Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
EMBO J ; 42(17): e112847, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37365982

ABSTRACT

The paralogs CUL4A and CUL4B assemble cullin-RING E3 ubiquitin ligase (CRL) complexes regulating multiple chromatin-associated cellular functions. Although they are structurally similar, we found that the unique N-terminal extension of CUL4B is heavily phosphorylated during mitosis, and the phosphorylation pattern is perturbed in the CUL4B-P50L mutation causing X-linked intellectual disability (XLID). Phenotypic characterization and mutational analysis revealed that CUL4B phosphorylation is required for efficient progression through mitosis, controlling spindle positioning and cortical tension. While CUL4B phosphorylation triggers chromatin exclusion, it promotes binding to actin regulators and to two previously unrecognized CUL4B-specific substrate receptors (DCAFs), LIS1 and WDR1. Indeed, co-immunoprecipitation experiments and biochemical analysis revealed that LIS1 and WDR1 interact with DDB1, and their binding is enhanced by the phosphorylated N-terminal domain of CUL4B. Finally, a human forebrain organoid model demonstrated that CUL4B is required to develop stable ventricular structures that correlate with onset of forebrain differentiation. Together, our study uncovers previously unrecognized DCAFs relevant for mitosis and brain development that specifically bind CUL4B, but not the CUL4B-P50L patient mutant, by a phosphorylation-dependent mechanism.


Subject(s)
Mitosis , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Chromatin , Brain/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism
2.
Nat Rev Neurosci ; 22(4): 223-236, 2021 04.
Article in English | MEDLINE | ID: mdl-33633402

ABSTRACT

Neural stem cells (NSCs) generate new neurons throughout life in the mammalian brain. Adult-born neurons shape brain function, and endogenous NSCs could potentially be harnessed for brain repair. In this Review, focused on hippocampal neurogenesis in rodents, we highlight recent advances in the field based on novel technologies (including single-cell RNA sequencing, intravital imaging and functional observation of newborn cells in behaving mice) and characterize the distinct developmental steps from stem cell activation to the integration of newborn neurons into pre-existing circuits. Further, we review current knowledge of how levels of neurogenesis are regulated, discuss findings regarding survival and maturation of adult-born cells and describe how newborn neurons affect brain function. The evidence arguing for (and against) lifelong neurogenesis in the human hippocampus is briefly summarized. Finally, we provide an outlook of what is needed to improve our understanding of the mechanisms and functional consequences of adult neurogenesis and how the field may move towards more translational relevance in the context of acute and chronic neural injury and stem cell-based brain repair.


Subject(s)
Hippocampus/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Cell Proliferation/physiology , Humans
3.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35815653

ABSTRACT

Asymmetric segregation of cellular components regulates the fate and behavior of somatic stem cells. Similar to dividing budding yeast and precursor cells in Caenorhabditis elegans, it has been shown that mouse neural progenitors establish a diffusion barrier in the membrane of the endoplasmic reticulum (ER), which has been associated with asymmetric partitioning of damaged proteins and cellular age. However, the existence of an ER diffusion barrier in human cells remains unknown. Here, we used fluorescence loss in photobleaching (FLIP) imaging to show that human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived neural progenitor cells establish an ER diffusion barrier during cell division. The human ER diffusion barrier is regulated via lamin-dependent mechanisms and is associated with asymmetric segregation of mono- and polyubiquitylated damaged proteins. Further, forebrain regionalized organoids derived from hESCs were used to show the establishment of an ER membrane diffusion barrier in more naturalistic tissues, mimicking early steps of human brain development. Thus, the data provided here show that human neural progenitors establish a diffusion barrier during cell division in the membrane of the ER, which may allow for asymmetric segregation of cellular components, contributing to the fate and behavior of human neural progenitor cells.


