← Back

Cortical Development

Topic spotlight
TopicWorld Wide

cortical development

Discover seminars, jobs, and research tagged with cortical development across World Wide.
30 curated items25 Seminars5 ePosters
Updated over 1 year ago
30 items · cortical development
30 results
SeminarNeuroscience

Investigating dynamiCa++l mechanisms underlying cortical development and disease

Georgia Panagiotakos
Icahn School of Medicine at Mount Sinai
May 7, 2024
SeminarNeuroscience

Modeling human brain development and disease: the role of primary cilia

Kyrousi Christina
Medical School, National and Kapodistrian University of Athens, Athens, Greece
Apr 23, 2024

Neurodevelopmental disorders (NDDs) impose a global burden, affecting an increasing number of individuals. While some causative genes have been identified, understanding the human-specific mechanisms involved in these disorders remains limited. Traditional gene-driven approaches for modeling brain diseases have failed to capture the diverse and convergent mechanisms at play. Centrosomes and cilia act as intermediaries between environmental and intrinsic signals, regulating cellular behavior. Mutations or dosage variations disrupting their function have been linked to brain formation deficits, highlighting their importance, yet their precise contributions remain largely unknown. Hence, we aim to investigate whether the centrosome/cilia axis is crucial for brain development and serves as a hub for human-specific mechanisms disrupted in NDDs. Towards this direction, we first demonstrated species-specific and cell-type-specific differences in the cilia-genes expression during mouse and human corticogenesis. Then, to dissect their role, we provoked their ectopic overexpression or silencing in the developing mouse cortex or in human brain organoids. Our findings suggest that cilia genes manipulation alters both the numbers and the position of NPCs and neurons in the developing cortex. Interestingly, primary cilium morphology is disrupted, as we find changes in their length, orientation and number that lead to disruption of the apical belt and altered delamination profiles during development. Our results give insight into the role of primary cilia in human cortical development and address fundamental questions regarding the diversity and convergence of gene function in development and disease manifestation. It has the potential to uncover novel pharmacological targets, facilitate personalized medicine, and improve the lives of individuals affected by NDDs through targeted cilia-based therapies.

SeminarNeuroscience

Investigating activity-dependent processes during cortical neuronal assembly in development and disease

Simona Lodato
Humanitas University
Mar 19, 2024
SeminarNeuroscience

Astrocyte reprogramming / activation and brain homeostasis

Thomaidou Dimitra
Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
Dec 12, 2023

Astrocytes are multifunctional glial cells, implicated in neurogenesis and synaptogenesis, supporting and fine-tuning neuronal activity and maintaining brain homeostasis by controlling blood-brain barrier permeability. During the last years a number of studies have shown that astrocytes can also be converted into neurons if they force-express neurogenic transcription factors or miRNAs. Direct astrocytic reprogramming to induced-neurons (iNs) is a powerful approach for manipulating cell fate, as it takes advantage of the intrinsic neural stem cell (NSC) potential of brain resident reactive astrocytes. To this end, astrocytic cell fate conversion to iNs has been well-established in vitro and in vivo using combinations of transcription factors (TFs) or chemical cocktails. Challenging the expression of lineage-specific TFs is accompanied by changes in the expression of miRNAs, that post-transcriptionally modulate high numbers of neurogenesis-promoting factors and have therefore been introduced, supplementary or alternatively to TFs, to instruct direct neuronal reprogramming. The neurogenic miRNA miR-124 has been employed in direct reprogramming protocols supplementary to neurogenic TFs and other miRNAs to enhance direct neurogenic conversion by suppressing multiple non-neuronal targets. In our group we aimed to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced-neurons (iNs) on its own both in vitro and in vivo and elucidate its independent mechanism of reprogramming action. Our in vitro data indicate that miR-124 is a potent driver of the reprogramming switch of astrocytes towards an immature neuronal fate. Elucidation of the molecular pathways being triggered by miR-124 by RNA-seq analysis revealed that miR-124 is sufficient to instruct reprogramming of cortical astrocytes to immature induced-neurons (iNs) in vitro by down-regulating genes with important regulatory roles in astrocytic function. Among these, the RNA binding protein Zfp36l1, implicated in ARE-mediated mRNA decay, was found to be a direct target of miR-124, that be its turn targets neuronal-specific proteins participating in cortical development, which get de-repressed in miR-124-iNs. Furthermore, miR-124 is potent to guide direct neuronal reprogramming of reactive astrocytes to iNs of cortical identity following cortical trauma, a novel finding confirming its robust reprogramming action within the cortical microenvironment under neuroinflammatory conditions. In parallel to their reprogramming properties, astrocytes also participate in the maintenance of blood-brain barrier integrity, which ensures the physiological functioning of the central nervous system and gets affected contributing to the pathology of several neurodegenerative diseases. To study in real time the dynamic physical interactions of astrocytes with brain vasculature under homeostatic and pathological conditions, we performed 2-photon brain intravital imaging in a mouse model of systemic neuroinflammation, known to trigger astrogliosis and microgliosis and to evoke changes in astrocytic contact with brain vasculature. Our in vivo findings indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, however this event is at compensated by the cross-talk of astrocytes with activated microglia, safeguarding blood vessel coverage and maintenance of blood-brain integrity.

