Neurogenesis
neurogenesis
Dr. Hayder Amin
The position focused on developing a brain-inspired computational model using parallel, non-linear algorithms to investigate the neurogenesis complexity in large-scale systems. The successful applicant will specifically develop a neurogenic-plasticity-inspired bottom-up computational metamodel using our unique experimentally derived multidimensional parameters for a cortico-hippocampal circuit. The project aims to link computational modeling to experimental neuroscience to provide an explicit bidirectional prediction for complex performance and neurogenic network reserve for functional compensation to the brain demands in health and disease.
Cellular Crosstalk in Brain Development, Evolution and Disease
Cellular crosstalk is an essential process during brain development and is influenced by numerous factors, including cell morphology, adhesion, the local extracellular matrix and secreted vesicles. Inspired by mutations associated with neurodevelopmental disorders, we focus on understanding the role of extracellular mechanisms essential for the proper development of the human brain. Therefore, we combine 2D and 3D in vitro human models to better understand the molecular and cellular mechanisms involved in progenitor proliferation and fate, migration and maturation of excitatory and inhibitory neurons during human brain development and tackle the causes of neurodevelopmental disorders.
Rejuvenating the Alzheimer’s brain: Challenges & Opportunities
Pharmacological exploitation of neurotrophins and their receptors to develop novel therapeutic approaches against neurodegenerative diseases and brain trauma
Neurotrophins (NGF, BDNF, NT-3) are endogenous growth factors that exert neuroprotective effects by preventing neuronal death and promoting neurogenesis. They act by binding to their respective high-affinity, pro-survival receptors TrkA, TrkB or TrkC, as well as to p75NTR death receptor. While these molecules have been shown to significantly slow or prevent neurodegeneration, their reduced bioavailability and inability to penetrate the blood-brain-barrier limit their use as potential therapeutics. To bypass these limitations, our research team has developed and patented small-sized, lipophilic compounds which selectively resemble neurotrophins’ effects, presenting preferable pharmacological properties and promoting neuroprotection and repair against neurodegeneration. In addition, the combination of these molecules with 3D cultured human neuronal cells, and their targeted delivery in the brain ventricles through soft robotic systems, could offer novel therapeutic approaches against neurodegenerative diseases and brain trauma.
Cellular and genetic mechanisms of cerebral cortex folding
One of the most prominent features of the human brain is the fabulous size of the cerebral cortex and its intricate folding, both of which emerge during development. Over the last few years, work from my lab has shown that specific cellular and genetic mechanisms play central roles in cortex folding, particularly linked to neural stem and progenitor cells. Key mechanisms include high rates of neurogenesis, high abundance of basal Radial Glia Cells (bRGCs), and neuron migration, all of which are intertwined during development. We have also shown that primary cortical folds follow highly stereotyped patterns, defined by a spatial-temporal protomap of gene expression within germinal layers of the developing cortex. I will present recent findings from my laboratory revealing novel cellular and genetic mechanisms that regulate cortex expansion and folding. We have uncovered the contribution of epigenetic regulation to the establishment of the cortex folding protomap, modulating the expression levels of key transcription factors that control progenitor cell proliferation and cortex folding. At the single cell level, we have identified an unprecedented diversity of cortical progenitor cell classes in the ferret and human embryonic cortex. These are differentially enriched in gyrus versus sulcus regions and establish parallel cell lineages, not observed in mouse. Our findings show that genetic and epigenetic mechanisms in gyrencephalic species diversify cortical progenitor cell types and implement parallel cell linages, driving the expansion of neurogenesis and patterning cerebral cortex folds.
Astrocyte reprogramming / activation and brain homeostasis
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.
Epigenetic rewiring in Schinzel-Giedion syndrome
During life, a variety of specialized cells arise to grant the right and timely corrected functions of tissues and organs. Regulation of chromatin in defining specialized genomic regions (e.g. enhancers) plays a key role in developmental transitions from progenitors into cell lineages. These enhancers, properly topologically positioned in 3D space, ultimately guide the transcriptional programs. It is becoming clear that several pathologies converge in differential enhancer usage with respect to physiological situations. However, why some regulatory regions are physiologically preferred, while some others can emerge in certain conditions, including other fate decisions or diseases, remains obscure. Schinzel-Giedion syndrome (SGS) is a rare disease with symptoms such as severe developmental delay, congenital malformations, progressive brain atrophy, intractable seizures, and infantile death. SGS is caused by mutations in the SETBP1 gene that results in its accumulation further leading to the downstream accumulation of SET. The oncoprotein SET has been found as part of the histone chaperone complex INHAT that blocks the activity of histone acetyltransferases suggesting that SGS may (i) represent a natural model of alternative chromatin regulation and (ii) offer chances to study downstream (mal)adaptive mechanisms. I will present our work on the characterization of SGS in appropriate experimental models including iPSC-derived cultures and mouse.
Establishment and aging of the neuronal DNA methylation landscape in the hippocampus
The hippocampus is a brain region with key roles in memory formation, cognitive flexibility and emotional control. Yet hippocampal function is impaired severely during aging and in neurodegenerative diseases, and impairments in hippocampal function underlie age-related cognitive decline. Accumulating evidence suggests that the deterioration of the neuron-specific epigenetic landscape during aging contributes to their progressive, age-related dysfunction. For instance, we have recently shown that aging is associated with pronounced alterations of neuronal DNA methylation patterns in the hippocampus. Because neurons are generated mostly during development with limited replacement in the adult brain, they are particularly long-lived cells and have to maintain their cell-type specific gene expression programs life-long in order to preserve brain function. Understanding the epigenetic mechanisms that underlie the establishment and long-term maintenance of neuron-specific gene expression programs, will help us to comprehend the sources and consequences of their age-related deterioration. In this talk, I will present our recent work that investigated the role of DNA methylation in the establishment of neuronal gene expression programs and neuronal function, using adult neurogenesis in the hippocampus as a model. I will then describe the effects of aging on the DNA methylation landscape in the hippocampus and discuss the malleability of the aging neuronal methylome to lifestyle and environmental stimulation.
Circuit solutions for programming actions
The hippocampus is one of the few regions in the adult mammalian brain which is endowed with life-long neurogenesis. Despite intense investigation, it remains unclear how neurons newly-generated may retain unique functions that contribute to modulate hippocampal information processing and cognition. In this talk, I will present some recent findings revealing how enhanced forms of plasticity in adult-born neurons underlie the way they become incorporated into pre-existing networks in response to experience.
Epigenome regulation in neocortex expansion and generation of neuronal subtypes
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
Untitled Seminar
G. Quattrocolo: Cajal-Retzius cells in the postnatal hippocampus; F. Garcia-Moreno: Mosaic evolutionary history of brain circuits through the lens of neurogenesis
Retinal neurogenesis and lamination: What to become, where to become it and how to move from there!
The vertebrate retina is an important outpost of the central nervous system, responsible for the perception and transmission of visual information. It consists of five different types of neurons that reproducibly laminate into three layers, a process of crucial importance for the organ’s function. Unsurprisingly, impaired fate decisions as well as impaired neuronal migrations and lamination lead to impaired retinal function. However, how processes are coordinated at the cellular and tissue level and how variable or robust retinal formation is, is currently still underexplored. In my lab, we aim to shed light on these questions from different angles, studying on the one hand differentiation phenomena and their variability and on the other hand the downstream migration and lamination phenomena. We use zebrafish as our main model system due to its excellent possibilities for live imaging and quantitative developmental biology. More recently we also started to use human retinal organoids as a comparative system. We further employ cross disciplinary approaches to address these issues combining work of cell and developmental biology, biomechanics, theory and computer science. Together, this allows us to integrate cell with tissue-wide phenomena and generate an appreciation of the reproducibility and variability of events.
Spatio-temporal control of adult neurogenesis for on-demand brain plasticity
Stem cell approaches to understand acquired and genetic epilepsies
The Hsieh lab focuses on the mechanisms that promote neural stem cell self-renewal and differentiation in embryonic and adult brain. Using mouse models, video-EEG monitoring, viral techniques, and imaging/electrophysiological approaches, we elucidated many of the key transcriptional/epigenetic regulators of adult neurogenesis and showed aberrant new neuron integration in adult rodent hippocampus contribute to circuit disruption and seizure development. Building on this work, I will present our recent studies describing how GABA-mediated Ca2+ activity regulates the production of aberrant adult-born granule cells. In a new direction of my laboratory, we are using human induced pluripotent stem cells and brain organoid models as approaches to understand brain development and disease. Mutations in one gene, Aristaless-related homeobox (ARX), are of considerable interest since they are known to cause a common spectrum of neurodevelopmental disorders including epilepsy, autism, and intellectual disability. We have generated cortical and subpallial organoids from patients with poly-alanine expansion mutations in ARX. To understand the nature of ARX mutations in the organoid system, we are currently performing cellular, molecular, and physiological analyses. I will present these data to gain a comprehensive picture of the effect of ARX mutations in brain development. Since we do not understand how human brain development is affected by ARX mutations that contribute to epilepsy, we believe these studies will allow us to understand the mechanism of pathogenesis of ARX mutations, which has the potential to impact the diagnosis and care of patients.
Molecular and cellular mechanisms controlling neural stem cell activity
Neural stem cells (NSCs) generate new neurons throughout life. We use imaging-, genome editing-, and transgenesis-based approaches as well as cellular models of human diseases using pluripotent embryonic cells to study the molecular and cellular framework of NSC biology in the developing and adult brain. Aim of our research is to understand how physiologic and disease-associated alterations of neurogenesis are translated into stem cell-associated plastic changes in the developing and adult brain on a molecular, cellular, and behavioral level.
Converging mechanisms of epileptogenesis after brain injury
Traumatic brain injury (TBI), a leading cause of acquired epilepsy, results in primary cellular injury as well as secondary neurophysiological and inflammatory responses which contribute to epileptogenesis. I will present our recent studies identifying a role for neuro-immune interactions, specifically, the innate immune receptor Toll-like receptor 4 (TLR4), in enhancing network excitability and cell loss in hippocampal dentate gyrus early after concussive brain injury. I will describe results indicating that the transient post-traumatic increases in dentate neurogenesis which occurs during the same early post-injury period augments dentate network excitability and epileptogenesis. I will provide evidence for the beneficial effects of targeting TLR4 and neurogenesis early after brain injury in limiting epileptogenesis. We will discuss potential mechanisms for convergence of the post-traumatic neuro-immune and neurogenic changes and the implications for therapies to reduce neurological deficits and epilepsy after brain injury.
Neural stem cells as biomarkers of cognitive aging and dementia
Adult hippocampal neurogenesis is implicated in memory formation and mood regulation. The Thuret lab investigates environmental and molecular mechanisms controlling the production of these adult-born neurons and how they impact mental health. We study neurogenesis in healthy ageing as well as in the context of diseases such as Alzheimer’s and depression. By approaching neurogenesis in health and disease, the strategy is two folds: (i) Validating the neurogenic process as a target for prevention and pharmacological interventions. (ii) Developing neurogenesis as a biomarker of disease prediction and progression. In this talk, I will focus on presenting some recent human studies demonstrating how hippocampal neural stem cells fate can be used as biomarkers of cognitive aging and dementia.
Adult neurogenesis in mouse hippocampus
Dr. Aixa V. Morales has been working for more than 20 years in the field of Developmental Biology and from 2005, she is the PI of the laboratory on “Molecular Control of Neurogenesis” at Cajal Institute. Along these years, she has contributed to understanding the control of neurogenesis during development, the dorsoventral specification of neural progenitors, and the temporal control of the migration of neural crest cells. More recently, her lab interest moved towards understanding modulation of adult neurogenesis. Her lab current interest is the control of quiescence, as a mechanism of long-term neural stem cell maintenance in adult niches.
Gut Feelings: The Microbiota-Gut-Brain Axis Across the Lifespan
The microbiota-gut-brain axis is emerging as a research area of increasing interest for those investigating the biological and physiological basis of brain development and behaviour during early life, adolescence & ageing. The routes of communication between the gut and brain include the vagus nerve, the immune system, tryptophan metabolism, via the enteric nervous system or by way of microbial metabolites such as short chain fatty acids. Studies in animal models have shown that the development of an appropriate stress response is dependent on the microbiota. Developmentally, a variety of factors can impact the microbiota in early life including mode of birth delivery, antibiotic exposure, mode of nutritional provision, infection, stress as well as host genetics. Recently, the gut microbiota has been implicated in regulating the stress response, and social behaviour. Moreover, fundamental brain processes from adult hippocampal neurogenesis to myelination to microglia activation have been shown to be regulated by the microbiome. Further studies will focus on understanding the mechanisms underlying such brain effects and how they can be exploited by microbiota-targeted interventions including ‘psychobiotics’ and diet
Brain Awareness Week @ IITGN
Traumatic injury in the nervous system leads to devastating consequences such as paralysis. The regenerative capacity of the nervous system is limited in adulthood. In this talk, Dr. Anindya would be sharing how the simple nematode C. elegans with its known connectome can inform us about the biology of nervous system repair.