Subject(s)
Cell Division , Endoplasmic Reticulum , Neural Stem Cells , Diffusion , Endoplasmic Reticulum/metabolism , Humans , Neural Stem Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-34996870

ABSTRACT

Fate and behavior of neural progenitor cells are tightly regulated during mammalian brain development. Metabolic pathways, such as glycolysis and oxidative phosphorylation, that are required for supplying energy and providing molecular building blocks to generate cells govern progenitor function. However, the role of de novo lipogenesis, which is the conversion of glucose into fatty acids through the multienzyme protein fatty acid synthase (FASN), for brain development remains unknown. Using Emx1Cre-mediated, tissue-specific deletion of Fasn in the mouse embryonic telencephalon, we show that loss of FASN causes severe microcephaly, largely due to altered polarity of apical, radial glia progenitors and reduced progenitor proliferation. Furthermore, genetic deletion and pharmacological inhibition of FASN in human embryonic stem cell-derived forebrain organoids identifies a conserved role of FASN-dependent lipogenesis for radial glia cell polarity in human brain organoids. Thus, our data establish a role of de novo lipogenesis for mouse and human brain development and identify a link between progenitor-cell polarity and lipid metabolism.


Subject(s)
Brain/metabolism , Fatty Acid Synthases/metabolism , Lipogenesis/physiology , Animals , Body Patterning , Brain/growth & development , Brain/pathology , Fatty Acid Synthase, Type I , Fatty Acid Synthases/genetics , Humans , Lipid Metabolism , Lipogenesis/genetics , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Transcriptome
5.
Am J Pathol ; 193(12): 2144-2155, 2023 12.
Article in English | MEDLINE | ID: mdl-37741454

ABSTRACT

Mechanisms responsible for the pathogenesis of diabetic retinal disease remain incompletely understood, but they likely involve multiple cellular targets, including photoreceptors. Evidence suggests that dysregulated de novo lipogenesis in photoreceptors is a critical early target of diabetes. Following on this observation, the present study aimed to determine whether two interventions shown to improve diabetic retinopathy in mice-pharmacologic visual cycle inhibition and prolonged dark adaptation-reduce photoreceptor anabolic lipid metabolism. Elevated retinal lipid biosynthetic signaling was observed in two mouse models of diabetes, with both models showing reduced retinal AMP-activated kinase (AMPK) signaling, elevated acetyl CoA carboxylase (ACC) signaling, and increased activity of fatty acid synthase, which promotes lipotoxicity in photoreceptors. Although retinal AMPK-ACC axis signaling was dependent on systemic glucose fluctuations in healthy animals, mice with diabetes lacked such regulation. Visual cycle inhibition and prolonged dark adaptation reversed abnormal retinal AMPK-ACC signaling in mice with diabetes. Although visual cycle inhibition reduced the severity of diabetic retinopathy in control mice, as assessed by retinal capillary atrophy, this intervention was ineffective in fatty acid synthase gain-of-function mice. These results suggest that early diabetic retinopathy is characterized by glucose-driven elevations in retinal lipid biosynthetic activity, and that two interventions known to increase photoreceptor glucose demands alleviate disease by reversing these signals.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Retinal Degeneration , Mice , Animals , AMP-Activated Protein Kinases/metabolism , Diabetic Retinopathy/metabolism , Glucose , Fatty Acid Synthases , Lipids
6.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34244440

ABSTRACT

Oligodendrocyte precursor cells (OPCs) retain the capacity to remyelinate axons in the corpus callosum (CC) upon demyelination. However, the dynamics of OPC activation, mode of cell division, migration, and differentiation on a single-cell level remain poorly understood due to the lack of longitudinal observations of individual cells within the injured brain. After inducing focal demyelination with lysophosphatidylcholin in the CC of adult mice, we used two-photon microscopy to follow for up to 2 mo OPCs and their differentiating progeny, genetically labeled through conditional recombination driven by the regulatory elements of the gene Achaete-scute homolog 1. OPCs underwent several rounds of symmetric and asymmetric cell divisions, producing a subset of daughter cells that differentiates into myelinating oligodendrocytes. While OPCs continue to proliferate, differentiation into myelinating oligodendrocytes declines with time, and death of OPC-derived daughter cells increases. Thus, chronic in vivo imaging delineates the cellular principles leading to remyelination in the adult brain, providing a framework for the development of strategies to enhance endogenous brain repair in acute and chronic demyelinating disease.