SeminarNeuroscience

Mechanisms of human cortical development and neuropsychiatric disease

Luis de la Torre-Ubieta
University of California Los Angeles
May 30, 2023
SeminarNeuroscience

Myelin Formation and Oligodendrocyte Biology in Epilepsy

Angelika Mühlebner
Universitair Medisch Centrum Utrecht
Feb 15, 2023

Epilepsy is one of the most common neurological diseases according to the World Health Organization (WHO) affecting around 70 million people worldwide [WHO]. Patients who suffer from epilepsy also suffer from a variety of neuro-psychiatric co-morbidities, which they can experience as crippling as the seizure condition itself. Adequate organization of cerebral white matter is utterly important for cognitive development. The failure of integration of neurologic function with cognition is reflected in neuro-psychiatric disease, such as autism spectrum disorder (ASD). However, in epilepsy we know little about the importance of white matter abnormalities in epilepsy-associated co-morbidities. Epilepsy surgery is an important therapy strategy in patients where conventional anti-epileptic drug treatment fails . On histology of the resected brain samples, malformations of cortical development (MCD) are common among the epilepsy surgery population, especially focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC). Both pathologies are associated with constitutive activation of the mTOR pathway. Interestingly, some type of FCD is morphological similar to TSC cortical tubers including the abnormalities of the white matter. Hypomyelination with lack of myelin-producing cells, the oligodendrocytes, within the lesional area is a striking phenomenon. Impairment of the complex myelination process can have a major impact on brain function. In the worst case leading to distorted or interrupted neurotransmissions. It is still unclear whether the observed myelin pathology in epilepsy surgical specimens is primarily related to the underlying malformation process or is just a secondary phenomenon of recurrent epileptic seizures creating a toxic micro-environment which hampers myelin formation. Interestingly, mTORC1 has been implicated as key signal for myelination, thus, promoting the maturation of oligodendrocytes . These results, however, remain controversial. Regardless of the underlying pathophysiologic mechanism, alterations of myelin dynamics, depending on their severity, are known to be linked to various kinds of developmental disorders or neuropsychiatric manifestations.

SeminarNeuroscienceRecording

Brain mosaicism in epileptogenic cortical malformations

Stéphanie Baulac
ICM Paris
Jan 31, 2023

Focal Cortical Dysplasia (FCD) is the most common focal cortical malformation leading to intractable childhood focal epilepsy. In recent years, we and others have shown that FCD type II is caused by mosaic mutations in genes within the PI3K-AKT-mTOR-signaling pathway. Hyperactivation of the mTOR pathway accounts for neuropathological abnormalities and seizure occurrence in FCD. We further showed from human surgical FCDII tissue that epileptiform activity correlates with the density of mutated dysmorphic neurons, supporting their pro-epileptogenic role. The level of mosaicism, as defined by variant allele frequency (VAF) is thought to correlate with the size and regional brain distribution of the lesion such that when a somatic mutation occurs early during the cortical development, the dysplastic area is smaller than if it occurs later. Novel approaches based on the detection of cell-free DNA from the CSF and from trace tissue adherent to SEEG electrodes promise future opportunities for genetic testing during the presurgical evaluation of refractory epilepsy patients or in those that are not eligible for surgery. In utero-based electroporation mouse models allow to express somatic mutation during neurodevelopment and recapitulate most neuropathological and clinical features of FCDII, establishing relevant preclinical mouse models for developing precision medicine strategies.

SeminarNeuroscience

Myelin Formation and Oligodendrocyte Biology in Epilepsy

Angelika Mühlebner
Universitair Medisch Centrum Utrecht
Oct 18, 2022

Epilepsy is one of the most common neurological diseases according to the World Health Organization (WHO) affecting around 70 million people worldwide [WHO]. Patients who suffer from epilepsy also suffer from a variety of neuro-psychiatric co-morbidities, which they can experience as crippling as the seizure condition itself. Adequate organization of cerebral white matter is utterly important for cognitive development. The failure of integration of neurologic function with cognition is reflected in neuro-psychiatric disease, such as autism spectrum disorder (ASD). However, in epilepsy we know little about the importance of white matter abnormalities in epilepsy-associated co-morbidities. Epilepsy surgery is an important therapy strategy in patients where conventional anti-epileptic drug treatment fails . On histology of the resected brain samples, malformations of cortical development (MCD) are common among the epilepsy surgery population, especially focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC). Both pathologies are associated with constitutive activation of the mTOR pathway. Interestingly, some type of FCD is morphological similar to TSC cortical tubers including the abnormalities of the white matter. Hypomyelination with lack of myelin-producing cells, the oligodendrocytes, within the lesional area is a striking phenomenon. Impairment of the complex myelination process can have a major impact on brain function. In the worst case leading to distorted or interrupted neurotransmissions. It is still unclear whether the observed myelin pathology in epilepsy surgical specimens is primarily related to the underlying malformation process or is just a secondary phenomenon of recurrent epileptic seizures creating a toxic micro-environment which hampers myelin formation. Interestingly, mTORC1 has been implicated as key signal for myelination, thus, promoting the maturation of oligodendrocytes . These results, however, remain controversial. Regardless of the underlying pathophysiologic mechanism, alterations of myelin dynamics, depending on their severity, are known to be linked to various kinds of developmental disorders or neuropsychiatric manifestations.

SeminarNeuroscience

Epigenome regulation in neocortex expansion and generation of neuronal subtypes

Tran Tuoc, PhD
Ruhruniversität-Bochum, Humangenetik
Aug 23, 2022

Evolutionarily, the expansion of the human neocortex accounts for many of the unique cognitive abilities of humans. This expansion appears to reflect the increased proliferative potential of basal progenitors (BPs) in mammalian evolution. Further cortical progenitors generate both glutamatergic excitatory neurons (ENs) and GABAergic inhibitory interneurons (INs) in human cortex, whereas they produce exclusively ENs in rodents. The increased proliferative capacity and neuronal subtype generation of cortical progenitors in mammalian evolution may have evolved through epigenetic alterations. However, whether or how the epigenome in cortical progenitors differs between humans and other species is unknown. Here, we report that histone H3 acetylation is a key epigenetic regulation in BP profiling of sorted BPs, we show that H3K9 acetylation is low in murine BPs and high in amplification, neuronal subtype generation and cortical expansion. Through epigenetic profiling of sorted BPs, we show that H3K9 acetylation is low in murine BPs and high in human BPs. Elevated H3K9ac preferentially increases BP proliferation, increasing the size and folding of the normally smooth mouse neocortex. Furthermore, we found that the elevated H3 acetylation activates expression of IN genes in in developing mouse cortex and promote proliferation of IN progenitor-like cells in cortex of Pax6 mutant mouse models. Mechanistically, H3K9ac drives the BP amplification and proliferation of these IN progenitor-like cells by increasing expression of the evolutionarily regulated gene, TRNP1. Our findings demonstrate a previously unknown mechanism that controls neocortex expansion and generation of neuronal subtypes. Keywords: Cortical development, neurogenesis, basal progenitors, cortical size, gyrification, excitatory neuron, inhibitory interneuron, epigenetic profiling, epigenetic regulation, H3 acetylation, H3K9ac, TRNP1, PAX6

SeminarNeuroscience

Investigating activity-dependent processes in cerebral cortex development and disease

Simona Lodato
Humanitas University
Jul 19, 2022

The cerebral cortex contains an extraordinary diversity of excitatory projection neuron (PN) and inhibitory interneurons (IN), wired together to form complex circuits. Spatiotemporally coordinated execution of intrinsic molecular programs by PNs and INs and activity-dependent processes, contribute to cortical development and cortical microcircuits formation. Alterations of these delicate processes have often been associated to neurological/neurodevelopmental disorders. However, despite the groundbreaking discovery that spontaneous activity in the embryonic brain can shape regional identities of distinct cortical territories, it is still unclear whether this early activity contributes to define subtype-specific neuronal fate as well as circuit assembly. In this study, we combined in utero genetic perturbations via CRISPR/Cas9 system and pharmacological inhibition of selected ion channels with RNA-sequencing and live imaging technologies to identify the activity-regulated processes controlling the development of different cortical PN classes, their wiring and the acquisition of subtype specific features. Moreover, we generated human induced pluripotent stem cells (iPSCs) form patients affected by a severe, rare and untreatable form of developmental epileptic encephalopathy. By differentiating cortical organoids form patient-derived iPSCs we create human models of early electrical alterations for studying molecular, structural and functional consequences of the genetic mutations during cortical development. Our ultimate goal is to define the activity-conditioned processes that physiologically occur during the development of cortical circuits, to identify novel therapeutical paths to address the pathological consequences of neonatal epilepsies.

SeminarNeuroscience

‘Studying cortical development through the lens of human disorders’

Gaia Novarino
Institute of Science and Technology Austria
Mar 16, 2022
SeminarNeuroscienceRecording

Dissecting the 3D regulatory landscape of the developing cerebral cortex with single-cell epigenomics

Boyan Bonev, PhD
Ludwig-Maximilians-Universität München
Mar 1, 2022

Understanding how different epigenetic layers are coordinated to facilitate robust lineage decisions during development is one of the fundamental questions in regulatory genomics. Using single-cell epigenomics coupled with cell-type specific high-throughput mapping of enhancer activity, DNA methylation and the 3D genome landscape in vivo, we dissected how the epigenome is rewired during cortical development. We identified and functionally validated key transcription factors such as Neurog2 which underlie regulatory dynamics and coordinate rewiring across multiple epigenetic layers to ensure robust lineage specification. This work showcases the power of high-throughput integrative genomics to dissect the molecular rules of cell fate decisions in the brain and more broadly, how to apply them to evolution and disease.

SeminarNeuroscience

Studying cortical development through the lens of autism spectrum disorders

Gaia Novarino
Institute of Science and Technology Austria
Feb 22, 2022
SeminarNeuroscienceRecording

Malformation of cortical development: the genesis of epileptogenic networks

Alfonso Represa
INSERM, Institut de Neurobiologie de la Méditerranée
Jun 1, 2021

Malformations of cortical development (MCDs) result from alterations of one or combined developmental steps, including progenitors proliferation, neuronal migration and differentiation. They are important cause of childhood epilepsy and frequently associate cognitive deficits and behavioral alterations. Though the genetic basis of MCDs have known prominent progress during the past decade, including the identification of somatic, mosaic mutations responsible for focal MCDs, the pathophysiological mechanisms linking malformations to epileptogenesis remain elusive. In this seminar I will present data from my team and from the literature addressing this topic in two different MCDs types, the subcortical band heterotopia as a model of cortical migration defect and mTOR- dependent MCDs , that characterize by cortical dyslamination and neuronal differentiation defects.

SeminarNeuroscienceRecording

Understanding and treating epilepsy in tuberous sclerosis complex

Angelique Bordey
Yale University
May 4, 2021

Tuberous sclerosis complex (TSC) and focal cortical dysplasia type II (FCDII) are caused by mutations in mTOR pathway genes leading to mTOR hyperactivity, focal malformations of cortical development (fMCD), and seizures in 80-90% of the patients. The current definitive treatments for epilepsy are surgical resection or treatment with everolimus, which inhibits mTOR activity (only approved for TSC). Because both options have severe limitations, there is a major need to better understand the mechanisms leading to seizures to improve life-long epilepsy treatment in TSC and FCDII. To investigate such mechanisms, we recently developed a murine model of fMCD-associated epilepsy that recapitulates the human TSC and FCDII disorders. fMCD are defined by the presence of misplaced, dysmorphic cortical neurons expressing hyperactive mTOR – for simplicity we will refer to these as “mutant” neurons. In our model and in human TSC tissue, we made a surprising finding that mutant neurons express HCN4 channels, which are not normally functionally expressed in cortical neurons, and increased levels of filamin A (FLNA). FLNA is an actin-crossing linking molecule that has also multiple binding partners inside cells. These data led us to ask several important questions: (1) As HCN4 channels are responsible for the pacemaking activity of the heart, can HCN4 channel expression lead to repetitive firing of mutant neurons resulting in seizures? (2) HCN4 is the most cAMP-sensitive of the four HCN isoforms. Does increase in cAMP lead to the firing of mutant neurons? (3) Does increase in FLNA contribute to neuronal alterations and seizures? (4) Is the abnormal HCN4 and FLNA expression in mutant neurons due to mTOR? These questions will be discussed and addressed in the lecture.

SeminarNeuroscienceRecording

Molecular and activity-dependent mechanisms of cortical development underlying corpus callosum dysgenesis

Linda Richards
Queensland Brain Institute, University of Queensland
Feb 10, 2021
SeminarNeuroscienceRecording

Studying cortical development through the lens of human disorders

Gaia Novarino
Institute of Science and Technology Austria
Oct 21, 2020
SeminarNeuroscienceRecording

Common developmental mechanisms underlie multiple brain disorders linked to corpus callosum dysgenesis. (Simultaneous translation to Spanish)

Linda J. Richards AO, FAA, FAHMS, PhD.
Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
Oct 18, 2020

The corpus callosum is the largest fibre tract in the brain of placental mammals and connects the two cerebral hemispheres. Corpus callosum dysgenesis is a developmental brain disorder that is commonly genetic and occurs in approximately 1:4000 live births. It is easily diagnosed by MRI or prenatal ultrasound and is found in isolation or together with other brain anomalies, or with other organ system defects in a large number of different congenital syndromes. Callosal dysgenesis is a structural brain wiring disorder that can impact brain function and cognition in heterogeneous ways. We aim to understand how early developmental mechanisms lead to circuit alterations that ultimately impact behaviour and cognition. Translated to Spanish by MD and Medical interpreter Trinidad Ott. El cuerpo calloso es el tracto de fibras más grande del cerebro de los mamíferos placentarios y conecta los dos hemisferios cerebrales. La disgenesia del cuerpo calloso es un trastorno del desarrollo del cerebro que comunmente es genético y ocurre en aproximadamente 1: 4000 nacidos vivos. Se diagnostica fácilmente mediante resonancia magnética o ecografía prenatal y se encuentra aislado o junto con otras anomalías cerebrales, o con otros defectos del sistema de órganos en un gran número de síndromes congénitos diferentes. La disgenesia callosa es un trastorno estructural del cableado cerebral que puede afectar la función cerebral y la cognición de formas heterogéneas. Nuestro objetivo es comprender cómo los primeros mecanismos del desarrollo conducen a alteraciones en los circuitos que, en última instancia, afectan el comportamiento y la cognición. Traducción al español por la Doctora e Intérprete Médica Trinidad Ott.

SeminarNeuroscience

Misplaced and misconnected: circuit-level defects in malformations of cortical development

Jean-Bernard Manent
Mediterranean Institute of Neurobiology - INMED, Marseille, France
Jul 13, 2020

During histogenesis of the cerebral cortex, a proper laminar placement of defined numbers of specific cellular types is necessary to ensure proper functional connectivity patterns. There is a wide range of cortical malformations causing epilepsy and intellectual disability in humans, characterized with various degrees of neuronal misplacement, aberrant circuit organization or abnormal folding patterns. Although progress in human neurogenetics and brain imaging techniques have considerably advanced the identification of their causative genes, the pathophysiological mechanisms associated with defective cerebral cortex development remain poorly understood. In my presentation, I will outline some of our recent works in rodent models illustrating how misplaced neurons forming grey matter heterotopia, a cortical malformation subtype, interfere with the proper development of cortical circuits, and induce both local and distant circuitry changes associated with the subsequent emergence of epilepsy.

SeminarNeuroscienceRecording

RNA on the brain: Dynamic control of cortical development and disease

Debby Silver
Duke University Medical Center
Jun 24, 2020
SeminarNeuroscienceRecording

The thalamus that speaks to the cortex: spontaneous activity in the developing brain

Guillermina Lopez Bendito
Instituto de Neurociencias, Alicante (Spain)
Jun 21, 2020

Our research team runs several related projects studying the cellular and molecular mechanisms involved in the development of axonal connections in the brain. In particular, our aim is to uncover the principles underlying thalamocortical axonal wiring, maintenance and ultimately the rewiring of connections, through an integrated and innovative experimental programme. The development of the thalamocortical wiring requires a precise topographical sorting of its connections. Each thalamic nucleus receives specific sensory information from the environment and projects topographically to its corresponding cortical. A second level of organization is achieved within each area, where thalamocortical connections display an intra-areal topographical organization, allowing the generation of accurate spatial representations within each cortical area. Therefore, the level of organization and specificity of the thalamocortical projections is much more complex than other projection systems in the CNS. The central hypothesis of our laboratory is that thalamocortical input influences and maintains the functional architecture of the sensory cortices. We also believe that rewiring and plasticity events can be triggered by activity-dependent mechanisms in the thalamus. Three major questions are been focused in the laboratory: i) the role of spontaneous patterns of activity in thalamocortical wiring and cortical development, ii) the role of the thalamus and its connectivity in the neuroplastic cortical changes following sensory deprivation, and iii) reprogramming thalamic cells for sensory circuit restoration. Within these projects we are using several experimental programmes, these include: optical imaging, manipulation of gene expression in vivo, cell and molecular biology, biochemistry, cell culture, sensory deprivation paradigms and electrophysiology. The results derived from our investigations will contribute to our understating of how reprogramming of cortical wiring takes place following brain damage and how cortical structure is maintained.

SeminarNeuroscienceRecording

Species-specific mechanisms of the timing of human cortical development

Pierre Vanderhaeghen
VIB KULeuven Center for Brain & Disease Research
Jun 3, 2020

The human brain, in particular the cerebral cortex, has undergone rapid expansion and increased complexity during recent evolution. One striking feature of human corticogenesis is that it is highly protracted in time, from prenatal stages of neurogenesis (taking months instead of days in the mouse), to postnatal stages of neuronal maturation and circuit formation (taking years instead of weeks in the mouse). This prolonged development is thought to contribute in an important fashion to increased cortical size, but also enhanced circuit complexity and plasticity. Here we will discuss how the species-specific temporal patterning of corticogenesis is largely intrinsic to cortical progenitors and neurons, and involves human-specific genes and cell properties that underlie human brain evolution, as well as our selective sensitivity to certain brain diseases.

ePoster

Influenza-induced maternal inflammation and effect on cortical development

Nathalia Cardozo da Conceicao, Vera Nikovics, Navneet Vasistha, Konstantin Khodosevich

FENS Forum 2024

ePoster

Investigating the role of centrosome-cilia axis in human cortical development orchestration and malformations

Athanasia Rapti, Marta Labeur, George E. Baltatzis, Panagiotis Politis, Stavros Taraviras, Elisabeth B Binder, Silvia Cappello, Christina Kyrousi

FENS Forum 2024

ePoster

The maternal gut microbiota regulates embryonic cortical development in mice

Hugo Blair, Lorena Morales, Alexandre J.C. Cergneux, Jennifer Morael, Valentine Turpin, Jennifer Shearer, John F. Cryan, María R. Aburto

FENS Forum 2024

ePoster

The role of SMARCA2 (BRM) subunit of SWI/SNF/BAF complex in cortical development

HsiangYi Lai, Yi-Hsuan Liu, Jin-Wu Tsai

FENS Forum 2024

ePoster

A single-cell multiomic atlas of human cortical development in Down syndrome identifies candidate mechanisms underlying intellectual disability

Michael Lattke, Jon Conesa, Mickey Hughes, Vincenzo De Paola

FENS Forum 2024