1
|
Hosseini SM, Panahi-Azar A, Sheybani-Arani M, Morovatshoar R, Mirzadeh M, Salimi Asl A, Naghdipour Mirsadeghi M, Khajavi-Mayvan F. Vitamins, minerals and their maternal levels' role in brain development: An updated literature-review. Clin Nutr ESPEN 2024; 63:31-45. [PMID: 38907995 DOI: 10.1016/j.clnesp.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 03/26/2024] [Accepted: 05/16/2024] [Indexed: 06/24/2024]
Abstract
One's neurobehavioural and mental health are built during the exact and complex process of brain development. It is thought that fetal development is where neuropsychiatric disorders first emerged. Behavioural patterns can change as a result of neuropsychiatric illnesses. The incidence is rising quickly; nevertheless, providing exceptional care remains a significant challenge for families and healthcare systems. It has been demonstrated that one of the main factors causing the transmission of these diseases is maternal exposure. Through physiologic pathways, maternal health and intrauterine exposures can affect brain development. Our attention has been focused on epigenetic factors, particularly in the gestational environment, which may be responsible for human neurodegenerative diseases since our main mental development occurs during the nine months of intrauterine life. After thoroughly searching numerous databases, this study examined the effect of fat-soluble vitamins, water-soluble vitamins, and minerals and their maternal-level effect on brain development.
Collapse
Affiliation(s)
| | - Ava Panahi-Azar
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | | | - Reza Morovatshoar
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Mahdieh Mirzadeh
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Ali Salimi Asl
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Misa Naghdipour Mirsadeghi
- Department of Gynecology, School of Medicine, Reproductive Health Research Center, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | | |
Collapse
|
2
|
von Bohlen Und Halbach O. Neurotrophic Factors and Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:223-254. [PMID: 37962797 DOI: 10.1007/978-3-031-36159-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spines are highly dynamic structures that play important roles in neuronal plasticity. The morphologies and the numbers of dendritic spines are highly variable, and this diversity is correlated with the different morphological and physiological features of this neuronal compartment. Dendritic spines can change their morphology and number rapidly, allowing them to adapt to plastic changes. Neurotrophic factors play important roles in the brain during development. However, these factors are also necessary for a variety of processes in the postnatal brain. Neurotrophic factors, especially members of the neurotrophin family and the ephrin family, are involved in the modulation of long-lasting effects induced by neuronal plasticity by acting on dendritic spines, either directly or indirectly. Thereby, the neurotrophic factors play important roles in processes attributed, for example, to learning and memory.
Collapse
|
3
|
Gross I, Brandt N, Vonk D, Köper F, Wöhlbrand L, Rabus R, Witt M, Heep A, Plösch T, Hipp MS, Bräuer AU. Plasticity-Related Gene 5 Is Expressed in a Late Phase of Neurodifferentiation After Neuronal Cell-Fate Determination. Front Cell Neurosci 2022; 16:797588. [PMID: 35496908 PMCID: PMC9053830 DOI: 10.3389/fncel.2022.797588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/17/2022] [Indexed: 12/27/2022] Open
Abstract
During adult neurogenesis, neuronal stem cells differentiate into mature neurons that are functionally integrated into the existing network. One hallmark during the late phase of this neurodifferentiation process is the formation of dendritic spines. These morphological specialized structures form the basis of most excitatory synapses in the brain, and are essential for neuronal communication. Additionally, dendritic spines are affected in neurological disorders, such as Alzheimer’s disease or schizophrenia. However, the mechanisms underlying spinogenesis, as well as spine pathologies, are poorly understood. Plasticity-related Gene 5 (PRG5), a neuronal transmembrane protein, has previously been linked to spinogenesis in vitro. Here, we analyze endogenous expression of the PRG5 protein in different mouse brain areas, as well as on a subcellular level. We found that native PRG5 is expressed dendritically, and in high abundance in areas characterized by their regenerative capacity, such as the hippocampus and the olfactory bulb. During adult neurogenesis, PRG5 is specifically expressed in a late phase after neuronal cell-fate determination associated with dendritic spine formation. On a subcellular level, we found PRG5 not to be localized at the postsynaptic density, but at the base of the synapse. In addition, we showed that PRG5-induced formation of membrane protrusions is independent from neuronal activity, supporting a possible role in the morphology and stabilization of spines.
Collapse
Affiliation(s)
- Isabel Gross
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Nicola Brandt
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Danara Vonk
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Franziska Köper
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Perinatal Neurobiology Research Group, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Lars Wöhlbrand
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Ralf Rabus
- General and Molecular Microbiology, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Martin Witt
- Department of Anatomy, University Medical Center Rostock, Rostock, Germany
| | - Axel Heep
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Perinatal Neurobiology Research Group, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Torsten Plösch
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Perinatal Neurobiology Research Group, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Mark S. Hipp
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anja U. Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- School of Medicine and Health Sciences, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
- *Correspondence: Anja U. Bräuer,
| |
Collapse
|
4
|
Chondroitin sulfate proteoglycan-5 forms perisynaptic matrix assemblies in the adult rat cortex. Cell Signal 2020; 74:109710. [PMID: 32653642 DOI: 10.1016/j.cellsig.2020.109710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022]
Abstract
Composition of the brain extracellular matrix changes in time as maturation proceeds. Chondroitin sulfate proteoglycan 5 (CSPG-5), also known as neuroglycan C, has been previously associated to differentiation since it shapes neurite growth and synapse forming. Here, we show that this proteoglycan persists in the postnatal rat brain, and its expression is higher in cortical regions with plastic properties, including hippocampus and the medial prefrontal cortex at the end of the second postnatal week. Progressively accumulating after birth, CSPG-5 typically concentrates around glutamatergic and GABAergic terminals in twelve-week old rat hippocampus. CSPG-5-containing perisynaptic matrix rings often appear at the peripheral margin of perineuronal nets. Electron microscopy and analysis of synaptosomal fraction showed that CSPG-5 accumulates around, and is associated to synapses, respectively. In vitro analyses suggest that neurons, but less so astrocytes, express CSPG-5 in rat primary neocortical cultures, and CSPG-5 produced by transfected neuroblastoma cells appear at endings and contact points of neurites. In human subjects, CSPG-5 expression shifts in brain areas of the default mode network of suicide victims, which may reflect an impact in the pathogenesis of psychiatric diseases or support diagnostic power.
Collapse
|
5
|
Winokurow N, Schumacher S. A role for polycystin-1 and polycystin-2 in neural progenitor cell differentiation. Cell Mol Life Sci 2019; 76:2851-2869. [PMID: 30895336 PMCID: PMC11105687 DOI: 10.1007/s00018-019-03072-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/17/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
Polycystin-1 (PC1) and polycystin-2 (PC2) are transmembrane proteins encoded by the Pkd1 and Pkd2 genes, respectively. Mutations in these genes are causative for the development of autosomal-dominant polycystic kidney disease. A prominent feature of this disease is an unbalanced cell proliferation. PC1 and PC2 physically interact to form a complex, which localizes to the primary cilia of renal epithelial cells. Recently, PC1 and PC2 have also been described to be present in primary cilia of radial glial cells (RGCs) and to contribute to the planar cell polarity of late RGCs and E1 ependymal cells. As neural progenitor cells (NPCs), early RGCs have to balance proliferation for expansion, or for self-renewal and differentiation to generate neurons. It is not known whether the polycystins play a role in this process. Here, we show that PC1 and PC2 are expressed in RGCs of the developing mouse cerebral cortex during neurogenesis. Loss-of-function analysis and cell-based assays reveal that a reduction of PC1 or PC2 expression leads to increased NPC proliferation, while the differentiation to neurons becomes impaired. The increased NPC proliferation is preceded by enhanced Notch signaling and accompanied by a rise in the number of symmetric cell divisions. The transcription factor STAT3 seems to be mechanistically important for polycystin signaling in NPCs as either STAT3 knockdown or inhibition of STAT3 function abrogates the increased proliferation driven by reduced polycystin expression. Our findings indicate that PC1 and PC2 are critical for maintaining a balance between proliferation and differentiation of NPCs.
Collapse
Affiliation(s)
- Natalie Winokurow
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
6
|
Hillen AEJ, Burbach JPH, Hol EM. Cell adhesion and matricellular support by astrocytes of the tripartite synapse. Prog Neurobiol 2018; 165-167:66-86. [PMID: 29444459 DOI: 10.1016/j.pneurobio.2018.02.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/25/2017] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Astrocytes contribute to the formation, function, and plasticity of synapses. Their processes enwrap the neuronal components of the tripartite synapse, and due to this close interaction they are perfectly positioned to modulate neuronal communication. The interaction between astrocytes and synapses is facilitated by cell adhesion molecules and matricellular proteins, which have been implicated in the formation and functioning of tripartite synapses. The importance of such neuron-astrocyte integration at the synapse is underscored by the emerging role of astrocyte dysfunction in synaptic pathologies such as autism and schizophrenia. Here we review astrocyte-expressed cell adhesion molecules and matricellular molecules that play a role in integration of neurons and astrocytes within the tripartite synapse.
Collapse
Affiliation(s)
- Anne E J Hillen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands; Department of Pediatrics/Child Neurology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Yu Q, Wang B, Zhao T, Zhang X, Tao L, Shi J, Sun X, Ding Q. NaHS Protects against the Impairments Induced by Oxygen-Glucose Deprivation in Different Ages of Primary Hippocampal Neurons. Front Cell Neurosci 2017; 11:67. [PMID: 28326019 PMCID: PMC5339257 DOI: 10.3389/fncel.2017.00067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 02/24/2017] [Indexed: 01/16/2023] Open
Abstract
Brain ischemia leads to poor oxygen supply, and is one of the leading causes of brain damage and/or death. Neuroprotective agents are thus in great need for treatment purpose. Using both young and aged primary cultured hippocampal neurons as in vitro models, we investigated the effect of sodium hydrosulfide (NaHS), an exogenous donor of hydrogen sulfide, on oxygen-glucose deprivation (OGD) damaged neurons that mimick focal cerebral ischemia/reperfusion (I/R) induced brain injury. NaHS treatment (250 μM) protected both young and aged hippocampal neurons, as indicated by restoring number of primary dendrites by 43.9 and 68.7%, number of dendritic end tips by 59.8 and 101.1%, neurite length by 36.8 and 66.7%, and spine density by 38.0 and 58.5% in the OGD-damaged young and aged neurons, respectively. NaHS treatment inhibited growth-associated protein 43 downregulation, oxidative stress in both young and aged hippocampal neurons following OGD damage. Further studies revealed that NaHS treatment could restore ERK1/2 activation, which was inhibited by OGD-induced protein phosphatase 2 (PP2A) upregulation. Our results demonstrated that NaHS has potent protective effects against neuron injury induced by OGD in both young and aged hippocampal neurons.
Collapse
Affiliation(s)
- Qian Yu
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Binrong Wang
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Tianzhi Zhao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Xiangnan Zhang
- Division of Scientific Research, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Lei Tao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Jinshan Shi
- Department of Anesthesiology, Guizhou Provincial People's Hospital Guiyang, China
| | - Xude Sun
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Qian Ding
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| |
Collapse
|
8
|
Ambrogini P, Betti M, Galati C, Di Palma M, Lattanzi D, Savelli D, Galli F, Cuppini R, Minelli A. α-Tocopherol and Hippocampal Neural Plasticity in Physiological and Pathological Conditions. Int J Mol Sci 2016; 17:E2107. [PMID: 27983697 PMCID: PMC5187907 DOI: 10.3390/ijms17122107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 12/25/2022] Open
Abstract
Neuroplasticity is an "umbrella term" referring to the complex, multifaceted physiological processes that mediate the ongoing structural and functional modifications occurring, at various time- and size-scales, in the ever-changing immature and adult brain, and that represent the basis for fundamental neurocognitive behavioral functions; in addition, maladaptive neuroplasticity plays a role in the pathophysiology of neuropsychiatric dysfunctions. Experiential cues and several endogenous and exogenous factors can regulate neuroplasticity; among these, vitamin E, and in particular α-tocopherol (α-T), the isoform with highest bioactivity, exerts potent effects on many plasticity-related events in both the physiological and pathological brain. In this review, the role of vitamin E/α-T in regulating diverse aspects of neuroplasticity is analyzed and discussed, focusing on the hippocampus, a brain structure that remains highly plastic throughout the lifespan and is involved in cognitive functions. Vitamin E-mediated influences on hippocampal synaptic plasticity and related cognitive behavior, on post-natal development and adult hippocampal neurogenesis, as well as on cellular and molecular disruptions in kainate-induced temporal seizures are described. Besides underscoring the relevance of its antioxidant properties, non-antioxidant functions of vitamin E/α-T, mainly involving regulation of cell signaling molecules and their target proteins, have been highlighted to help interpret the possible mechanisms underlying the effects on neuroplasticity.
Collapse
Affiliation(s)
- Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michele Betti
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Claudia Galati
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michael Di Palma
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - David Savelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Andrea Minelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| |
Collapse
|
9
|
Jahani-Asl A, Cheng C, Zhang C, Bonni A. Pathogenesis of Börjeson-Forssman-Lehmann syndrome: Insights from PHF6 function. Neurobiol Dis 2016; 96:227-235. [PMID: 27633282 DOI: 10.1016/j.nbd.2016.09.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/07/2016] [Accepted: 09/07/2016] [Indexed: 02/07/2023] Open
Abstract
Intellectual disability encompasses a large set of neurodevelopmental disorders of cognition that are more common in males than females. Although mutations in over 100 X-linked genes associated to intellectual disability have been identified, only a few X-linked intellectual disability proteins have been intensively studied. Hence, the molecular mechanisms underlying the majority of X-linked intellectual disability disorders remain poorly understood. A substantial fraction of X-linked intellectual disability genes encode nuclear proteins, suggesting that elucidating their functions in the regulation of transcription may provide novel insights into the pathogenesis of intellectual disability. Recent studies have uncovered mechanisms by which mutations of the gene encoding plant homeodomain (PHD)-like finger protein 6 (PHF6) contribute to the pathogenesis of the X-linked intellectual disability disorder Börjeson-Forssman-Lehmann syndrome (BFLS). PHF6 plays a critical role in the migration of neurons in the mouse cerebral cortex in vivo, and patient-specific mutations disrupt the ability of PHF6 to promote neuronal migration. Interestingly, PHF6 physically associates with the PAF1 transcriptional elongation complex and thereby drives neuronal migration in the cerebral cortex. PHF6 also interacts with the NuRD chromatin remodeling complex and with the nucleolar transcriptional regulator UBF, though the biological role of these interactions remains to be characterized. In other studies, PHF6 mRNA has been identified as the target of the microRNA miR-128 in the cerebral cortex, providing new insights into regulation of PHF6 function in neuronal migration. Importantly, deregulation of PHF6 function in neuronal migration triggers the formation of white matter heterotopias that harbor neuronal hyperexcitability, which may be relevant to the pathogenesis of intellectual disability and seizures in BFLS. Collectively, these studies are beginning to provide key insights into the molecular pathogenesis of BFLS.
Collapse
Affiliation(s)
- Arezu Jahani-Asl
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA; Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H3T 1E2, Canada; Lady Davis Research Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Cheng Cheng
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA
| | - Chi Zhang
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
10
|
Schulz J, Franke K, Frick M, Schumacher S. Different roles of the small GTPases Rac1, Cdc42, and RhoG in CALEB/NGC-induced dendritic tree complexity. J Neurochem 2016; 139:26-39. [DOI: 10.1111/jnc.13735] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/24/2016] [Accepted: 07/08/2016] [Indexed: 12/01/2022]
Affiliation(s)
- Jana Schulz
- Institute of Molecular and Cellular Anatomy; Ulm University; Ulm Germany
| | - Kristin Franke
- Institute of Molecular and Cellular Anatomy; Ulm University; Ulm Germany
| | - Manfred Frick
- Institute of General Physiology; Ulm University; Ulm Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy; Ulm University; Ulm Germany
| |
Collapse
|
11
|
Zhang Y, He Q, Dong J, Jia Z, Hao F, Shan C. Effects of epigallocatechin-3-gallate on proliferation and differentiation of mouse cochlear neural stem cells: Involvement of PI3K/Akt signaling pathway. Eur J Pharm Sci 2016; 88:267-73. [PMID: 27012759 DOI: 10.1016/j.ejps.2016.03.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/11/2016] [Accepted: 03/11/2016] [Indexed: 12/29/2022]
Abstract
Since the majority of hearing impaired patients suffer from the significant loss of sensory hair cells and associated neurons, stem cell-based approaches hold great promise by replacing the damaged tissues in the ears. For instance, stem cells from the spiral ganglion could be isolated and expanded to regenerate neural structures of the inner ear. It is thus necessary to explore the potential procedures that may promote the proliferation and differentiation of such cochlear neural stem cells. In the present study, we study the effects of epigallocatechin-3-gallate (EGCG), a known antioxidant, for potential therapeutic use in NSC regeneration. At a non-toxic concentration, EGCG stimulated both proliferation and neurosphere formation in isolated mouse cochlear neural stem cell (NSC) in vitro. Specifically, the neural differentiation of NSC was promoted by EGCG treatment. The up-regulated neural function by EGCG was also supported by the increased calcium spike frequencies and enhanced neurite complexity in NSC-differentiated neurons. Finally, the induced neuron differentiation and Akt activation of cochlear NSC by EGCG were blocked by PI3 kinase inhibition. These data suggested that EGCG acts through phosphoinositide 3-kinase (PI3K)/Akt signaling in cochlea NSC to promote cell growth and neuron differentiation, which may be exploited for the treatment of hearing loss.
Collapse
Affiliation(s)
- Yubo Zhang
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China
| | - Qiang He
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China
| | - Jinhui Dong
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China
| | - Zhanwei Jia
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China
| | - Fang Hao
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China
| | - Chunguang Shan
- E.N.T. Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang 50000, China.
| |
Collapse
|
12
|
Wang Y, Yang L, Yang D. Tanshinone IIA Rescued the Impairments of Primary Hippocampal Neurons Induced by BV2 Microglial Over-Activation. Neurochem Res 2015; 40:1497-508. [PMID: 26012368 DOI: 10.1007/s11064-015-1624-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/16/2022]
Abstract
Activated microglia plays an important role in monitoring the microenvironment and prune neural process in healthy neural tissue, in order to maintain synaptic homeostasis. However, hyperactive microglia may release various cytotoxic factors and induce neuroinflammation, which cause neuronal damages leading to neurodegenerative diseases. Tanshinone IIA (TSA), an extract from traditional Chinese medicine, features potent anti-apoptotic and anti-inflammatory effects both in vitro and in vivo. But little is known on the effects of TSA on microglial-over-activation-induced neural impairments. In this study, by employing murine BV2 cell lines as well as the combinations of ELISA assay, immunostaining, western blotting analysis and RT-PCR, we found that TSA has the potential to exhibit anti-inflammatory effects. We hereby demonstrated that TSA rescued neural growth and development in the primary cultured hippocampal neurons from impairments caused by BV2 microglial over-activation insult. The results show that TSA attenuated the BV2 cell activation by lipopolysaccharide (LPS) stimulation through suppressing the NF-кB signal pathway. Also, conditioned mediums (CM) from TSA treated and activated BV2 cells protected against LPS-CM-induced neuronal death. Furthermore, TSA treatment could recover the inhibitory effects of LPS-CM on growth cone extension, neurite sprouting and outgrowth, as well as spinogenesis. Our findings support that TSA is capable of inhibiting BV2 cell over-activation thus has potential protective effects in the cultured hippocampal neurons. This study may lay a foundation for using TSA to restore cerebral injuries after severe neuroinflammation.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | | | | |
Collapse
|
13
|
|
14
|
Gu X, Meng S, Liu S, Jia C, Fang Y, Li S, Fu C, Song Q, Lin L, Wang X. miR-124 represses ROCK1 expression to promote neurite elongation through activation of the PI3K/Akt signal pathway. J Mol Neurosci 2013; 52:156-65. [PMID: 24338057 DOI: 10.1007/s12031-013-0190-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/18/2013] [Indexed: 12/22/2022]
Abstract
Recent studies have demonstrated an important role for miR-124, the most abundant and well-conserved brain-specific microRNA(miRNA), in promoting neurite outgrowth and elongation during neuronal differentiation. This miRNA's target genes and the mechanisms that execute this role remain unclear. In this study, we identified ROCK1, a small GTPase Rho kinase, as a direct target of miR-124 for regulating neurite elongation. miR-124 significantly inhibited ROCK1 expression in M17 cells. Inhibiting ROCK1 promoted neurite elongation, and the overexpression of ROCK1 strongly repressed the neurite elongation-enhancing effect of miR-124 in M17 cells. We determined that Akt functions as a novel ROCK1 downstream effector in regulating neurite outgrowth and elongation.
Collapse
Affiliation(s)
- Xi Gu
- Department of Neurobiology, Southern Medical University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Ramamurthy S, Chang E, Cao Y, Zhu J, Ronnett GV. AMPK activation regulates neuronal structure in developing hippocampal neurons. Neuroscience 2013; 259:13-24. [PMID: 24295634 DOI: 10.1016/j.neuroscience.2013.11.048] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/21/2013] [Accepted: 11/23/2013] [Indexed: 12/25/2022]
Abstract
AMP-activated protein kinase (AMPK) is a serine/threonine kinase that functions as a cellular and whole organism energy sensor to regulate ATP-consuming (anabolic) and ATP-generating (catabolic) pathways. The heterotrimeric AMPK complex consists of a catalytic α-subunit, regulatory β-subunit, and an AMP/ATP-binding γ-subunit. Several alternate isoforms exist for each subunit (α1, α2, β1, β2, γ1, γ2 and γ3). However, little is known of the expression pattern or function of the individual catalytic complexes in regulating neuronal structure. In this study, we examined the role of AMPK subunits in differentiating hippocampal neurons. We found that during development, the expression of AMPK subunits increase and that activation of AMPK by energetic stress inhibits neuronal development at multiple stages, not only during axon outgrowth, but also during dendrite growth and arborization. The presence of a single functional AMPK catalytic complex was sufficient to mediate these inhibitory effects of energetic stress. Activation of AMPK mediates these effects by suppressing both the mTOR and Akt signaling pathways. These findings demonstrate that the energy-sensing AMPK pathway regulates neuronal structure in distinct regions of developing neurons at multiple stages of development.
Collapse
Affiliation(s)
- S Ramamurthy
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA
| | - E Chang
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA
| | - Y Cao
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA
| | - J Zhu
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA
| | - G V Ronnett
- Department of Neuroscience, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Department of Biological Chemistry, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 855 N Wolfe Street, Baltimore, MD 21205, USA; Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
16
|
Cottet S, Jüttner R, Voirol N, Chambon P, Rathjen FG, Schorderet DF, Escher P. Retinal pigment epithelium protein of 65 kDA gene-linked retinal degeneration is not modulated by chicken acidic leucine-rich epidermal growth factor-like domain containing brain protein/Neuroglycan C/ chondroitin sulfate proteoglycan 5. Mol Vis 2013; 19:2312-20. [PMID: 24265546 PMCID: PMC3834887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/14/2013] [Indexed: 10/26/2022] Open
Abstract
PURPOSE To analyze in vivo the function of chicken acidic leucine-rich epidermal growth factor-like domain containing brain protein/Neuroglycan C (gene symbol: Cspg5) during retinal degeneration in the Rpe65⁻/⁻ mouse model of Leber congenital amaurosis. METHODS We resorted to mice with targeted deletions in the Cspg5 and retinal pigment epithelium protein of 65 kDa (Rpe65) genes (Cspg5⁻/⁻/Rpe65⁻/⁻). Cone degeneration was assessed with cone-specific peanut agglutinin staining. Transcriptional expression of rhodopsin (Rho), S-opsin (Opn1sw), M-opsin (Opn1mw), rod transducin α subunit (Gnat1), and cone transducin α subunit (Gnat2) genes was assessed with quantitative PCR from 2 weeks to 12 months. The retinal pigment epithelium (RPE) was analyzed at P14 with immunodetection of the retinol-binding protein membrane receptor Stra6. RESULTS No differences in the progression of retinal degeneration were observed between the Rpe65⁻/⁻ and Cspg5⁻/⁻/Rpe65⁻/⁻ mice. No retinal phenotype was detected in the late postnatal and adult Cspg5⁻/⁻ mice, when compared to the wild-type mice. CONCLUSIONS Despite the previously reported upregulation of Cspg5 during retinal degeneration in Rpe65⁻/⁻ mice, no protective effect or any involvement of Cspg5 in disease progression was identified.
Collapse
Affiliation(s)
- Sandra Cottet
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| | | | | | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Collège de France, Strasbourg, France
| | | | - Daniel F. Schorderet
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland,EPFL-Ecole Polytechnique Fédérale, Lausanne, Switzerland
| | - Pascal Escher
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Schmeisser MJ, Kühl SJ, Schoen M, Beth NH, Weis TM, Grabrucker AM, Kühl M, Boeckers TM. The Nedd4-binding protein 3 (N4BP3) is crucial for axonal and dendritic branching in developing neurons. Neural Dev 2013; 8:18. [PMID: 24044555 PMCID: PMC3849298 DOI: 10.1186/1749-8104-8-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 08/28/2013] [Indexed: 11/22/2022] Open
Abstract
Background Circuit formation in the nervous system essentially relies on the proper development of neurons and their processes. In this context, the ubiquitin ligase Nedd4 is a crucial modulator of axonal and dendritic branching. Results Herein we characterize the Nedd4-binding protein 3 (N4BP3), a Fezzin family member, during nerve cell development. In developing rat primary hippocampal neurons, endogenous N4BP3 localizes to neuronal processes, including axons and dendrites. Transient in vitro knockdown of N4BP3 in hippocampal cultures during neuritogenesis results in impaired branching of axons and dendrites. In line with these findings, in vivo knockdown of n4bp3 in Xenopus laevis embryos results in severe alteration of cranial nerve branching. Conclusions We introduce N4BP3 as a novel molecular element for the correct branching of neurites in developing neurons and propose a central role for an N4BP3-Nedd4 complex in neurite branching and circuit formation.
Collapse
|
18
|
Jüttner R, Montag D, Craveiro RB, Babich A, Vetter P, Rathjen FG. Impaired presynaptic function and elimination of synapses at premature stages during postnatal development of the cerebellum in the absence of CALEB (CSPG5/neuroglycan C). Eur J Neurosci 2013; 38:3270-80. [PMID: 23889129 DOI: 10.1111/ejn.12313] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 06/17/2013] [Indexed: 12/25/2022]
Abstract
Chicken acidic leucine-rich EGF-like domain-containing brain protein (CALEB), also known as chondroitin sulfate proteoglycan (CSPG)5 or neuroglycan C, is a neural chondroitin sulfate-containing and epidermal growth factor (EGF)-domain-containing transmembrane protein that is implicated in synaptic maturation. Here, we studied the role of CALEB within the developing cerebellum. Adult CALEB-deficient mice displayed impaired motor coordination in Rota-Rod experiments. Analysis of the neuronal connectivity of Purkinje cells by patch-clamp recordings demonstrated impairments of presynaptic maturation of inhibitory synapses. GABAergic synapses on Purkinje cells revealed decreased evoked amplitudes, altered paired-pulse facilitation and reduced depression after repetitive stimulation at early postnatal but not at mature stages. Furthermore, the elimination of supernumerary climbing fiber synapses on Purkinje cells was found to occur at earlier developmental stages in the absence of CALEB. For example, at postnatal day 8 in wild-type mice, 54% of Purkinje cells had three or more climbing fiber synapses in contrast to mutants where this number was decreased to less than 25%. The basic properties of the climbing fiber Purkinje cell synapse remained unaffected. Using Sholl analysis of dye-injected Purkinje cells we revealed that the branching pattern of the dendritic tree of Purkinje cells was not impaired in CALEB-deficient mice. The alterations observed by patch-clamp recordings correlated with a specific pattern and timing of expression of CALEB in Purkinje cells, i.e. it is dynamically regulated during development from a high chondroitin sulfate-containing form to a non-chondroitin sulfate-containing form. Thus, our results demonstrated an involvement of CALEB in the presynaptic differentiation of cerebellar GABAergic synapses and revealed a new role for CALEB in synapse elimination in Purkinje cells.
Collapse
Affiliation(s)
- René Jüttner
- Max-Delbrück-Centrum, Robert-Rössle-Straße 10, 13092, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Tapias V, Greenamyre JT, Watkins SC. Automated imaging system for fast quantitation of neurons, cell morphology and neurite morphometry in vivo and in vitro. Neurobiol Dis 2013; 54:158-68. [PMID: 23220621 PMCID: PMC3604080 DOI: 10.1016/j.nbd.2012.11.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 11/20/2012] [Accepted: 11/28/2012] [Indexed: 12/21/2022] Open
Abstract
Quantitation of neurons using stereologic approaches reduces bias and systematic error, but is time-consuming and labor-intensive. Accurate methods for quantifying neurons in vitro are lacking; conventional methodologies are limited in reliability and application. The morphological properties of the soma and neurites are a key aspect of neuronal phenotype and function, but the assays commonly used in such evaluations are beset with several methodological drawbacks. Herein we describe automated techniques to quantify the number and morphology of neurons (or any cell type, e.g., astrocytes) and their processes with high speed and accuracy. Neuronal quantification from brain tissue using a motorized stage system yielded results that were statistically comparable to those generated by stereology. The approach was then adapted for in vitro neuron and neurite outgrowth quantification. To determine the utility of our methods, rotenone was used as a neurotoxicant leading to morphological changes in neurons and cell death, astrocytic activation, and loss of neurites. Importantly, our technique counted about 8 times as many neurons in less than 5-10% of the time taken by manual stereological analysis.
Collapse
Affiliation(s)
- Victor Tapias
- Department of Neurology, University of Pittsburgh, USA.
| | | | | |
Collapse
|
20
|
Abstract
RhoG is a member of the Rho family of small GTPases sharing highest sequence similarity with Rac and Cdc42. Mig-2 and Mtl represent the functional equivalents of RhoG in Caenorhabditis elegans and Drosophila, respectively. RhoG has attracted great interest because it plays a central role in the regulation of cytoskeletal reorganization in various physiological and pathophysiological situations. For example, it is fundamental to phagocytotic processes, is able to regulate gene expression, cell survival and proliferation, and is involved in cell migration and in the invasion of pathogenic bacteria. The activation of Rac1 via an ELMO/Dock180 module has been elaborated to be important for RhoG signaling. Although a stimulatory role for neurite outgrowth in the pheochromocytoma PC12 cell line has been assigned to RhoG, the exact function of this GTPase for the development of the processes of primary neurons remains to be clarified. In this view, we discuss the impact of RhoG on axonal and dendritic differentiation, its role as a conductor of Rac1 and Cdc42 activity and the functional regulation of RhoG expression by the microRNA miR-124.
Collapse
Affiliation(s)
- Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany.
| | | |
Collapse
|
21
|
Urbanska M, Gozdz A, Swiech LJ, Jaworski J. Mammalian target of rapamycin complex 1 (mTORC1) and 2 (mTORC2) control the dendritic arbor morphology of hippocampal neurons. J Biol Chem 2012; 287:30240-56. [PMID: 22810227 DOI: 10.1074/jbc.m112.374405] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dendrites are the main site of information input into neurons. Their development is a multistep process controlled by mammalian target of rapamycin (mTOR) among other proteins. mTOR is a serine/threonine protein kinase that forms two functionally distinct complexes in mammalian cells: mTORC1 and mTORC2. However, the one that contributes to mammalian neuron development remains unknown. This work used short hairpin RNA against Raptor and Rictor, unique components of mTORC1 and mTORC2, respectively, to dissect mTORC involvement in this process. We provide evidence that both mTOR complexes are crucial for the proper dendritic arbor morphology of hippocampal neurons. These two complexes are required for dendritic development both under basal conditions and upon the induction of mTOR-dependent dendritic growth. We also identified Akt as a downstream effector of mTORC2 needed for proper dendritic arbor morphology, the action of which required mTORC1 and p70S6K1.
Collapse
Affiliation(s)
- Malgorzata Urbanska
- International Institute of Molecular and Cell Biology, 02-109, Warsaw, Poland
| | | | | | | |
Collapse
|
22
|
miR-124-regulated RhoG reduces neuronal process complexity via ELMO/Dock180/Rac1 and Cdc42 signalling. EMBO J 2012; 31:2908-21. [PMID: 22588079 DOI: 10.1038/emboj.2012.130] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/05/2012] [Indexed: 12/19/2022] Open
Abstract
The small GTPase RhoG plays a central role in actin remodelling during diverse biological processes such as neurite outgrowth, cell migration, phagocytosis of apoptotic cells, and the invasion of pathogenic bacteria. Although it is known that RhoG stimulates neurite outgrowth in the rat pheochromocytoma PC12 cell line, neither the physiological function nor the regulation of this GTPase in neuronal differentiation is clear. Here, we identify RhoG as an inhibitor of neuronal process complexity, which is regulated by the microRNA miR-124. We find that RhoG inhibits dendritic branching in hippocampal neurons in vitro and in vivo. RhoG also inhibits axonal branching, acting via an ELMO/Dock180/Rac1 signalling pathway. However, RhoG inhibits dendritic branching dependent on the small GTPase Cdc42. Finally, we show that the expression of RhoG in neurons is suppressed by the CNS-specific microRNA miR-124 and connect the regulation of RhoG expression by miR-124 to the stimulation of neuronal process complexity. Thus, RhoG emerges as a cellular conductor of Rac1 and Cdc42 activity, in turn regulated by miR-124 to control axonal and dendritic branching.
Collapse
|
23
|
Regulation of axonal HCN1 trafficking in perforant path involves expression of specific TRIP8b isoforms. PLoS One 2012; 7:e32181. [PMID: 22363812 PMCID: PMC3283722 DOI: 10.1371/journal.pone.0032181] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 01/23/2012] [Indexed: 12/22/2022] Open
Abstract
The functions of HCN channels in neurons depend critically on their subcellular localization, requiring fine-tuned machinery that regulates subcellular channel trafficking. Here we provide evidence that regulatory mechanisms governing axonal HCN channel trafficking involve association of the channels with specific isoforms of the auxiliary subunit TRIP8b. In the medial perforant path, which normally contains HCN1 channels in axon terminals in immature but not in adult rodents, we found axonal HCN1 significantly increased in adult mice lacking TRIP8b (TRIP8b−/−). Interestingly, adult mice harboring a mutation that results in expression of only the two most abundant TRIP8b isoforms (TRIP8b[1b/2]−/−) exhibited an HCN1 expression pattern similar to wildtype mice, suggesting that presence of one or both of these isoforms (TRIP8b(1a), TRIP8b(1a-4)) prevents HCN1 from being transported to medial perforant path axons in adult mice. Concordantly, expression analyses demonstrated a strong increase of expression of both TRIP8b isoforms in rat entorhinal cortex with age. However, when overexpressed in cultured entorhinal neurons of rats, TRIP8b(1a), but not TRIP8b(1a-4), altered substantially the subcellular distribution of HCN1 by promoting somatodendritic and reducing axonal expression of the channels. Taken together, we conclude that TRIP8b isoforms are important regulators of HCN1 trafficking in entorhinal neurons and that the alternatively-spliced isoform TRIP8b(1a) could be responsible for the age-dependent redistribution of HCN channels out of perforant path axon terminals.
Collapse
|
24
|
Abstract
Dendritic arbors are compartments of neurons dedicated to receiving synaptic inputs. Their shape is an outcome of both the intrinsic genetic program and environmental signals. The microtubules and actin cytoskeleton are both crucial for proper dendritic morphology, but how they interact is unclear. The present study demonstrates that microtubule plus-end tracking protein CLIP-170 and actin-binding protein IQGAP1 regulate dendrite morphology of rat neurons by coordinating the interaction between microtubules and the actin cytoskeleton. Moreover, we show that mTOR kinase interacts with CLIP-170 and is needed for efficient formation of a protein complex containing CLIP-170 and IQGAP1. Dynamic microtubules, CLIP-170, and IQGAP1 are required for proper dendritic arbor morphology and PI3K-mTOR-induced increase in dendritic arbor complexity. Moreover, CLIP-170 and IQGAP1 knockdown modulates dendritic arbor growth via regulation of the actin cytoskeleton. We postulate that mTOR controls dendritic arbor morphology by enhancing cross talk between dynamic microtubules and actin through CLIP-170 and IQGAP1.
Collapse
|
25
|
Betti M, Ambrogini P, Minelli A, Floridi A, Lattanzi D, Ciuffoli S, Bucherelli C, Prospero E, Frontini A, Santarelli L, Baldi E, Benetti F, Galli F, Cuppini R. Maternal dietary loads of α-tocopherol depress protein kinase C signaling and synaptic plasticity in rat postnatal developing hippocampus and promote permanent deficits in adult offspring. J Nutr Biochem 2011; 22:60-70. [DOI: 10.1016/j.jnutbio.2009.11.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 11/17/2009] [Accepted: 11/30/2009] [Indexed: 01/12/2023]
|
26
|
Nakanishi K, Tokita Y, Aono S, Ida M, Matsui F, Higashi Y, Oohira A. Neuroglycan C, a brain-specific chondroitin sulfate proteoglycan, interacts with pleiotrophin, a heparin-binding growth factor. Neurochem Res 2010; 35:1131-7. [PMID: 20369290 DOI: 10.1007/s11064-010-0164-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2010] [Indexed: 12/24/2022]
Abstract
Neuroglycan C (NGC) is a transmembrane-type chondroitin sulfate proteoglycan that promotes neurite outgrowth. To identify the ligand of NGC, we applied a detergent-solubilized membrane fraction of fetal rat brains to an NGC-immobilized affinity column. Several proteins were eluted from the column including an 18 kDa-band protein recognized by an anti-pleiotrophin antibody. The binding of pleiotrophin (PTN) to NGC was confirmed by a quartz crystal microbalance method and had a Kd of 8.7 nM. PTN bound to the acidic amino acid cluster of the NGC extracellular domain. In addition, PTN bound to both chondroitin sulfate-bearing NGC and chondroitinase-treated NGC prepared from the neonatal rat brain. These results suggest that NGC interacts with PTN.
Collapse
Affiliation(s)
- Keiko Nakanishi
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi, Japan.
| | | | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Abstract
Our understanding of the mechanisms involved in the formation of the complex arrangement of neurons and their interconnections within the brain has made significant progress in recent years. Current research has uncovered a network of intracellular signaling events that provide precise coordination of a diverse array of cellular responses, including trafficking events, cytoskeletal remodeling, gene transcription, and protein ubiquitination and translation. This chapter considers the specific cellular responses controlled by the phosphatidylinositol 3-kinase (PI3K) signaling pathway, which is instructive with regard to a number of important steps involved in the development of the brain. These range from the mediation of extrinsic signals - such as growth factors, axon guidance cues, and extracellular matrix components - to intrinsic effectors, such as downstream signaling components that act, for example, at the translation level. PI3K signaling is, consequently, at the heart of controlling neuronal migration and neuronal morphogenesis, as well as dendrite and synapse development. Many neurobehavioral disorders arise as a consequence of subtle developmental abnormalities. Unsurprisingly, therefore, aberrant PI3K signaling has been indicated by many studies to be a contributing factor to the pathophysiology of disorders such as schizophrenia and autism. In this chapter, we will focus on the specific, yet divergent, cellular processes that are achieved through PI3K signaling in neurons and are key to brain development.
Collapse
Affiliation(s)
- Kathryn Waite
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, London, UK
| | | |
Collapse
|
29
|
Trimbuch T, Beed P, Vogt J, Schuchmann S, Maier N, Kintscher M, Breustedt J, Schuelke M, Streu N, Kieselmann O, Brunk I, Laube G, Strauss U, Battefeld A, Wende H, Birchmeier C, Wiese S, Sendtner M, Kawabe H, Kishimoto-Suga M, Brose N, Baumgart J, Geist B, Aoki J, Savaskan NE, Bräuer AU, Chun J, Ninnemann O, Schmitz D, Nitsch R. Synaptic PRG-1 modulates excitatory transmission via lipid phosphate-mediated signaling. Cell 2009; 138:1222-35. [PMID: 19766573 DOI: 10.1016/j.cell.2009.06.050] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 04/30/2009] [Accepted: 06/19/2009] [Indexed: 11/27/2022]
Abstract
Plasticity related gene-1 (PRG-1) is a brain-specific membrane protein related to lipid phosphate phosphatases, which acts in the hippocampus specifically at the excitatory synapse terminating on glutamatergic neurons. Deletion of prg-1 in mice leads to epileptic seizures and augmentation of EPSCs, but not IPSCs. In utero electroporation of PRG-1 into deficient animals revealed that PRG-1 modulates excitation at the synaptic junction. Mutation of the extracellular domain of PRG-1 crucial for its interaction with lysophosphatidic acid (LPA) abolished the ability to prevent hyperexcitability. As LPA application in vitro induced hyperexcitability in wild-type but not in LPA(2) receptor-deficient animals, and uptake of phospholipids is reduced in PRG-1-deficient neurons, we assessed PRG-1/LPA(2) receptor-deficient animals, and found that the pathophysiology observed in the PRG-1-deficient mice was fully reverted. Thus, we propose PRG-1 as an important player in the modulatory control of hippocampal excitability dependent on presynaptic LPA(2) receptor signaling.