Subject(s)
Aging/physiology , Corpus Callosum/diagnostic imaging , Imaging, Three-Dimensional , Remyelination/physiology , Animals , Cell Death , Cell Division , Cell Movement , Clone Cells , Mice , Oligodendrocyte Precursor Cells/cytology
7.
Hippocampus ; 33(4): 402-411, 2023 04.
Article in English | MEDLINE | ID: mdl-36256493

ABSTRACT

Neural stem cells (NSCs) generate new neurons throughout life in the mammalian hippocampus. The distinct developmental steps in the course of adult neurogenesis, including NSC activation, expansion, and neuronal integration, are increasingly well characterized down to the molecular level. However, substantial gaps remain in our knowledge about regulators and mechanisms involved in this biological process. This review highlights three long-standing unknowns. First, we discuss potency and identity of NSCs and the quest for a unifying model of short- and long-term self-renewal dynamics. Next, we examine cell death, specifically focusing on the early demise of newborn cells. Then, we outline the current knowledge on cell integration dynamics, discussing which (if any) neurons are replaced by newly added neurons in the hippocampal circuits. For each of these unknowns, we summarize the trajectory of studies leading to the current state of knowledge. Finally, we offer suggestions on how to fill the remaining gaps by taking advantage of novel technology to reveal currently hidden secrets in the course of adult hippocampal neurogenesis.


Subject(s)
Hippocampus , Neural Stem Cells , Adult , Humans , Animals , Infant, Newborn , Hippocampus/physiology , Neurogenesis/physiology , Neurons/physiology , Neural Stem Cells/physiology , Mammals
8.
EMBO J ; 38(17): e103013, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31432524

ABSTRACT

The activity of stem cells in the adult brain is controlled by various niche-dependent mechanisms. A new article by Lepko et al (2019) shows that proliferation of neural stem cells in the ventricular-subventricular zone is regulated by choroid plexus-derived miR-204, identifying a novel mechanism of how the delicate balance between stem cell quiescence and activation is controlled.


Subject(s)
MicroRNAs , Neural Stem Cells , Adult , Brain , Choroid Plexus , Humans , Stem Cell Niche
9.
Nature ; 542(7639): 49-54, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28024299

ABSTRACT

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Lymphangiogenesis , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Acetates/pharmacology , Acetyl Coenzyme A/metabolism , Acetylation/drug effects , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epigenesis, Genetic , Female , Histones/metabolism , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymphangiogenesis/drug effects , Lymphangiogenesis/genetics , Lymphatic Vessels/drug effects , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Protein Biosynthesis , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Umbilical Arteries/cytology , Up-Regulation
10.
J Biol Chem ; 297(3): 101104, 2021 09.
Article in English | MEDLINE | ID: mdl-34425110

ABSTRACT

Diabetic retinopathy (DR) is an increasingly frequent cause of blindness across populations; however, the events that initiate pathophysiology of DR remain elusive. Strong preclinical and clinical evidence suggests that abnormalities in retinal lipid metabolism caused by diabetes may account for the origin of this disease. A major arm of lipid metabolism, de novo biosynthesis, is driven by elevation in available glucose, a common thread binding all forms of vision loss in diabetes. Therefore, we hypothesized that aberrant retinal lipid biogenesis is an important promoter of early DR. In murine models, we observed elevations of diabetes-associated retinal de novo lipogenesis ∼70% over control levels. This shift was primarily because of activation of fatty acid synthase (FAS), a rate-limiting enzyme in the biogenic pathway. Activation of FAS was driven by canonical glucose-mediated disinhibition of acetyl-CoA carboxylase, a major upstream regulatory enzyme. Mutant mice expressing gain-of-function FAS demonstrated increased vulnerability to DR, whereas those with FAS deletion in rod photoreceptors maintained preserved visual responses upon induction of diabetes. Excess retinal de novo lipogenesis-either because of diabetes or because of FAS gain of function-was associated with modestly increased levels of palmitate-containing phosphatidylcholine species in synaptic membranes, a finding with as yet uncertain significance. These findings implicate glucose-dependent increases in photoreceptor de novo lipogenesis in the early pathogenesis of DR, although the mechanism of deleterious action of this pathway remains unclear.