Novel mechanisms of neurogenesis and neural repair
In order to re-install neurogenesis after loss of neurons upon injury or neurodegeneration, we need to understand the basic principles of neurogenesis. I will first discuss about our discovery of a novel centrosome protein (Camargo et al., 2019) and discuss unpublished work about the great diversity of interphase centrosome proteomes and their relevance for neurodevelopmental disorders. I would then present work on a master regulator of neural stem cell amplification and brain folding (Stahl et al., 2013; Esgleas et al., 2020) to proceed presenting data on utilizing some of these factors for turning astrocytes into neurons. I will present data on the critical role of mitochondria in this conversion process (Gascon et al., 2016, Russo et al., 2020) and how it regulates the speed of conversion also showing unpublished data. If time permits I may touch on recent progress in in vivo reprogramming (Mattugini et al., 2019). Taken together, these data highlight the surprising specificity and importance of organelle diversity from centrosome, nucleolus and mitochondria as key regulators in development and reprogramming.
Vulnerable periods of brain development in ion channelopathies
Brain and neuronal network development depend on a complex sequence of events, which include neurogenesis, migration, differentiation, synaptogenesis, and synaptic pruning. Perturbations to any of these processes, for example associated with ion channel gene mutations (i.e., channelopathies), can underlie neurodevelopmental disorders such as neonatal and infantile epilepsies, strongly impair psychomotor development and cause persistent deficits in cognition, motor skills, or motor control. The therapeutic options available are very limited, and prophylactic therapies for patients at an increased risk of developing such epilepsies do not exist yet. By using genetic mouse models in which we controlled the activities of Kv7/M or HCN/h-channels during different developmental periods, we obtained offspring with distinct neurological phenotypes that could not simply be reversed by the re-introduction of the affected ion channel in juvenile or adult animals. The results indicate that channelopathy/mutation-specific treatments of neonatal and infantile epilepsies and their comorbidities need to be targeted to specific sensitive periods.
Genetic screening and modeling of human-specific neurogenesis in cerebral organoids
How development sculpts memory circuits
In mammals, the selective transformation of transient experience into stored memory occurs in the hippocampus, which develops representations of specific events in the context in which they occur. In this talk, I will focus on the development of hippocampal circuits and the self-organized dynamics embedded in them since the latter critically support the role of the hippocampus in memory. I will discuss evidence that adult hippocampal cells and circuits are remarkably sculpted by development, as early as embryonic neurogenesis. We argue that these primary developmental programs provide a scaffold onto which later experience of the external world can be grafted. Next, I will present data on the emergence of recurrent connectivity and self-organized dynamics in hippocampal circuits and outline the critical turn points and discontinuities in that developmental journey.
Adult Neurogenesis, Enriched Environments, and the Neurobiology of Early Life-style Dependent Resilience
“Changing Memory on the Fly, re-evaluation of learned behaviour I n Drosophila” “Metabolic Regulation of Neural Stem Cells” “The answer is in the sauce”
Species-specific mechanisms of the timing of human cortical development
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.
Neuroscience Investigations in the Virgin Lands of African Biodiversity
Africa is blessed with a rich diversity and abundance in rodent and avian populations. This natural endowment on the continent portends research opportunities to study unique anatomical profiles and investigate animal models that may confer better neural architecture to study neurodegenerative diseases, adult neurogenesis, stroke and stem cell therapies. To this end, African researchers are beginning to pay closer attention to some of her indigenous rodents and birds in an attempt to develop spontaneous laboratory models for homegrown neuroscience-based research. For this presentation, I will be showing studies in our lab, involving cellular neuroanatomy of two rodents, the African giant rat (AGR) and Greater cane rat (GCR), Eidolon Bats (EB) and also the Striped Owl (SO). Using histological stains (Cresyl violet and Rapid Golgi) and immunohistochemical biomarkers (GFAP, NeuN, CNPase, Iba-1, Collagen 2, Doublecortin, Ki67, Calbindin, etc), and Electron Microscopy, morphology and functional organizations of neuronal and glial populations of the AGR , GCR, EB and SO brains have been described, with our work ongoing. In addition, the developmental profiles of the prenatal GCR brains have been chronicled across its entire gestational period. Brains of embryos/foetuses were harvested for gross morphological descriptions and then processed using immunofluorescence biomarkers to determine the pattern, onset, duration and peak of neurogenesis (Pax6, Tbr1, Tbr2, NF, HuCD, MAP2) and the onset and peak of glial cell expressions and myelination in the prenatal GCR. The outcome of these research efforts has shown unique neuroanatomical expressions and networks amongst Africa’s rich biodiversity. It is hopeful that continuous effort in this regard will provide sufficient basic research data on neural developments and cellular neuroanatomy with subsequent translational consequences.
Adducins affect cortical neurogenesis by regulating basal progenitors' abundance
FENS Forum 2024
Adult hippocampal neurogenesis, the post-mortem delay, and prolonged aldehyde fixation: The enemies within
FENS Forum 2024
Adult neurogenesis regulates social dominance and anxiety
FENS Forum 2024
Adult neurogenesis in the Drosophila olfactory system
FENS Forum 2024
Analysis of differences in hippocampal adult neurogenesis induced by acute mild and severe seizures in young mice
FENS Forum 2024
Characterization of adult neurogenesis in Acomys cahirinus by lineage tracing analysis
FENS Forum 2024
The control of stem cell quiescence in early postnatal dentate gyrus is essential for establishing long-lasting adult hippocampal neurogenesis
FENS Forum 2024
DNA damage during early neurogenesis impairs interneuron migration without altering their ultimate cortical positioning
FENS Forum 2024
Dentate granule cell neurogenesis in adult mice following denervation in the entorhinal cortex lesion model
FENS Forum 2024
Does methamphetamine exposure alter hippocampal neurogenesis during preweaning period of laboratory rat?
FENS Forum 2024
Early life stress & the developmental dynamics of hypothalamic neurogenesis
FENS Forum 2024
Elucidating distinctive roles of Hippo pathway members YAP and TAZ in human in vitro neurogenesis
FENS Forum 2024
Environmental enrichment effects on hippocampal microglia and adult neurogenesis
FENS Forum 2024
Female microglia and neurogenesis respond differently to social defeat stress compared to males
FENS Forum 2024
Hippocampal neurogenesis changes in a sex and region-specific manner in adult mice subjected to maternal separation as an early life stress
FENS Forum 2024
Human pluripotent stem cell-derived ectomesenchymal cells promote neurogenesis and neurite outgrowth in rats with hypoxic-ischemic brain injury through the ERK/CREB signaling pathway
FENS Forum 2024
Hyperbaric oxygenation enhances neurogenesis in subventricular zone after traumatic brain injury
FENS Forum 2024
The impact of THC on neurogenesis and stress response: An in vitro investigation in astrocytes
FENS Forum 2024
Impact of type 2 diabetes and high-intensity interval exercise on neurogenesis, angiogenesis, and the accumulation of lipid droplets in the hippocampus
FENS Forum 2024
Increased GABAergic neurogenesis in human cortical organoids with schizophrenia-associated SETD1A mutations
FENS Forum 2024
Investigating the effect of meropenem on neurogenesis in adult rats
FENS Forum 2024
Metformin administration mitigates early life stress-induced disruptions in hippocampal neurogenesis
FENS Forum 2024
Nucleoporin 153 deficiency in adult neural stem cells defines a pathological protein-network signature and defective neurogenesis in a mouse model of Alzheimer’s disease
FENS Forum 2024
Olfactory neurogenesis after visual impairment: A role in sensory compensation?
FENS Forum 2024
Pharmacological activation of the dopamine D4 receptor prevents morphine-induced impairment of adult neurogenesis in the subventricular zone: Functional implications in odor discrimination learning
FENS Forum 2024
PIANP/LEDA-1 influences adult hippocampal neurogenesis and cerebellar morphology
FENS Forum 2024
New players in neurodevelopment: Role of meningeal macrophages in developmental neurogenesis
FENS Forum 2024
Pregnancy-responsive pools of adult neural stem cells for transient neurogenesis in mothers
FENS Forum 2024
Regulatory roles of mitochondria and metabolism in human and mouse neurogenesis
FENS Forum 2024
Sequential neurogenesis in zebrafish habenula gives rise to distinct functional microcircuits
FENS Forum 2024
Studying the effect of fosfomycin on hippocampal neurogenesis in adult rats
FENS Forum 2024
Telencephalic stab wound injury induces regenerative angiogenesis and neurogenesis in zebrafish: Unveiling the role of microglia along with VEGF and its expression
FENS Forum 2024
Unraveling the impact of high-caloric diets on juvenile rat hippocampal GABAergic system, neurogenesis, and astrocyte morphology
FENS Forum 2024
Viral encephalitis causes rapid transient depletion of neuronal progenitor cells and chronically alters neurogenesis in the adult mouse dentate gyrus
FENS Forum 2024