. In this way we aimed to simulate the multiple layers of germ cell types that support spermatogenesis in vivo in the testis. We found that in this optimized culture system, although independent of the undifferentiated mGSCs, meiotic chromosome synapsis was complete and XY body appeared normal. However, meiotic recombination still occurred insufficiently and only few meiotic crossovers were formed, leading to MI-spermatocytes displaying univalent chromosomes (paired sister chromatids). Therefore, considering that meiotic checkpoints are not necessarily fully functional in vitro, meiotic crossover formation should be closely monitored when mimicking gametogenesis in vitro to prevent generation of aneuploid gametes.


Subject(s)
Chromosome Pairing/physiology , Chromosomes/physiology , Meiosis/physiology , Aneuploidy , Animals , Azoospermia/congenital , Azoospermia/physiopathology , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Male , Mice , Mice, Inbred DBA , Sertoli Cells/physiology , Spermatocytes/physiology , Spermatogenesis/physiology , Spermatozoa/physiology , Testis/physiology
5.
Nat Commun ; 12(1): 5827, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625553

ABSTRACT

During meiosis, chromosomes exhibit dramatic changes in morphology and intranuclear positioning. How these changes influence homolog pairing, alignment, and recombination remain elusive. Using Hi-C, we systematically mapped 3D genome architecture throughout all meiotic prophase substages during mouse spermatogenesis. Our data uncover two major chromosome organizational features varying along the chromosome axis during early meiotic prophase, when homolog alignment occurs. First, transcriptionally active and inactive genomic regions form alternating domains consisting of shorter and longer chromatin loops, respectively. Second, the force-transmitting LINC complex promotes the alignment of ends of different chromosomes over a range of up to 20% of chromosome length. Both features correlate with the pattern of homolog interactions and the distribution of recombination events. Collectively, our data reveal the influences of transcription and force on meiotic chromosome structure and suggest chromosome organization may provide an infrastructure for the modulation of meiotic recombination in higher eukaryotes.


Subject(s)
Meiosis/physiology , Animals , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Flow Cytometry , Homologous Recombination/genetics , Homologous Recombination/physiology , Humans , In Situ Hybridization, Fluorescence , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , RNA-Seq , Spermatocytes/metabolism
6.
Genes (Basel) ; 12(9)2021 08 29.
Article in English | MEDLINE | ID: mdl-34573341

ABSTRACT

We analyzed the synapsis and recombination between Z and W chromosomes in the oocytes of nine neognath species: domestic chicken Gallus gallus domesticus, grey goose Anser anser, black tern Chlidonias niger, common tern Sterna hirundo, pale martin Riparia diluta, barn swallow Hirundo rustica, European pied flycatcher Ficedula hypoleuca, great tit Parus major and white wagtail Motacilla alba using immunolocalization of SYCP3, the main protein of the lateral elements of the synaptonemal complex, and MLH1, the mismatch repair protein marking mature recombination nodules. In all species examined, homologous synapsis occurs in a short region of variable size at the ends of Z and W chromosomes, where a single recombination nodule is located. The remaining parts of the sex chromosomes undergo synaptic adjustment and synapse non-homologously. In 25% of ZW bivalents of white wagtail, synapsis and recombination also occur at the secondary pairing region, which probably resulted from autosome-sex chromosome translocation. Using FISH with a paint probe specific to the germline-restricted chromosome (GRC) of the pale martin on the oocytes of the pale martin, barn swallow and great tit, we showed that both maternally inherited songbird chromosomes (GRC and W) share common sequences.


Subject(s)
Birds/genetics , Chromosome Pairing/physiology , Recombination, Genetic , Sex Chromosomes , Animals , Chickens/genetics , Female , In Situ Hybridization, Fluorescence , MutL Protein Homolog 1/genetics , Oocytes/physiology , Pachytene Stage/genetics , Passeriformes/genetics
7.
Nat Struct Mol Biol ; 28(8): 681-693, 2021 08.
Article in English | MEDLINE | ID: mdl-34373646

ABSTRACT

The synaptonemal complex (SC) is a supramolecular protein assembly that mediates synapsis between homologous chromosomes during meiosis. SC elongation along the chromosome length (up to 24 µm) depends on its midline α-fibrous component SYCE2-TEX12. Here, we report X-ray crystal structures of human SYCE2-TEX12 as an individual building block and on assembly within a fibrous lattice. We combine these structures with mutagenesis, biophysics and electron microscopy to reveal the hierarchical mechanism of SYCE2-TEX12 fiber assembly. SYCE2-TEX12's building blocks are 2:2 coiled coils that dimerize into 4:4 hetero-oligomers and interact end-to-end and laterally to form 10-nm fibers that intertwine within 40-nm bundled micrometer-long fibers that define the SC's midline structure. This assembly mechanism bears striking resemblance with intermediate filament proteins vimentin, lamin and keratin. Thus, SYCE2-TEX12 exhibits behavior typical of cytoskeletal proteins to provide an α-fibrous SC backbone that structurally underpins synaptic elongation along meiotic chromosomes.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosome Pairing/physiology , Synaptonemal Complex/metabolism , Crystallography, X-Ray , Cytoskeletal Proteins/metabolism , Humans , Keratins/metabolism , Lamins/metabolism , Meiosis/physiology , Protein Structure, Quaternary , Vimentin/metabolism
8.
Elife ; 102021 06 08.
Article in English | MEDLINE | ID: mdl-34100718

ABSTRACT

Three-dimensional eukaryotic genome organization provides the structural basis for gene regulation. In Drosophila melanogaster, genome folding is characterized by somatic homolog pairing, where homologous chromosomes are intimately paired from end to end; however, how homologs identify one another and pair has remained mysterious. Recently, this process has been proposed to be driven by specifically interacting 'buttons' encoded along chromosomes. Here, we turned this hypothesis into a quantitative biophysical model to demonstrate that a button-based mechanism can lead to chromosome-wide pairing. We tested our model using live-imaging measurements of chromosomal loci tagged with the MS2 and PP7 nascent RNA labeling systems. We show solid agreement between model predictions and experiments in the pairing dynamics of individual homologous loci. Our results strongly support a button-based mechanism of somatic homolog pairing in Drosophila and provide a theoretical framework for revealing the molecular identity and regulation of buttons.


Subject(s)
Chromosome Pairing , Chromosomes , Models, Genetic , Animals , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Chromosomes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , Drosophila melanogaster , Embryo, Nonmammalian , Female , Genome, Insect/genetics , Male , Microscopy, Confocal
9.
Sci Rep ; 11(1): 9055, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907215

ABSTRACT

The cholinergic midbrain is involved in a wide range of motor and cognitive processes. Cholinergic neurons of the pedunculopontine (PPN) and laterodorsal tegmental nucleus (LDT) send long-ranging axonal projections that target sensorimotor and limbic areas in the thalamus, the dopaminergic midbrain and the striatal complex following a topographical gradient, where they influence a range of functions including attention, reinforcement learning and action-selection. Nevertheless, a comprehensive examination of the afferents to PPN and LDT cholinergic neurons is still lacking, partly due to the neurochemical heterogeneity of this region. Here we characterize the whole-brain input connectome to cholinergic neurons across distinct functional domains (i.e. PPN vs LDT) using conditional transsynaptic retrograde labeling in ChAT::Cre male and female rats. We reveal that input neurons are widely distributed throughout the brain but segregated into specific functional domains. Motor related areas innervate preferentially the PPN, whereas limbic related areas preferentially innervate the LDT. The quantification of input neurons revealed that both PPN and LDT receive similar substantial inputs from the superior colliculus and the output of the basal ganglia (i.e. substantia nigra pars reticulata). Notably, we found that PPN cholinergic neurons receive preferential inputs from basal ganglia structures, whereas LDT cholinergic neurons receive preferential inputs from limbic cortical areas. Our results provide the first characterization of inputs to PPN and LDT cholinergic neurons and highlight critical differences in the connectome among brain cholinergic systems thus supporting their differential roles in behavior.


Subject(s)
Brain Mapping/methods , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/physiology , Chromosome Pairing/physiology , Neural Pathways/physiology , Pedunculopontine Tegmental Nucleus/physiology , Tegmentum Mesencephali/physiology , Animals , Female , Male , Pedunculopontine Tegmental Nucleus/anatomy & histology , Rats , Tegmentum Mesencephali/anatomy & histology
10.
Plant Physiol ; 185(4): 1783-1797, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33793950

ABSTRACT

Meiotic recombination (MR) drives novel combinations of alleles and contributes to genomic diversity in eukaryotes. In this study, we showed that heat stress (36°C-38°C) over the fertile threshold fully abolished crossover formation in Arabidopsis (Arabidopsis thaliana). Cytological and genetic studies in wild-type plants and syn1 and rad51 mutants suggested that heat stress reduces generation of SPO11-dependent double-strand breaks (DSBs). In support, the abundance of recombinase DMC1, which is required for MR-specific DSB repair, was significantly reduced under heat stress. In addition, high temperatures induced disassembly and/or instability of the ASY4- but not the SYN1-mediated chromosome axis. At the same time, the ASY1-associated lateral element of the synaptonemal complex (SC) was partially affected, while the ZYP1-dependent central element of SC was disrupted, indicating that heat stress impairs SC formation. Moreover, expression of genes involved in DSB formation; e.g. SPO11-1, PRD1, 2, and 3 was not impacted; however, recombinase RAD51 and chromosome axis factors ASY3 and ASY4 were significantly downregulated under heat stress. Taken together, these findings revealed that heat stress inhibits MR via compromised DSB formation and homolog synapsis, which are possible downstream effects of the impacted chromosome axis. Our study thus provides evidence shedding light on how increasing environmental temperature influences MR in Arabidopsis.


Subject(s)
Arabidopsis/genetics , Arabidopsis/physiology , Chromosome Pairing/physiology , DNA Breaks, Double-Stranded , Heat-Shock Response/genetics , Heat-Shock Response/physiology , Meiosis/physiology , Chromosome Pairing/genetics , Genetic Variation , Genotype , Meiosis/genetics
11.
Exp Cell Res ; 399(2): 112455, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33400935

ABSTRACT

During meiosis, homologous chromosomes exchange genetic material. This exchange or meiotic recombination is mediated by a proteinaceous scaffold known as the Synaptonemal complex (SC). Any defects in its formation produce failures in meiotic recombination, chromosome segregation and meiosis completion. It has been proposed that DNA repair events that will be resolved by crossover between homologous chromosomes are predetermined by the SC. Hence, structural analysis of the organization of the DNA in the SC could shed light on the process of crossover interference. In this work, we employed an ultrastructural DNA staining technique on mouse testis and followed nuclei of pachytene cells. We observed structures organized similarly to the SCs stained with conventional techniques. These structures, presumably the DNA in the SCs, are delineating the edges of both lateral elements and no staining was observed between them. DNA in the LEs resembles two parallel tracks. However, a bubble-like staining pattern in certain regions of the SC was observed. Furthermore, this staining pattern is found in SCs formed between non-homologous chromosomes, in SCs formed between sister chromatids and in SCs without lateral elements, suggesting that this particular organization of the DNA is determined by the synapsis of the chromosomes despite their lack of homology or the presence of partially formed SCs.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Meiosis/physiology , Synaptonemal Complex/metabolism , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatids/chemistry , Chromatids/metabolism , Chromatids/ultrastructure , Chromosome Pairing/physiology , DNA/chemistry , DNA/ultrastructure , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Male , Mice , Mice, Knockout , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Structure, Quaternary , Synaptonemal Complex/physiology , Synaptonemal Complex/ultrastructure
12.
Int J Mol Sci ; 23(1)2021 Dec 25.
Article in English | MEDLINE | ID: mdl-35008637

ABSTRACT

The assembly of synaptic protein-DNA complexes by specialized proteins is critical for bringing together two distant sites within a DNA molecule or bridging two DNA molecules. The assembly of such synaptosomes is needed in numerous genetic processes requiring the interactions of two or more sites. The molecular mechanisms by which the protein brings the sites together, enabling the assembly of synaptosomes, remain unknown. Such proteins can utilize sliding, jumping, and segmental transfer pathways proposed for the single-site search process, but none of these pathways explains how the synaptosome assembles. Here we used restriction enzyme SfiI, that requires the assembly of synaptosome for DNA cleavage, as our experimental system and applied time-lapse, high-speed AFM to directly visualize the site search process accomplished by the SfiI enzyme. For the single-site SfiI-DNA complexes, we were able to directly visualize such pathways as sliding, jumping, and segmental site transfer. However, within the synaptic looped complexes, we visualized the threading and site-bound segment transfer as the synaptosome-specific search pathways for SfiI. In addition, we visualized sliding and jumping pathways for the loop dissociated complexes. Based on our data, we propose the site-search model for synaptic protein-DNA systems.


Subject(s)
DNA/metabolism , Proteins/metabolism , Binding Sites/physiology , Chromosome Pairing/physiology , DNA Restriction Enzymes/metabolism , Plasmids/metabolism , Protein Binding/physiology , Synaptosomes/metabolism
13.
Plant J ; 105(6): 1665-1676, 2021 03.
Article in English | MEDLINE | ID: mdl-33346910

ABSTRACT

Allopolyploid wheat (Triticum aestivum L.) carries three pairs of homoeologous genomes but its meiotic pairing is diploid-like. This is the effect of the Ph (pairing homoeologous) system which restricts chromosome pairing to strictly homologous. Ph1 is the locus with the strongest effect. Disabling Ph1 permits pairing between homoeologues and is routinely used in chromosome engineering to introgress alien variation into breeding stocks. Whereas the efficiency of Ph1 and the general pattern of homoeologous crossovers in its absence are quite well known from numerous studies, other characteristics of such crossovers remain unknown. This study analyzed the crossover points in four sets of the ph1b-induced recombinants between wheat homologues as well as between three wheat and rye (Secale cereale) homoeologous chromosome arms, and compared them to crossovers between homologues in a reference wheat population. The results show the Ph1 locus also controls crossing over of homologues, and the general patterns of homologous (with Ph1) and homoeologous (with ph1b) crossing over are the same. In all intervals analyzed, homoeologous crossovers fell within the range of frequency distribution of homologous crossovers among individual families of the reference population. No specific DNA sequence characteristics were identified that could be recognized by the Ph1 locus; the only difference between homologous and homoeologous crossing over appears to be in frequency. It is concluded that the Ph1 locus likely recognizes DNA sequence similarity; crossing over is permitted between very similar sequences. In the absence of Ph1 dissimilarities are ignored, in proportion to the level of the sequence divergence.


Subject(s)
Chromosomes, Plant/genetics , Secale/genetics , Triticum/genetics , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Crossing Over, Genetic/genetics , Plant Breeding
14.
Nature ; 589(7840): 96-102, 2021 01.
Article in English | MEDLINE | ID: mdl-33208951

ABSTRACT

The hippocampus has a major role in encoding and consolidating long-term memories, and undergoes plastic changes during sleep1. These changes require precise homeostatic control by subcortical neuromodulatory structures2. The underlying mechanisms of this phenomenon, however, remain unknown. Here, using multi-structure recordings in macaque monkeys, we show that the brainstem transiently modulates hippocampal network events through phasic pontine waves known as pontogeniculooccipital waves (PGO waves). Two physiologically distinct types of PGO wave appear to occur sequentially, selectively influencing high-frequency ripples and low-frequency theta events, respectively. The two types of PGO wave are associated with opposite hippocampal spike-field coupling, prompting periods of high neural synchrony of neural populations during periods of ripple and theta instances. The coupling between PGO waves and ripples, classically associated with distinct sleep stages, supports the notion that a global coordination mechanism of hippocampal sleep dynamics by cholinergic pontine transients may promote systems and synaptic memory consolidation as well as synaptic homeostasis.


Subject(s)
Geniculate Bodies/physiology , Hippocampus/physiology , Occipital Lobe/physiology , Pons/physiology , Sleep/physiology , Theta Rhythm/physiology , Animals , Chromosome Pairing/physiology , Female , Homeostasis , Macaca/physiology , Memory Consolidation/physiology , Neuronal Plasticity , Sleep Stages/physiology
15.
Behav Brain Res ; 391: 112705, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32473844

ABSTRACT

Pairing vagus nerve stimulation (VNS) with rehabilitation has emerged as a potential strategy to improve recovery after neurological injury, an effect ascribed to VNS-dependent enhancement of synaptic plasticity. Previous studies demonstrate that pairing VNS with forelimb training increases forelimb movement representations in motor cortex. However, it is not known whether VNS-dependent enhancement of plasticity is restricted to forelimb training or whether VNS paired with other movements could induce plasticity of other motor representations. We tested the hypothesis that VNS paired with orofacial movements associated with chewing during an unskilled task would drive a specific increase in jaw representation in motor cortex compared to equivalent behavioral experience without VNS. Rats performed a behavioral task in which VNS at a specified intensity between 0 and 1.2 mA was paired with chewing 200 times per day for five days. Intracortical microstimulation (ICMS) was then used to document movement representations in motor cortex. VNS paired with chewing at 0.8 mA significantly increased motor cortex jaw representation compared to equivalent behavioral training without stimulation (Bonferroni-corrected unpaired t-test, p < 0.01). Higher and lower intensities failed to alter cortical plasticity. No changes in other movement representations or total motor cortex area were observed between groups. These results demonstrate that 0.8 mA VNS paired with training drives robust plasticity specific to the paired movement, is not restricted to forelimb representations, and occurs with training on an unskilled task. This suggests that moderate intensity VNS may be a useful adjuvant to enhance plasticity and support benefits of rehabilitative therapies targeting functions beyond upper limb movement.


Subject(s)
Conditioning, Psychological/physiology , Motor Cortex/physiology , Neuronal Plasticity/physiology , Animals , Chromosome Pairing/physiology , Female , Mastication/physiology , Motor Cortex/metabolism , Movement/physiology , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Vagus Nerve/metabolism , Vagus Nerve/physiology , Vagus Nerve Stimulation/methods
16.
Mol Hum Reprod ; 26(7): 485-497, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32402064

ABSTRACT

More than 50% of cases of primary ovarian insufficiency (POI) and nonobstructive azoospermia in humans are classified as idiopathic infertility. Meiotic defects may relate to at least some of these cases. Mutations in genes coding for synaptonemal complex (SC) components have been identified in humans, and hypothesized to be causative for the observed infertile phenotype. Mutation SYCE1 c.721C>T (former c.613C>T)-a familial mutation reported in two sisters with primary amenorrhea-was the first such mutation found in an SC central element component-coding gene. Most fundamental mammalian oogenesis events occur during the embryonic phase, and eventual defects are identified many years later, thus leaving few possibilities to study the condition's etiology and pathogenesis. Aiming to validate an approach to circumvent this difficulty, we have used the CRISPR/Cas9 technology to generate a mouse model with an SYCE1 c.721C>T equivalent genome alteration. We hereby present the characterization of the homozygous mutant mice phenotype, compared to their wild type and heterozygous littermates. Our results strongly support a causative role of this mutation for the POI phenotype in human patients, and the mechanisms involved would relate to defects in homologous chromosome synapsis. No SYCE1 protein was detected in homozygous mutants and Syce1 transcript level was highly diminished, suggesting transcript degradation as the basis of the infertility mechanism. This is the first report on the generation of a humanized mouse model line for the study of an infertility-related human mutation in an SC component-coding gene, thus representing a proof of principle.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Point Mutation/genetics , Primary Ovarian Insufficiency/genetics , Animals , Chromosome Pairing/genetics , Chromosome Pairing/physiology , DNA-Binding Proteins/genetics , Female , Flow Cytometry , Genetic Predisposition to Disease/genetics , Homozygote , Humans , Immunohistochemistry , Meiosis/genetics , Meiosis/physiology , Mice , Mutation/genetics
17.
PLoS One ; 15(4): e0230787, 2020.
Article in English | MEDLINE | ID: mdl-32275724

ABSTRACT

The mechanisms of epileptic discharge generation and spread are not yet fully known. A recently proposed simple biophysical model of interictal and ictal discharges, Epileptor-2, reproduces well the main features of neuronal excitation and ionic dynamics during discharge generation. In order to distinguish between two hypothesized mechanisms of discharge propagation, we extend the model to the case of two-dimensional propagation along the cortical neural tissue. The first mechanism is based on extracellular potassium diffusion, and the second is the propagation of spikes and postsynaptic signals along axons and dendrites. Our simulations show that potassium diffusion is too slow to reproduce an experimentally observed speed of ictal wavefront propagation (tenths of mm/s). By contrast, the synaptic mechanism predicts well the speed and synchronization of the pre-ictal bursts before the ictal front and the afterdischarges in the ictal core. Though this fact diminishes the role of diffusion and electrodiffusion, the model nevertheless highlights the role of potassium extrusion during neuronal excitation, which provides a positive feedback that changes at the ictal wavefront the balance of excitation versus inhibition in favor of excitation. This finding may help to find a target for a treatment to prevent seizure propagation.


Subject(s)
Dendrites/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Potassium/metabolism , Seizures/metabolism , Seizures/physiopathology , Action Potentials/physiology , Axons/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Chromosome Pairing/physiology , Models, Neurological , Neurons/metabolism , Synaptic Potentials/physiology
18.
Proc Natl Acad Sci U S A ; 117(18): 10055-10066, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32312822

ABSTRACT

Synaptic activity in neurons leads to the rapid activation of genes involved in mammalian behavior. ATP-dependent chromatin remodelers such as the BAF complex contribute to these responses and are generally thought to activate transcription. However, the mechanisms keeping such "early activation" genes silent have been a mystery. In the course of investigating Mendelian recessive autism, we identified six families with segregating loss-of-function mutations in the neuronal BAF (nBAF) subunit ACTL6B (originally named BAF53b). Accordingly, ACTL6B was the most significantly mutated gene in the Simons Recessive Autism Cohort. At least 14 subunits of the nBAF complex are mutated in autism, collectively making it a major contributor to autism spectrum disorder (ASD). Patient mutations destabilized ACTL6B protein in neurons and rerouted dendrites to the wrong glomerulus in the fly olfactory system. Humans and mice lacking ACTL6B showed corpus callosum hypoplasia, indicating a conserved role for ACTL6B in facilitating neural connectivity. Actl6b knockout mice on two genetic backgrounds exhibited ASD-related behaviors, including social and memory impairments, repetitive behaviors, and hyperactivity. Surprisingly, mutation of Actl6b relieved repression of early response genes including AP1 transcription factors (Fos, Fosl2, Fosb, and Junb), increased chromatin accessibility at AP1 binding sites, and transcriptional changes in late response genes associated with early response transcription factor activity. ACTL6B loss is thus an important cause of recessive ASD, with impaired neuron-specific chromatin repression indicated as a potential mechanism.


Subject(s)
Autism Spectrum Disorder/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Hippocampus/pathology , Actins/genetics , Adenosine Triphosphate/genetics , Animals , Autism Spectrum Disorder/pathology , Behavior, Animal/physiology , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Dendrites/genetics , Dendrites/physiology , Disease Models, Animal , Gene Expression Regulation/genetics , Hippocampus/metabolism , Humans , Mice , Mice, Knockout , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Transcription Factors/genetics
19.
Elife ; 92020 03 10.
Article in English | MEDLINE | ID: mdl-32149606

ABSTRACT

Chromosome segregation during male meiosis is tailored to rapidly generate multitudes of sperm. Little is known about mechanisms that efficiently partition chromosomes to produce sperm. Using live imaging and tomographic reconstructions of spermatocyte meiotic spindles in Caenorhabditis elegans, we find the lagging X chromosome, a distinctive feature of anaphase I in C. elegans males, is due to lack of chromosome pairing. The unpaired chromosome remains tethered to centrosomes by lengthening kinetochore microtubules, which are under tension, suggesting that a 'tug of war' reliably resolves lagging. We find spermatocytes exhibit simultaneous pole-to-chromosome shortening (anaphase A) and pole-to-pole elongation (anaphase B). Electron tomography unexpectedly revealed spermatocyte anaphase A does not stem solely from kinetochore microtubule shortening. Instead, movement of autosomes is largely driven by distance change between chromosomes, microtubules, and centrosomes upon tension release during anaphase. Overall, we define novel features that segregate both lagging and paired chromosomes for optimal sperm production.


Subject(s)
Chromosome Pairing/physiology , Chromosome Segregation/physiology , Meiosis/physiology , Spermatocytes/physiology , Spindle Apparatus/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins , Male , X Chromosome
20.
Life Sci Alliance ; 3(3)2020 03.
Article in English | MEDLINE | ID: mdl-32051254

ABSTRACT

In mitotic cells, establishment of sister chromatid cohesion requires acetylation of the cohesin subunit SMC3 (acSMC3) by ESCO1 and/or ESCO2. Meiotic cohesin plays additional but poorly understood roles in the formation of chromosome axial elements (AEs) and synaptonemal complexes. Here, we show that levels of ESCO2, acSMC3, and the pro-cohesion factor sororin increase on meiotic chromosomes as homologs synapse. These proteins are less abundant on the largely unsynapsed sex chromosomes, whose sister chromatid cohesion appears weaker throughout the meiotic prophase. Using three distinct conditional Esco2 knockout mouse strains, we demonstrate that ESCO2 is essential for male gametogenesis. Partial depletion of ESCO2 in prophase I spermatocytes delays chromosome synapsis and further weakens cohesion along sex chromosomes, which show extensive separation of AEs into single chromatids. Unsynapsed regions of autosomes are associated with the sex chromatin and also display split AEs. This study provides the first evidence for a specific role of ESCO2 in mammalian meiosis, identifies a particular ESCO2 dependence of sex chromosome cohesion and suggests support of autosomal synapsis by acSMC3-stabilized cohesion.


Subject(s)
Acetyltransferases/metabolism , Chromatids/metabolism , Chromosome Pairing/physiology , Acetylation , Acetyltransferases/genetics , Acetyltransferases/physiology , Animals , Cell Cycle Proteins , Chromatids/genetics , Chromosomal Proteins, Non-Histone , Chromosome Pairing/genetics , Chromosome Segregation/genetics , Chromosome Segregation/physiology , Chromosome Structures/metabolism , Gametogenesis/genetics , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Sex Chromosomes/metabolism , Spermatocytes/metabolism , Synaptonemal Complex/metabolism , Cohesins
SELECTION OF CITATIONS
SEARCH DETAIL
...