Subject(s)
Diabetic Retinopathy/etiology , Lipogenesis/physiology , Photoreceptor Cells, Vertebrate/physiology , Acetyl-CoA Carboxylase/metabolism , Animals , Diabetes Mellitus/metabolism , Diabetic Retinopathy/metabolism , Fatty Acid Synthases/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Lipid Metabolism/physiology , Mice , Mice, Inbred C57BL , Photoreceptor Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retina/pathology
11.
Proc Natl Acad Sci U S A ; 116(51): 25688-25696, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31772009

ABSTRACT

Neural stem cells (NSCs) generate neurons and glial cells throughout embryonic and postnatal brain development. The role of S-palmitoylation (also referred to as S-acylation), a reversible posttranslational lipid modification of proteins, in regulating the fate and activity of NSCs remains largely unknown. We used an unbiased screening approach to identify proteins that are S-acylated in mouse NSCs and showed that bone morphogenic protein receptor 1a (BMPR1a), a core mediator of BMP signaling, is palmitoylated. Genetic manipulation of S-acylated sites affects the localization and trafficking of BMPR1a and leads to altered BMP signaling. Strikingly, defective palmitoylation of BMPR1a modulates NSC function within the mouse brain, resulting in enhanced oligodendrogenesis. Thus, we identified a mechanism regulating the behavior of NSCs and provided the framework to characterize dynamic posttranslational lipid modifications of proteins in the context of NSC biology.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Lipoylation/physiology , Neural Stem Cells , Neurogenesis/physiology , Animals , Bone Morphogenetic Protein Receptors, Type I/chemistry , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cells, Cultured , Mice , Neural Stem Cells/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
12.
Nature ; 493(7431): 226-30, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23201681

ABSTRACT

Mechanisms controlling the proliferative activity of neural stem and progenitor cells (NSPCs) have a pivotal role to ensure life-long neurogenesis in the mammalian brain. How metabolic programs are coupled with NSPC activity remains unknown. Here we show that fatty acid synthase (Fasn), the key enzyme of de novo lipogenesis, is highly active in adult NSPCs and that conditional deletion of Fasn in mouse NSPCs impairs adult neurogenesis. The rate of de novo lipid synthesis and subsequent proliferation of NSPCs is regulated by Spot14, a gene previously implicated in lipid metabolism, that we found to be selectively expressed in low proliferating adult NSPCs. Spot14 reduces the availability of malonyl-CoA, which is an essential substrate for Fasn to fuel lipogenesis. Thus, we identify here a functional coupling between the regulation of lipid metabolism and adult NSPC proliferation.


Subject(s)
Adult Stem Cells/metabolism , Fatty Acid Synthases/metabolism , Lipogenesis , Neural Stem Cells/metabolism , Adult Stem Cells/cytology , Animals , Cell Proliferation , Dentate Gyrus/metabolism , Fatty Acid Synthases/deficiency , Fatty Acid Synthases/genetics , Gene Expression Profiling , Hippocampus/cytology , Hippocampus/metabolism , Malonyl Coenzyme A/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
J Neurosci ; 36(28): 7407-14, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27413151

ABSTRACT

UNLABELLED: The hippocampal dentate gyrus is critically involved in learning and memory. However, methods for imaging the activity of its principal neurons, the dentate gyrus granule cells, are missing. Here we demonstrate chronic two-photon imaging of granule cell population activity in awake mice using a cortical window implant that leaves the hippocampal formation intact and does not lead to obvious alteration of animal behavior. Using virus delivery, we targeted expression of genetically encoded calcium indicators specifically to dentate gyrus granule cells. Calcium imaging of granule cell activity 600-800 µm below the hippocampal surface was facilitated by using 1040 nm excitation of the red indicator R-CaMP1.07, but was also achieved using the green indicator GCaMP6s. We found that the rate of calcium transients was increased during wakefulness relative to an extremely low rate during anesthesia; however, activity still remained sparse with, on average, approximately one event per 2-5 min per cell across the granule cell population. Comparing periods of running on a ladder wheel and periods of resting, we furthermore identified state-dependent differences in the active granule cell population, with some cells displaying highest activity level during running and others during resting. Typically, cells did not maintain a clear state preference in their activity pattern across days. Our approach opens new avenues to elucidate granule cell function, plasticity mechanisms, and network computation in the adult dentate gyrus. SIGNIFICANCE STATEMENT: We describe a technique that allows for chronic, functional imaging of dentate gyrus granule cells in awake, behaving mice in an intact hippocampal circuitry using genetically encoded calcium indicators. This novel approach enables the analyses of individual granule cell activity over time and provides a powerful tool to elucidate the mechanisms underlying structural and functional plasticity of the adult dentate gyrus.