Collapse
Affiliation(s)
- Thorsten Trimbuch
- Institute of Cell Biology and Neurobiology and NeuroCure, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Vodrazka P, Korostylev A, Hirschberg A, Swiercz JM, Worzfeld T, Deng S, Fazzari P, Tamagnone L, Offermanns S, Kuner R. The semaphorin 4D-plexin-B signalling complex regulates dendritic and axonal complexity in developing neurons via diverse pathways. Eur J Neurosci 2009; 30:1193-208. [PMID: 19788569 DOI: 10.1111/j.1460-9568.2009.06934.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Semaphorins and their receptors, plexins, have emerged as key regulators of various aspects of neuronal development. In contrast to the Plexin-A family, the cellular functions of Plexin-B family proteins in developing neurons are only poorly understood. An activation of Plexin-B1 via its ligand, semaphorin 4D (Sema4D), produces an acute collapse of axonal growth cones in hippocampal and retinal neurons over the early stages of neurite outgrowth. However, the functional role of Sema4D-Plexin-B interactions over subsequent stages of neurite development, differentiation and maturation has not been characterized. Here we addressed this question using morphogenetic assays and time-lapse imaging on developing rat hippocampal neurons as a model system. Interestingly, Sema4D treatment over several hours was observed to promote branching and complexity in hippocampal neurons via the activation of Plexin-B1. The activation of receptor tyrosine kinases and the Rho kinase following Sema4D treatment was found to control dendritic and axonal morphogenesis by differentially regulating branching and extension. Phosphoinositide-3-kinase, but not extracellular signal-regulated kinase 1/2, was observed to be important for the stimulatory effects of Sema4D on dendritic branching. Furthermore, we observed that the mammalian target of rapamycin is activated downstream of Plexin-B1 and contributes to Sema4D-induced effects on dendritic branching. In contrast, glycogen synthase kinase-3 beta, another effector of phosphoinositide-3-kinase signalling, was not involved. Thus, our results show that Sema4D-Plexin-B interactions modulate dendritic and axonal arborizations of developing neurons by co-ordinated and concerted activation of diverse signalling pathways.
Collapse
Affiliation(s)
- Peter Vodrazka
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Smorodchenko A, Rupprecht A, Sarilova I, Ninnemann O, Bräuer AU, Franke K, Schumacher S, Techritz S, Nitsch R, Schuelke M, Pohl EE. Comparative analysis of uncoupling protein 4 distribution in various tissues under physiological conditions and during development. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2309-19. [PMID: 19646951 DOI: 10.1016/j.bbamem.2009.07.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 07/21/2009] [Accepted: 07/23/2009] [Indexed: 11/17/2022]
Abstract
UCP4 is a member of the mitochondrial uncoupling protein subfamily and one of the three UCPs (UCP2, UCP4, UCP5), associated with the nervous system. Its putative functions include thermogenesis, attenuation of reactive oxidative species (ROS), regulation of mitochondrial calcium concentration and involvement in cell differentiation and apoptosis. Here we investigate UCP4's subcellular, cellular and tissue distribution, using an antibody designed specially for this study, and discuss the findings in terms of the protein's possible functions. Western blot and immunohistochemistry data confirmed that UCP4 is expressed predominantly in the central nervous system (CNS), as previously shown at mRNA level. No protein was found in heart, spleen, stomach, intestine, lung, thymus, muscles, adrenal gland, testis and liver. The reports revealing UCP4 mRNA in kidney and white adipose tissue were not confirmed at protein level. The amount of UCP4 varies in the mitochondria of different brain regions, with the highest protein content found in cortex. We show that UCP4 is present in fetal murine brain tissue as early as embryonic days 12-14 (E12-E14), which coincides with the beginning of neuronal differentiation. The UCP4 content in mitochondria decreases as the age of mice increases. UCP4 preferential expression in neurons and its developmental expression pattern under physiological conditions may indicate a specific protein function, e.g. in neuronal cell differentiation.
Collapse
Affiliation(s)
- Alina Smorodchenko
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Grabrucker AM, Vaida B, Bockmann J, Boeckers TM. Efficient targeting of proteins to post-synaptic densities of excitatory synapses using a novel pSDTarget vector system. J Neurosci Methods 2009; 181:227-34. [DOI: 10.1016/j.jneumeth.2009.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 05/14/2009] [Accepted: 05/15/2009] [Indexed: 11/16/2022]
|
33
|
Amendola J, Durand J. Morphological differences between wild-type and transgenic superoxide dismutase 1 lumbar motoneurons in postnatal mice. J Comp Neurol 2008; 511:329-41. [PMID: 18803237 DOI: 10.1002/cne.21818] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Quantitative analysis of the dendritic arborizations of wild-type (WT) and superoxide dismutase 1 (SOD1) postnatal mouse motoneurons was performed following intracellular staining and 3D reconstructions with Neurolucida system. The population of lumbar motoneurons was targeted in the caudal part of the L5 segment, and all labeled motoneurons were located within the same ventrolateral pool. Despite the similar size of the soma and the mean diameter of primary dendrites, the dendritic arborizations of the WT and SOD1 motoneurons showed significant differences in terms of their morphometric parameters. The metric and topological parameters of dendrites show that the total dendritic length and surface area and total number of segments, branching nodes, and tips per motoneuron were significantly higher in SOD1 motoneurons. Our main finding concerns a proliferation of dendritic branches starting at about 100 microm from the soma in the SOD1 motoneurons. However, the longest and mean dendritic paths from soma to terminations were similar, giving a comparable envelope of the dendritic fields. Indeed, the SOD1 motoneurons were larger as a result of abnormal branching. The results suggest that a defect in pruning mechanisms occurs during this developmental period. The abnormal growth of the dendritic arborizations and the reduced excitability of postnatal SOD1 motoneurons could be a neuroprotective response and would represent an early compensatory mechanism against the activity-induced toxicity.
Collapse
Affiliation(s)
- Julien Amendola
- UMR 6196, Centre National de la Recherche Scientifique, Laboratoire de Plasticité et Physiopathologie de la Motricité, Université de la Méditerranée, 13402 Marseille Cedex 20, France
| | | |
Collapse
|
34
|
Escher P, Cottet S, Aono S, Oohira A, Schorderet DF. Differential neuroglycan C expression during retinal degeneration in Rpe65-/- mice. Mol Vis 2008; 14:2126-35. [PMID: 19050768 PMCID: PMC2592529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 11/19/2008] [Indexed: 11/12/2022] Open
Abstract
PURPOSE An increased mRNA expression of the genes coding for the extracellular matrix proteins neuroglycan C (NGC), interphotoreceptor matrix proteoglycan 2 (IMPG2), and CD44 antigen (CD44) has been observed during retinal degeneration in mice with a targeted disruption of the Rpe65 gene (Rpe65-/- mouse). To validate these data, we analyzed this differential expression in more detail by characterizing retinal NGC mRNA isoform and protein expression during disease progression. METHODS Retinas from C57/Bl6 wild-type and Rpe65-/- mice, ranging 2 to 18 months of age, were used. NGC, IMPG2, and CD44 mRNA expression was assessed by oligonucleotide microarray, quantitative PCR, and in situ hybridization. Retinal NGC protein expression was analyzed by western blot and immunohistochemistry. RESULTS As measured by quantitative PCR, mRNA expression of NGC and CD44 was induced by about 2 fold to 3 fold at all time points in Rpe65-/- retinas, whereas initially 4 fold elevated IMPG2 mRNA levels progressively declined. NGC and IMPG2 mRNAs were expressed in the ganglion cell layer, the inner nuclear layer, and at the outer limiting membrane. NGC mRNA was also detected in retinal pigment epithelium cells (RPE), where its mRNA expression was not induced during retinal degeneration. NGC-I was the major isoform detected in the retina and the RPE, whereas NGC-III was barely detected and NGC-II could not be assessed. NGC protein expression was at its highest levels on the apical membrane of the RPE. NGC protein levels were induced in retinas from 2- and 4-month-old Rpe65-/- mice, and an increased amount of the activity-cleaved NGC ectodomain containing an epidermal growth factor (EGF)-like domain was detected. CONCLUSIONS During retinal degeneration in Rpe65-/- mice, NGC expression is induced in the neural retina, but not in the RPE, where NGC is expressed at highest levels.
Collapse
Affiliation(s)
- Pascal Escher
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| | - Sandra Cottet
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| | - Saichiko Aono
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan
| | - Atsuhiko Oohira
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan
| | - Daniel F. Schorderet
- Institute for Research in Ophthalmology, Sion, Switzerland,Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland,EPFL-Ecole Polytechnique Fédérale, Lausanne, Switzerland
| |
Collapse
|
35
|
Volonté C, D'Ambrosi N, Amadio S. Protein cooperation: from neurons to networks. Prog Neurobiol 2008; 86:61-71. [PMID: 18722498 DOI: 10.1016/j.pneurobio.2008.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 07/28/2008] [Indexed: 12/30/2022]
Abstract
A constant pattern through the development of cellular life is that not only cells but also subcellular components such as proteins, either being enzymes, receptors, signaling or structural proteins, strictly cooperate. Discerning how protein cooperation originated and propagates over evolutionary time, how proteins work together to a shared outcome far beyond mere interaction, thus represents a theoretical and experimental challenge for evolutionary, molecular, and computational biology, and a timely fruition also for biotechnology. In this review, we describe some basic principles sustaining not only cellular but especially protein cooperative behavior, with particular emphasis on neurobiological systems. We illustrate experimental results and numerical models substantiating that bench research, as well as computer analysis, indeed concurs in recognizing the natural propensity of proteins to cooperate. At the cellular level, we exemplify network connectivity in the thalamus, hippocampus and basal ganglia. At the protein level, we depict numerical models about the receptosome, the protein machinery connecting neurotransmitters or growth factors to specific, unique downstream effector proteins. We primarily focus on the purinergic P2/P1 receptor systems for extracellular purine and pyrimidine nucleotides/nucleosides. By spanning concepts such as single-molecule biology to membrane computing, we seek to stimulate a scientific debate on the implications of protein cooperation in neurobiological systems.
Collapse
Affiliation(s)
- Cinzia Volonté
- Santa Lucia Foundation/CNR, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | | | | |
Collapse
|
36
|
Brandt N, Franke K, Johannes S, Buck F, Harder S, Hassel B, Nitsch R, Schumacher S. B56beta, a regulatory subunit of protein phosphatase 2A, interacts with CALEB/NGC and inhibits CALEB/NGC-mediated dendritic branching. FASEB J 2008; 22:2521-33. [PMID: 18385213 DOI: 10.1096/fj.07-096115] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The development of dendritic arbors is critical in neuronal circuit formation, as dendrites are the primary sites of synaptic input. Morphologically specialized dendritic protrusions called spines represent the main postsynaptic compartment for excitatory neurotransmission. Recently, we demonstrated that chicken acidic leucine-rich epidermal growth factor (EGF) -like domain-containing brain protein/neuroglycan C (CALEB/NGC), a neural member of the EGF family, mediates dendritic tree and spine complexity but that the signaling pathways in the respective processes differ. For a more detailed characterization of these signal transduction pathways, we performed a yeast two-hybrid screen to identify proteins that interact with CALEB/NGC. Our results show that B56beta, a regulatory subunit of protein phosphatase 2A, interacts with CALEB/NGC and inhibits CALEB/NGC-mediated dendritic branching but not spine formation. Binding of B56beta to CALEB/NGC was confirmed by several biochemical and immunocytochemical assays. Using affinity chromatography and mass spectrometry, we demonstrate that the whole protein phosphatase 2A trimer, including structural and catalytic subunits, binds to CALEB/NGC via B56beta. We show that CALEB/NGC induces the phosphorylation of Akt in dendrites. Previously described to interfere with Akt signaling, B56beta inhibits Akt phosphorylation and Akt-dependent dendritic branching but not Akt-independent spine formation induced by CALEB/NGC. Our results contribute to a better understanding of signaling specificity leading to neuronal process differentiation in sequential developmental events.
Collapse
Affiliation(s)
- Nicola Brandt
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Sirt1 contributes critically to the redox-dependent fate of neural progenitors. Nat Cell Biol 2008; 10:385-94. [PMID: 18344989 DOI: 10.1038/ncb1700] [Citation(s) in RCA: 362] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Accepted: 02/14/2008] [Indexed: 01/28/2023]
Abstract
Repair processes that are activated in response to neuronal injury, be it inflammatory, ischaemic, metabolic, traumatic or other cause, are characterized by a failure to replenish neurons and by astrogliosis. The underlying molecular pathways, however, are poorly understood. Here, we show that subtle alterations of the redox state, found in different brain pathologies, regulate the fate of mouse neural progenitor cells (NPCs) through the histone deacetylase (HDAC) Sirt1. Mild oxidation or direct activation of Sirt1 suppressed proliferation of NPCs and directed their differentiation towards the astroglial lineage at the expense of the neuronal lineage, whereas reducing conditions had the opposite effect. Under oxidative conditions in vitro and in vivo, Sirt1 was upregulated in NPCs, bound to the transcription factor Hes1 and subsequently inhibited pro-neuronal Mash1. In utero shRNA-mediated knockdown of Sirt1 in NPCs prevented oxidation-mediated suppression of neurogenesis and caused upregulation of Mash1 in vivo. Our results provide evidence for an as yet unknown metabolic master switch that determines the fate of neural progenitors.
Collapse
|
38
|
Swiech L, Perycz M, Malik A, Jaworski J. Role of mTOR in physiology and pathology of the nervous system. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1784:116-32. [PMID: 17913600 DOI: 10.1016/j.bbapap.2007.08.015] [Citation(s) in RCA: 269] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Revised: 08/09/2007] [Accepted: 08/10/2007] [Indexed: 01/04/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a serine-threonine protein kinase that regulates several intracellular processes in response to extracellular signals, nutrient availability, energy status of the cell and stress. mTOR regulates survival, differentiation and development of neurons. Axon growth and navigation, dendritic arborization, as well as synaptogenesis, depend on proper mTOR activity. In adult brain mTOR is crucial for synaptic plasticity, learning and memory formation, and brain control of food uptake. Recent studies reveal that mTOR activity is modified in various pathologic states of the nervous system, including brain tumors, tuberous sclerosis, cortical displasia and neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases. This review presents current knowledge about the role of mTOR in the physiology and pathology of the nervous system, with special focus on molecular targets acting downstream of mTOR that potentially contribute to neuronal development, plasticity and neuropathology.
Collapse
Affiliation(s)
- Lukasz Swiech
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | | |
Collapse
|