Subject(s)
Dentate Gyrus/cytology , Nerve Net/physiology , Neurons/physiology , Animals , Calcium/metabolism , Dentate Gyrus/diagnostic imaging , Exploratory Behavior/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Nerve Net/diagnostic imaging , Neurons/classification , Optogenetics , Wakefulness
14.
EMBO J ; 32(13): 1793-5, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23727887

ABSTRACT

During evolution, the mammalian brain massively expanded its size. However, the exact roles of distinct neural precursors, identified in the developing cortex during embryogenesis, for size expansion and surface folding (i.e., gyration) remain largely unknown. New findings by Nonaka-Kinoshita et al advance our understanding of embryonic neural precursor function by identifying cell type-selective functions for size expansion and folding, and challenge previously held concepts of mammalian brain development.


Subject(s)
Brain/physiology , Cell Differentiation , Cerebral Cortex/physiology , Embryo, Mammalian/physiology , Intermediate Filament Proteins/physiology , Nerve Tissue Proteins/physiology , Stem Cells/physiology , Animals , Nestin
15.
Development ; 141(10): 1983-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24803647

ABSTRACT

New neurons are generated throughout life in distinct regions of the mammalian brain. This process, called adult neurogenesis, has been implicated in physiological brain function, and failing or altered neurogenesis has been associated with a number of neuropsychiatric diseases. Here, we provide an overview of the mechanisms governing the neurogenic process in the adult brain and describe how new neurons may contribute to brain function in health and disease.


Subject(s)
Adult Stem Cells/physiology , Brain/growth & development , Neural Stem Cells/physiology , Neurogenesis/physiology , Adult , Animals , Brain/cytology , Brain/physiology , Humans , Mice , Signal Transduction/physiology , Stem Cell Niche/physiology
16.
Dev Dyn ; 245(7): 702-9, 2016 07.
Article in English | MEDLINE | ID: mdl-26890418

ABSTRACT

We here review the existing evidence linking adult hippocampal neurogenesis and human brain function in physiology and disease. Furthermore, we aim to point out where evidence is missing, highlight current promising avenues of investigation, and suggest future tools and approaches to foster the link between life-long neurogenesis and human brain function. Developmental Dynamics 245:702-709, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Neurogenesis/physiology , Adult , Brain/cytology , Brain/metabolism , Brain/physiology , Hippocampus/metabolism , Humans , Neurogenesis/genetics
17.
Development ; 140(13): 2823-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23720045

ABSTRACT

Neural stem/progenitor cells (NSPCs) generate new neurons throughout life in the mammalian hippocampus. Newborn granule cells mature over several weeks to functionally integrate into the pre-existing neural circuitry. Even though an increasing number of genes that regulate neuronal polarization and neurite extension have been identified, the cellular mechanisms underlying the extension of neurites arising from newborn granule cells remain largely unknown. This is mainly because of the current lack of longitudinal observations of neurite growth within the endogenous niche. Here we used a novel slice culture system of the adult mouse hippocampal formation combined with in vivo retroviral labeling of newborn neurons and longitudinal confocal imaging to analyze the mode and velocity of neurite growth extending from immature granule cells. Using this approach we show that dendritic processes show a linear growth pattern with a speed of 2.19±0.2 µm per hour, revealing a much faster growth dynamic than expected by snapshot-based in vivo time series. Thus, we here identified the growth pattern of neurites extending from newborn neurons within their niche and describe a novel technology that will be useful to monitor neuritic growth in physiological and disease states that are associated with altered dendritic morphology, such as rodent models of epilepsy.


Subject(s)
Neurogenesis/physiology , Neurons/cytology , Animals , Cell Differentiation/physiology , Dentate Gyrus/cytology , Hippocampus/cytology , In Vitro Techniques , Mice , Neurites , Neurogenesis/genetics , Neurons/metabolism
18.
Proc Natl Acad Sci U S A ; 110(17): 7062-7, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569253

ABSTRACT

New neurons are continuously added to the dentate gyrus of the adult mammalian brain. During the critical period of a few weeks after birth when newborn neurons progressively mature, a restricted fraction is competitively selected to survive in an experience-dependent manner, a condition for their contribution to memory processes. The mechanisms that control critical stages of experience-dependent functional incorporation of adult newborn neurons remain largely unknown. Here, we identify a unique transcriptional regulator of the functional integration of newborn neurons, the inducible immediate early gene zif268/egr1. We show that newborn neurons in zif268-KO mice undergo accelerated death during the critical period of 2-3 wk around their birth and exhibit deficient neurochemical and morphological maturation, including reduced GluR1 expression, increased NKCC1/KCC2b chloride cotransporter ratio, altered dendritic development, and marked spine growth defect. Investigating responsiveness of newborn neurons to activity-dependent expression of zif268 in learning, we demonstrate that in the absence of zif268, training in a spatial learning task during this critical period fails to recruit newborn neurons and promote their survival, leading to impaired long-term memory. This study reveals a previously unknown mechanism for the control of the selection, functional maturation, and experience-dependent recruitment of dentate gyrus newborn neurons that depends on the inducible immediate early gene zif268, processes that are critical for their contribution to hippocampal-dependent long-term memory.


Subject(s)
Dentate Gyrus/growth & development , Early Growth Response Protein 1/genetics , Maze Learning/physiology , Neurogenesis/physiology , Transcription Factors/genetics , Analysis of Variance , Animals , Bromodeoxyuridine , Dentate Gyrus/chemistry , Early Growth Response Protein 1/physiology , Immunohistochemistry , Mice , Mice, Knockout , Receptors, AMPA/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Symporters/metabolism , Transcription Factors/physiology , K Cl- Cotransporters
19.
Transfus Med Hemother ; 43(5): 321-326, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27781019

ABSTRACT

Acute or chronic injury of the adult mammalian brain is often associated with persistent functional deficits as its potential for regeneration and capacity to rebuild lost neural structures is limited. However, the discovery that neural stem cells (NSCs) persist throughout life in discrete regions of the brain, novel approaches to induce the formation of neuronal and glial cells, and recently developed strategies to generate tissue for exogenous cell replacement strategies opened novel perspectives how to regenerate the adult brain. Here, we will review recently developed approaches for brain repair and discuss future perspectives that may eventually allow for developing novel treatment strategies in acute and chronic brain injury.

20.
J Neurosci ; 34(15): 5222-32, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24719101

ABSTRACT

Controlling neural stem and progenitor cell (NSPC) proliferation is critical to maintain neurogenesis in the mammalian brain throughout life. However, it remains poorly understood how niche-derived cues such as ß1-integrin-mediated signaling are translated into NSPC-intrinsic molecular changes to regulate NSPC activity. Here we show that genetic deletion of integrin-linked kinase (ILK) increases NSPC proliferation through PINCH1/2-dependent enhancement of c-Jun N-terminal protein kinase activity in both neurogenic regions of the adult mouse brain. This effect downstream of ILK signaling is mediated through loss of Ras suppressor unit-1 (RSU-1), as virus-based reconstitution of RSU-1 expression rescued the ILK-dependent effects on NSPC proliferation. Thus, we here identified an intracellular signaling cascade linking extrinsic integrin-mediated signaling to NSPC proliferation and characterized a novel mechanism that regulates NSPC activity in the adult mammalian brain.


Subject(s)
Brain/metabolism , Cell Proliferation , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/growth & development , Cells, Cultured , Female , Gene Deletion , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , MAP Kinase Signaling System , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Protein Serine-Threonine Kinases/genetics , Stem Cell Niche , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL