1
|
Rao SS, Lago L, Volitakis I, Shukla JJ, McColl G, Finkelstein DI, Adlard PA. Deferiprone Treatment in Aged Transgenic Tau Mice Improves Y-Maze Performance and Alters Tau Pathology. Neurotherapeutics 2021; 18:1081-1094. [PMID: 33410108 PMCID: PMC8423882 DOI: 10.1007/s13311-020-00972-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 11/25/2022] Open
Abstract
The accumulation of neurofibrillary tangles (NFTs), which is composed of abnormally hyperphosphorylated tau aggregates, is the classic neuropathology associated with cognitive dysfunction in tauopathies such as Alzheimer's disease (AD). However, there is an emerging theory suggesting that dysregulation in cerebral iron may contribute to NFT formation. Iron is speculated to bind to tau and induce conformational changes of the protein, potentially leading to subsequent aggregation and cognitive decline. Deferiprone (DFP) is a clinically available iron chelator, which has demonstrated potential therapeutic advantages of chelating iron in neurodegenerative disorders, and is currently in clinical trials for AD. However, its effect on tau pathology remains unclear. Here, we report the effects of short-term DFP treatment (4 weeks, 100 mg/kg/daily, via oral gavage) in a mixed-gender cohort of the rTg(tauP301L)4510 mouse model of tauopathy. Our results revealed that DFP improved Y-maze and open field performance, accompanied by a 28% decrease in brain iron levels, measured by inductively coupled plasma mass spectrometry (ICP-MS) and reduced AT8-labeled p-tau within the hippocampus in transgenic tau mice. This data supports the notion that iron may play a neurotoxic role in tauopathies and may be a potential therapeutic target for this class of disorders that can be modulated by the clinically available metal chelator DFP.
Collapse
Affiliation(s)
- Shalini S Rao
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Larissa Lago
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Irene Volitakis
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Jay J Shukla
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Gawain McColl
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - David I Finkelstein
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Paul A Adlard
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
2
|
Owens LV, Benedetto A, Dawson N, Gaffney CJ, Parkin ET. Gene therapy-mediated enhancement of protective protein expression for the treatment of Alzheimer's disease. Brain Res 2021; 1753:147264. [PMID: 33422539 DOI: 10.1016/j.brainres.2020.147264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/22/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the leading form of dementia but lacks curative treatments. Current understanding of AD aetiology attributes the development of the disease to the misfolding of two proteins; amyloid-β (Aβ) and hyperphosphorylated tau, with their pathological accumulation leading to concomitant oxidative stress, neuroinflammation, and neuronal death. These processes are regulated at multiple levels to maintain homeostasis and avert disease. However, many of the relevant regulatory proteins appear to be downregulated in the AD-afflicted brain. Enhancement/restoration of these 'protective' proteins, therefore, represents an attractive therapeutic avenue. Gene therapy is a desirable means of achieving this because it is not associated with the side-effects linked to systemic protein administration, and sustained protein expression virtually eliminates compliance issues. The current article represents a focused and succinct review of the better established 'protective' protein targets for gene therapy enhancement/restoration rather than being designed as an exhaustive review incorporating less validated protein subjects. In addition, we will discuss how the risks associated with uncontrolled or irreversible gene expression might be mitigated through combining neuronal-specific promoters, inducible expression systems and localised injections. Whilst many of the gene therapy targets reviewed herein are yet to enter clinical trials, preclinical testing has thus far demonstrated encouraging potential for the gene therapy-based treatment of AD.
Collapse
Affiliation(s)
- Lauren V Owens
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Christopher J Gaffney
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Edward T Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| |
Collapse
|
3
|
Deslauriers J, Toth M, Scadeng M, McKenna BS, Bussell R, Gresack J, Rissman R, Risbrough VB, Brown GG. DTI-identified microstructural changes in the gray matter of mice overexpressing CRF in the forebrain. Psychiatry Res Neuroimaging 2020; 304:111137. [PMID: 32731113 PMCID: PMC7508966 DOI: 10.1016/j.pscychresns.2020.111137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 11/18/2022]
Abstract
Increased corticotroping releasing factor (CRF) contributes to brain circuit abnormalities associated with stress-related disorders including posttraumatic stress disorder. However, the causal relationship between CRF hypersignaling and circuit abnormalities associated with stress disorders is unclear. We hypothesized that increased CRF exposure induces changes in limbic circuit morphology and functions. An inducible, forebrain-specific overexpression of CRF (CRFOE) transgenic mouse line was used to longitudinally investigate its chronic effects on behaviors and microstructural integrity of several brain regions. Behavioral and diffusion tensor imaging studies were performed before treatment, after 3-4 wks of treatment, and again 3 mo after treatment ended to assess recovery. CRFOE was associated with increased perseverative movements only after 3 wks of treatment, as well as reduced fractional anisotropy at 3 wks in the medial prefrontal cortex and increased fractional anisotropy in the ventral hippocampus at 3 mo compared to the control group. In the dorsal hippocampus, mean diffusivity was lower in CRFOE mice both during and after treatment ended. Our data suggest differential response and recovery patterns of cortical and hippocampal subregions in response to CRFOE. Overall these findings support a causal relationship between CRF hypersignaling and microstructural changes in brain regions relevant to stress disorders.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California San Diego, La Jolla, CA; Veterans Affairs Center of Excellence for Stress and Mental Health, La Jolla, CA; Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada.
| | - Mate Toth
- Department of Psychiatry, University of California San Diego, La Jolla, CA; Veterans Affairs Center of Excellence for Stress and Mental Health, La Jolla, CA; Department of Translational Behavioral Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | - Miriam Scadeng
- Department of Radiology, University of California San Diego, La Jolla, CA; Department of Anatomy and Medical Imaging, University of Auckland, New Zealand
| | - Benjamin S McKenna
- Department of Psychiatry, University of California San Diego, La Jolla, CA; Veterans Affairs Center of Excellence for Stress and Mental Health, La Jolla, CA
| | - Robert Bussell
- Department of Translational Behavioral Neuroscience, Institute of Experimental Medicine, Budapest, Hungary
| | | | - Robert Rissman
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA; Veterans Affairs Center of Excellence for Stress and Mental Health, La Jolla, CA
| | - Gregory G Brown
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| |
Collapse
|
4
|
Ross JA, Alexis R, Reyes BAS, Risbrough V, Van Bockstaele EJ. Localization of amyloid beta peptides to locus coeruleus and medial prefrontal cortex in corticotropin releasing factor overexpressing male and female mice. Brain Struct Funct 2019; 224:2385-2405. [PMID: 31250157 PMCID: PMC7371412 DOI: 10.1007/s00429-019-01915-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/17/2019] [Indexed: 01/25/2023]
Abstract
A culmination of evidence from the literature points to the Locus Coeruleus (LC)-Norepinephrine system as an underappreciated and understudied area of research in the context of Alzheimer's Disease (AD). Stress is a risk factor for developing AD, and is supported by multiple clinical and preclinical studies demonstrating that amplification of the stress system disrupts cellular and molecular processes at the synapse, promoting the production and accumulation of the amyloid beta (Aβ42) peptide. Stress-induced activation of the LC is mediated by corticotropin releasing factor (CRF) and CRF receptors exhibit sex-biased stress signaling. Sex differences are evident in the neurochemical, morphological and molecular regulation of LC neurons by CRF, providing a compelling basis for the higher prevalence of stress-related disorders such as AD in females. In the present study, we examined the cellular substrates for interactions between Aβ and tyrosine hydroxylase a marker of noradrenergic somatodendritic processes in the LC, and Dopamine-β-Hydroxylase (DβH) in the infralimbic medial prefrontal cortex (ILmPFC) in mice conditionally overexpressing CRF in the forebrain (CRFOE) under a Doxycycline (DOX) regulated tetO promoter. CRFOE was sufficient to elicit a redistribution of Aβ peptides in the somatodendritic processes of the LC of male and female transgenic mice, without altering total Aβ42 protein expression levels. DOX treated groups exhibited lysosomal compartments with apparent lipofuscin and abnormal morphology, indicating potential dysfunction of these Aβ42-clearing compartments. In female DOX treated groups, swollen microvessels with lipid-laden vacuoles were also observed, a sign of blood-brain-barrier dysfunction. Finally, sex differences were observed in the prefrontal cortex, as females responded to DOX treatment with increased frequency of co-localization of Aβ42 and DβH in noradrenergic axon terminals compared to vehicle treated controls, while male groups showed no significant changes. We hypothesize that the observed sex differences in Aβ42 distribution in this model of CRF hypersignaling is based on increased responsivity of female rodent CRFR1 in the LC. Aβ42 production is enhanced during increased neuronal activation, therefore, the excitation of DOX treated female CRFOE LC neurons projecting to the mPFC may exhibit more frequent co-localization with Aβ due to increased neuronal activity of noradrenergic neurons.
Collapse
Affiliation(s)
- Jennifer A Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA.
| | - Rody Alexis
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| | - Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| | - Victoria Risbrough
- Department of Psychiatry, University of California, San Diego, CA, 92093, USA
- Center of Excellence for Stress and Mental Health, San Diego VA Health Services, La Jolla, CA, USA
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| |
Collapse
|
5
|
Procaspase activating compound 1 controls tetracycline repressor-regulated gene expression system. Biosci Rep 2019; 39:BSR20180793. [PMID: 30538170 PMCID: PMC6328932 DOI: 10.1042/bsr20180793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/29/2018] [Accepted: 12/07/2018] [Indexed: 11/17/2022] Open
Abstract
The tetracycline repressor (TetR)-regulated system is a widely used tool to study gene functions through control of its expression. Various effectors such as tetracycline (Tc) and doxycycline (Dox) quickly induce or shut down gene expression, but reversing gene expression has not been eligible due to long half-lives of such effectors. Here, we found that procaspase activating compound 1 (PAC-1) rapidly reduces transient expression of TetR-regulated green fluorescent protein (GFP) in mammalian cells. Next, we applied PAC-1 to control of expression of transient receptor potential melastatin 7 (TRPM7) protein, whose downstream cellular events can be monitored by cell morphological changes. We observed that PAC-1 quickly reduces TRPM7 expression, consequently affecting cell morphology regulated by TRPM7. The present study demonstrates the first small molecule that efficiently turns off the TetR-regulated gene expression in mammalian cells, thereby precisely regulating the expression level of target gene.
Collapse
|
6
|
Choi B, Lee HW, Mo S, Kim JY, Kim HW, Rhyu IJ, Hong E, Lee YK, Choi JS, Kim CH, Kim H. Inositol 1,4,5-trisphosphate 3-kinase A overexpressed in mouse forebrain modulates synaptic transmission and mGluR-LTD of CA1 pyramidal neurons. PLoS One 2018; 13:e0193859. [PMID: 29617377 PMCID: PMC5884490 DOI: 10.1371/journal.pone.0193859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/20/2018] [Indexed: 11/18/2022] Open
Abstract
Inositol 1,4,5-trisphosphate 3-kinase A (IP3K-A) regulates the level of the inositol polyphosphates, inositol trisphosphate (IP3) and inositol tetrakisphosphate to modulate cellular signaling and intracellular calcium homeostasis in the central nervous system. IP3K-A binds to F-actin in an activity-dependent manner and accumulates in dendritic spines, where it is involved in the regulation of synaptic plasticity. IP3K-A knockout mice exhibit deficits in some forms of hippocampus-dependent learning and synaptic plasticity, such as long-term potentiation in the dentate gyrus synapses of the hippocampus. In the present study, to further elucidate the role of IP3K-A in the brain, we developed a transgenic (Tg) mouse line in which IP3K-A is conditionally overexpressed approximately 3-fold in the excitatory neurons of forebrain regions, including the hippocampus. The Tg mice showed an increase in both presynaptic release probability of evoked responses, along with bigger synaptic vesicle pools, and miniature excitatory postsynaptic current amplitude, although the spine density or the expression levels of the postsynaptic density-related proteins NR2B, synaptotagmin 1, and PSD-95 were not affected. Hippocampal-dependent learning and memory tasks, including novel object recognition and radial arm maze tasks, were partially impaired in Tg mice. Furthermore, (R,S)-3,5-dihydroxyphenylglycine-induced metabotropic glutamate receptor long-term depression was inhibited in Tg mice and this inhibition was dependent on protein kinase C but not on the IP3 receptor. Long-term potentiation and depression dependent on N-methyl-d-aspartate receptor were marginally affected in Tg mice. In summary, this study shows that overexpressed IP3K-A plays a role in some forms of hippocampus-dependent learning and memory tasks as well as in synaptic transmission and plasticity by regulating both presynaptic and postsynaptic functions.
Collapse
Affiliation(s)
- Byungil Choi
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Hyun Woo Lee
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Seojung Mo
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Jin Yong Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Hyun Wook Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
| | - Eunhwa Hong
- Department of Psychology, Korea University, Seoul, Korea
| | - Yeon Kyung Lee
- Department of Psychology, Korea University, Seoul, Korea
| | - June-Seek Choi
- Department of Psychology, Korea University, Seoul, Korea
| | - Chong-Hyun Kim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology and Neuroscience Program, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Korea
- * E-mail: (C-HK); (HK)
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Brain Korea, Seoul, Korea
- * E-mail: (C-HK); (HK)
| |
Collapse
|
7
|
Liliom H, Tárnok K, Ábrahám Z, Rácz B, Hausser A, Schlett K. Protein kinase D exerts neuroprotective functions during oxidative stress via nuclear factor kappa B-independent signaling pathways. J Neurochem 2017; 142:948-961. [DOI: 10.1111/jnc.14131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Hanna Liliom
- Department of Physiology and Neurobiology; Eötvös Loránd University; Budapest Hungary
| | - Krisztián Tárnok
- Department of Physiology and Neurobiology; Eötvös Loránd University; Budapest Hungary
| | - Zsófia Ábrahám
- Department of Physiology and Neurobiology; Eötvös Loránd University; Budapest Hungary
| | - Bence Rácz
- Department of Anatomy and Histology; University of Veterinary Medicine; Budapest Hungary
| | - Angelika Hausser
- Institute of Cell Biology and Immunology; University Stuttgart; Stuttgart Germany
- Stuttgart Research Center Systems Biology; University of Stuttgart; Stuttgart Germany
| | - Katalin Schlett
- Department of Physiology and Neurobiology; Eötvös Loránd University; Budapest Hungary
- MTA-ELTE-NAP B - Neuronal Cell Biology Research Group; Eötvös Loránd University; Budapest Hungary
| |
Collapse
|
8
|
The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult brain. Neurobiol Learn Mem 2016; 135:100-114. [DOI: 10.1016/j.nlm.2016.07.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/23/2016] [Accepted: 07/28/2016] [Indexed: 11/22/2022]
|
9
|
Jul P, Volbracht C, de Jong IEM, Helboe L, Elvang AB, Pedersen JT. Hyperactivity with Agitative-Like Behavior in a Mouse Tauopathy Model. J Alzheimers Dis 2016; 49:783-95. [PMID: 26519432 DOI: 10.3233/jad-150292] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tauopathies, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), are characterized by formation of neurofibrillary tangles consisting of hyperphosphorylated tau. In addition to memory loss, patients experience behavioral symptoms such as agitation, aggression, depression, and insomnia. We explored the behavioral phenotype of a mouse model (rTg4510) carrying the human tau P301L mutation found in a familial form of FTD. We tested these mice in locomotor activity assays as well as in the Morris water maze to access spatial memory. In addition to cognitive impairments, rTg4510 mice exhibited a hyperactivity phenotype which correlated with progression of tau pathology and was dependent on P301L tau transgene expression. The hyperactive phenotype was characterized by significantly increased locomotor activity in a novel and in a simulated home cage environment together with a disturbed day/night cycle. The P301L-tau-dependent hyperactivity and agitative-like phenotype suggests that these mice may form a correlate to some of the behavioral disturbances observed in advanced AD and FTD.
Collapse
|
10
|
Marzo A, Galli S, Lopes D, McLeod F, Podpolny M, Segovia-Roldan M, Ciani L, Purro S, Cacucci F, Gibb A, Salinas PC. Reversal of Synapse Degeneration by Restoring Wnt Signaling in the Adult Hippocampus. Curr Biol 2016; 26:2551-2561. [PMID: 27593374 PMCID: PMC5070786 DOI: 10.1016/j.cub.2016.07.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/12/2016] [Indexed: 11/30/2022]
Abstract
Synapse degeneration occurs early in neurodegenerative diseases and correlates strongly with cognitive decline in Alzheimer’s disease (AD). The molecular mechanisms that trigger synapse vulnerability and those that promote synapse regeneration after substantial synaptic failure remain poorly understood. Increasing evidence suggests a link between a deficiency in Wnt signaling and AD. The secreted Wnt antagonist Dickkopf-1 (Dkk1), which is elevated in AD, contributes to amyloid-β-mediated synaptic failure. However, the impact of Dkk1 at the circuit level and the mechanism by which synapses disassemble have not yet been explored. Using a transgenic mouse model that inducibly expresses Dkk1 in the hippocampus, we demonstrate that Dkk1 triggers synapse loss, impairs long-term potentiation, enhances long-term depression, and induces learning and memory deficits. We decipher the mechanism involved in synapse loss induced by Dkk1 as it can be prevented by combined inhibition of the Gsk3 and RhoA-Rock pathways. Notably, after loss of synaptic connectivity, reactivation of the Wnt pathway by cessation of Dkk1 expression completely restores synapse number, synaptic plasticity, and long-term memory. These findings demonstrate the remarkable capacity of adult neurons to regenerate functional circuits and highlight Wnt signaling as a targetable pathway for neuronal circuit recovery after synapse degeneration. Wnt signaling is required for synapse integrity in the adult hippocampus Dkk1 induces synapse loss and deficits in synaptic plasticity and long-term memory Dkk1 disassembles synapses by activating the Gsk3 and Rock pathways Synapse loss and memory defects are reversible by reactivation of the Wnt pathway
Collapse
Affiliation(s)
- Aude Marzo
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Soledad Galli
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Douglas Lopes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Faye McLeod
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Marina Podpolny
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | | | - Lorenza Ciani
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Silvia Purro
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Francesca Cacucci
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Alasdair Gibb
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
11
|
Lauritzen KH, Hasan-Olive MM, Regnell CE, Kleppa L, Scheibye-Knudsen M, Gjedde A, Klungland A, Bohr VA, Storm-Mathisen J, Bergersen LH. A ketogenic diet accelerates neurodegeneration in mice with induced mitochondrial DNA toxicity in the forebrain. Neurobiol Aging 2016; 48:34-47. [PMID: 27639119 DOI: 10.1016/j.neurobiolaging.2016.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022]
Abstract
Mitochondrial genome maintenance plays a central role in preserving brain health. We previously demonstrated accumulation of mitochondrial DNA damage and severe neurodegeneration in transgenic mice inducibly expressing a mutated mitochondrial DNA repair enzyme (mutUNG1) selectively in forebrain neurons. Here, we examine whether severe neurodegeneration in mutUNG1-expressing mice could be rescued by feeding the mice a ketogenic diet, which is known to have beneficial effects in several neurological disorders. The diet increased the levels of superoxide dismutase 2, and mitochondrial mass, enzymes, and regulators such as SIRT1 and FIS1, and appeared to downregulate N-methyl-D-aspartic acid (NMDA) receptor subunits NR2A/B and upregulate γ-aminobutyric acid A (GABAA) receptor subunits α1. However, unexpectedly, the ketogenic diet aggravated neurodegeneration and mitochondrial deterioration. Electron microscopy showed structurally impaired mitochondria accumulating in neuronal perikarya. We propose that aggravation is caused by increased mitochondrial biogenesis of generally dysfunctional mitochondria. This study thereby questions the dogma that a ketogenic diet is unambiguously beneficial in mitochondrial disorders.
Collapse
Affiliation(s)
- Knut H Lauritzen
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Md Mahdi Hasan-Olive
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Christine E Regnell
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway; Center for Healthy Aging and Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Liv Kleppa
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Morten Scheibye-Knudsen
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Albert Gjedde
- Center for Healthy Aging and Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Klungland
- Institute of Medical Microbiology, Oslo University Hospital and University of Oslo, Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jon Storm-Mathisen
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Linda H Bergersen
- Synaptic Neurochemistry Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway; Center for Healthy Aging and Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Overexpression of Forebrain CRH During Early Life Increases Trauma Susceptibility in Adulthood. Neuropsychopharmacology 2016; 41:1681-90. [PMID: 26538448 PMCID: PMC4832031 DOI: 10.1038/npp.2015.338] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 09/30/2015] [Accepted: 10/29/2015] [Indexed: 01/02/2023]
Abstract
Although early-life stress is a significant risk factor for developing anxiety disorders, including posttraumatic stress disorder (PTSD), the underlying mechanisms are unclear. Corticotropin releasing hormone (CRH) is disrupted in individuals with PTSD and early-life stress and hence may mediate the effects of early-life stress on PTSD risk. We hypothesized that CRH hyper-signaling in the forebrain during early development is sufficient to increase response to trauma in adulthood. To test this hypothesis, we induced transient, forebrain-specific, CRH overexpression during early-life (pre-puberty, CRHOEdev) in double-mutant mice (Camk2a-rtta2 × tetO-Crh) and tested their behavioral and gene expression responses to the predator stress model of PTSD in adulthood. In one cohort of CRHOEdev exposed and unexposed mice, avoidance and arousal behaviors were examined 7-15 days after exposure to predator stress. In another cohort, gene expression changes in Crhr1, Crhr2, and Fkbp51 in forebrain of CRHOEdev exposed and unexposed mice were examined 7 days after predator stress. CRHOEdev induced robust increases in startle reactivity and reductions in startle inhibition independently of predator stress in both male and female mice. Avoidance behaviors after predator stress were highly dependent on sex and CRHOEdev exposure. Whereas stressed females exhibited robust avoidance responses that were not altered by CRHOEdev, males developed significant avoidance only when exposed to both CRHOEdev and stress. Quantitative real-time-PCR analysis indicated that CRHOEdev unexposed males exhibit significant changes in Crhr2 expression in the amygdala and bed nucleus stria terminalis in response to stress, whereas males exposed to CRHOEdev did not. Similar to CRHOEdev males, females exhibited no significant Crhr2 gene expression changes in response to stress. Cortical Fkbp51 expression was also significantly reduced by stress and CRHOEdev exposure in males, but not in females. These findings indicate that forebrain CRH hyper-signaling in early-life is sufficient to increase enduring effects of adult trauma and attenuate Crhr2 expression changes in response to stress in males. These data support growing evidence for significant sex differences in response to trauma, and support further study of CRHR2 as a candidate mechanism for PTSD risk.
Collapse
|
13
|
Bencsik N, Szíber Z, Liliom H, Tárnok K, Borbély S, Gulyás M, Rátkai A, Szűcs A, Hazai-Novák D, Ellwanger K, Rácz B, Pfizenmaier K, Hausser A, Schlett K. Protein kinase D promotes plasticity-induced F-actin stabilization in dendritic spines and regulates memory formation. J Cell Biol 2015; 210:771-83. [PMID: 26304723 PMCID: PMC4555815 DOI: 10.1083/jcb.201501114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/23/2015] [Indexed: 02/07/2023] Open
Abstract
PKD regulates the stabilization of the F-actin network within dendritic spines upon chemically induced plasticity changes and is needed for proper hippocampal LTP and spatial memory formation. Actin turnover in dendritic spines influences spine development, morphology, and plasticity, with functional consequences on learning and memory formation. In nonneuronal cells, protein kinase D (PKD) has an important role in stabilizing F-actin via multiple molecular pathways. Using in vitro models of neuronal plasticity, such as glycine-induced chemical long-term potentiation (LTP), known to evoke synaptic plasticity, or long-term depolarization block by KCl, leading to homeostatic morphological changes, we show that actin stabilization needed for the enlargement of dendritic spines is dependent on PKD activity. Consequently, impaired PKD functions attenuate activity-dependent changes in hippocampal dendritic spines, including LTP formation, cause morphological alterations in vivo, and have deleterious consequences on spatial memory formation. We thus provide compelling evidence that PKD controls synaptic plasticity and learning by regulating actin stability in dendritic spines.
Collapse
Affiliation(s)
- Norbert Bencsik
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Zsófia Szíber
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Hanna Liliom
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Krisztián Tárnok
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Sándor Borbély
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Márton Gulyás
- MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| | - Anikó Rátkai
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Attila Szűcs
- MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| | - Diána Hazai-Novák
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, H-1400 Budapest, Hungary
| | - Kornelia Ellwanger
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Bence Rácz
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, H-1400 Budapest, Hungary
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| |
Collapse
|
14
|
Lauritzen KH, Kleppa L, Aronsen JM, Eide L, Carlsen H, Haugen ØP, Sjaastad I, Klungland A, Rasmussen LJ, Attramadal H, Storm-Mathisen J, Bergersen LH. Impaired dynamics and function of mitochondria caused by mtDNA toxicity leads to heart failure. Am J Physiol Heart Circ Physiol 2015; 309:H434-49. [PMID: 26055793 DOI: 10.1152/ajpheart.00253.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/02/2015] [Indexed: 11/22/2022]
Abstract
Cardiac mitochondrial dysfunction has been implicated in heart failure of diverse etiologies. Generalized mitochondrial disease also leads to cardiomyopathy with various clinical manifestations. Impaired mitochondrial homeostasis may over time, such as in the aging heart, lead to cardiac dysfunction. Mitochondrial DNA (mtDNA), close to the electron transport chain and unprotected by histones, may be a primary pathogenetic site, but this is not known. Here, we test the hypothesis that cumulative damage of cardiomyocyte mtDNA leads to cardiomyopathy and heart failure. Transgenic mice with Tet-on inducible, cardiomyocyte-specific expression of a mutant uracil-DNA glycosylase 1 (mutUNG1) were generated. The mutUNG1 is known to remove thymine in addition to uracil from the mitochondrial genome, generating apyrimidinic sites, which obstruct mtDNA function. Following induction of mutUNG1 in cardiac myocytes by administering doxycycline, the mice developed hypertrophic cardiomyopathy, leading to congestive heart failure and premature death after ∼2 mo. The heart showed reduced mtDNA replication, severely diminished mtDNA transcription, and suppressed mitochondrial respiration with increased Pgc-1α, mitochondrial mass, and antioxidative defense enzymes, and finally failing mitochondrial fission/fusion dynamics and deteriorating myocardial contractility as the mechanism of heart failure. The approach provides a model with induced cardiac-restricted mtDNA damage for investigation of mtDNA-based heart disease.
Collapse
Affiliation(s)
- Knut H Lauritzen
- Department of Oral Biology, Brain and Muscle Energy Group, University of Oslo, Oslo, Norway; Department of Anatomy, Institute of Basic Medical Sciences, and Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway
| | - Liv Kleppa
- Department of Oral Biology, Brain and Muscle Energy Group, University of Oslo, Oslo, Norway; Department of Anatomy, Institute of Basic Medical Sciences, and Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo, Oslo, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Norway, Oslo, Norway
| | - Lars Eide
- Department of Medical Biochemistry, University of Oslo, Oslo, Norway
| | - Harald Carlsen
- Department of Nutrition Research, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Øyvind P Haugen
- Department of Oral Biology, Brain and Muscle Energy Group, University of Oslo, Oslo, Norway; Department of Anatomy, Institute of Basic Medical Sciences, and Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital Ullevål and University of Oslo, Oslo, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Norway, Oslo, Norway
| | - Arne Klungland
- Department of Nutrition Research, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Institute of Medical Microbiology, Oslo University Hospital, Oslo, Norway
| | - Lene Juel Rasmussen
- Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway; and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Jon Storm-Mathisen
- Department of Anatomy, Institute of Basic Medical Sciences, and Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway
| | - Linda H Bergersen
- Department of Oral Biology, Brain and Muscle Energy Group, University of Oslo, Oslo, Norway; Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
15
|
New Transgenic Technologies. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
16
|
Galli S, Lopes DM, Ammari R, Kopra J, Millar SE, Gibb A, Salinas PC. Deficient Wnt signalling triggers striatal synaptic degeneration and impaired motor behaviour in adult mice. Nat Commun 2014; 5:4992. [PMID: 25318560 PMCID: PMC4218967 DOI: 10.1038/ncomms5992] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 08/15/2014] [Indexed: 12/31/2022] Open
Abstract
Synapse degeneration is an early and invariant feature of neurodegenerative diseases. Indeed, synapse loss occurs prior to neuronal degeneration and correlates with the symptom severity of these diseases. However, the molecular mechanisms that trigger synaptic loss remain poorly understood. Here we demonstrate that deficient Wnt signalling elicits synaptic degeneration in the adult striatum. Inducible expression of the secreted Wnt antagonist Dickkopf1 (Dkk1) in adult mice (iDkk1) decreases the number of cortico-striatal glutamatergic synapses and of D1 and D2 dopamine receptor clusters. Synapse loss occurs in the absence of axon retraction or cell death. The remaining excitatory terminals contain fewer synaptic vesicles and have a reduced probability of evoked transmitter release. IDkk1 mice show impaired motor coordination and are irresponsive to amphetamine. These studies identify Wnts as key endogenous regulators of synaptic maintenance and suggest that dysfunction in Wnt signalling contributes to synaptic degeneration at early stages in neurodegenerative diseases.
Collapse
Affiliation(s)
- Soledad Galli
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Douglas M. Lopes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Rachida Ammari
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Sarah E. Millar
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Alasdair Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
17
|
Schmidt S, Berens C, Klotzsche M. A novel TetR-regulating peptide turns off rtTA-mediated activation of gene expression. PLoS One 2014; 9:e96546. [PMID: 24810590 PMCID: PMC4014509 DOI: 10.1371/journal.pone.0096546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/09/2014] [Indexed: 11/18/2022] Open
Abstract
Conditional regulation of gene expression is a powerful and indispensable method for analyzing gene function. The “Tet-On” system is a tool widely used for that purpose. Here, the transregulator rtTA mediates expression of a gene of interest after addition of the small molecule effector doxycycline. Although very effective in rapidly turning on gene expression, the system is hampered by the long half-life of doxycycline which makes shutting down gene expression rapidly very difficult to achieve. We isolated an rtTA-binding peptide by in vivo selection that acts as a doxycycline antagonist and leads to rapid and efficient shut down of rtTA-mediated reporter gene expression in a human cell line. This peptide represents the basis for novel effector molecules which complement the “Tet-system” by enabling the investigator to rapidly turn gene expression not just on at will, but now also off.
Collapse
Affiliation(s)
- Sebastian Schmidt
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Berens
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marcus Klotzsche
- Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
18
|
Toth M, Gresack JE, Bangasser DA, Plona Z, Valentino RJ, Flandreau EI, Mansuy IM, Merlo-Pich E, Geyer MA, Risbrough VB. Forebrain-specific CRF overproduction during development is sufficient to induce enduring anxiety and startle abnormalities in adult mice. Neuropsychopharmacology 2014; 39:1409-19. [PMID: 24326400 PMCID: PMC3988544 DOI: 10.1038/npp.2013.336] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/23/2013] [Accepted: 11/16/2013] [Indexed: 11/09/2022]
Abstract
Corticotropin releasing factor (CRF) regulates physiological and behavioral responses to stress. Trauma in early life or adulthood is associated with increased CRF in the cerebrospinal fluid and heightened anxiety. Genetic variance in CRF receptors is linked to altered risk for stress disorders. Thus, both heritable differences and environmentally induced changes in CRF neurotransmission across the lifespan may modulate anxiety traits. To test the hypothesis that CRF hypersignaling is sufficient to modify anxiety-related phenotypes (avoidance, startle, and conditioned fear), we induced transient forebrain-specific overexpression of CRF (CRFOE) in mice (1) during development to model early-life stress, (2) in adulthood to model adult-onset stress, or (3) across the entire postnatal lifespan to model heritable increases in CRF signaling. The consequences of these manipulations on CRF peptide levels and behavioral responses were examined in adulthood. We found that transient CRFOE during development decreased startle habituation and prepulse inhibition, and increased avoidance (particularly in females) recapitulating the behavioral effects of lifetime CRFOE despite lower CRF peptide levels at testing. In contrast, CRFOE limited to adulthood reduced contextual fear learning in females and increased startle reactivity in males but did not change avoidance or startle plasticity. These findings suggest that forebrain CRFOE limited to development is sufficient to induce enduring alterations in startle plasticity and anxiety, while forebrain CRFOE during adulthood results in a different phenotype profile. These findings suggest that startle circuits are particularly sensitive to forebrain CRFOE, and that the impact of CRFOE may be dependent on the time of exposure.
Collapse
Affiliation(s)
- Mate Toth
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Jodi E Gresack
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA,Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Debra A Bangasser
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA,Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Zach Plona
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rita J Valentino
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Isabelle M Mansuy
- Brain Research Institute, University and ETH Zürich, Zürich, Switzerland
| | - Emilio Merlo-Pich
- Neuroscience Disease Therapeutic Area, Pharmaceutical Division, F. Hoffman—La Roche, Basel, Switzerland
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA,Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC0804, La Jolla CA 92093-0804, USA, Tel: +1 16195433582, Fax: +1 16195432475, E-mail:
| |
Collapse
|
19
|
Heitz F, Johansson T, Baumgärtel K, Gecaj R, Pelczar P, Mansuy IM. Heritable and inducible gene knockdown in astrocytes or neurons in vivo by a combined lentiviral and RNAi approach. Front Cell Neurosci 2014; 8:62. [PMID: 24678290 PMCID: PMC3958736 DOI: 10.3389/fncel.2014.00062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/12/2014] [Indexed: 11/13/2022] Open
Abstract
Gene knockout by homologous recombination is a popular method to study gene functions in the mouse in vivo. However, its lack of temporal control has limited the interpretation of knockout studies because the complete elimination of a gene product often alters developmental processes, and can induce severe malformations or lethality. Conditional gene knockdown has emerged as a compelling alternative to gene knockout, an approach well-established in vitro but that remains challenging in vivo, especially in the adult brain. Here, we report a method for conditional and cell-specific gene knockdown in the mouse brain in vivo that combines Cre-mediated RNA interference (RNAi) with classical and lentivirus-mediated transgenesis. The method is based on the inducible expression of a silencing short hairpin RNA (shRNA) introduced in mice by lentivirus-mediated transgenesis, and on its activation by excision of a floxed stop EGFP reporter with an inducible Cre recombinase expressed in astrocytes or in neurons. This dual system should be of broad utility for comparative studies of gene functions in these two cell types in vivo.
Collapse
Affiliation(s)
- Fabrice Heitz
- Brain Research Institute, Medical Faculty of the University of Zürich and Department of Biology of the Swiss Federal Institute of Technology Zürich, Switzerland
| | - Torbjörn Johansson
- Institute of Pharmacology and Toxicology, Medical Faculty of the University of Zürich Zürich, Switzerland
| | - Karsten Baumgärtel
- Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| | - Rreze Gecaj
- Brain Research Institute, Medical Faculty of the University of Zürich and Department of Biology of the Swiss Federal Institute of Technology Zürich, Switzerland
| | - Pawel Pelczar
- Institute of Laboratory Animal Science, University of Zürich Zürich, Switzerland
| | - Isabelle M Mansuy
- Brain Research Institute, Medical Faculty of the University of Zürich and Department of Biology of the Swiss Federal Institute of Technology Zürich, Switzerland
| |
Collapse
|
20
|
Cook C, Dunmore JH, Murray ME, Scheffel K, Shukoor N, Tong J, Castanedes-Casey M, Phillips V, Rousseau L, Penuliar MS, Kurti A, Dickson DW, Petrucelli L, Fryer JD. Severe amygdala dysfunction in a MAPT transgenic mouse model of frontotemporal dementia. Neurobiol Aging 2013; 35:1769-77. [PMID: 24503275 DOI: 10.1016/j.neurobiolaging.2013.12.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 11/26/2013] [Accepted: 12/09/2013] [Indexed: 01/05/2023]
Abstract
Frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) is a neurodegenerative tauopathy caused by mutations in the tau gene (MAPT). Individuals with FTDP-17 have deficits in learning, memory, and language, in addition to personality and behavioral changes that are often characterized by a lack of social inhibition. Several transgenic mouse models expressing tau mutations have been tested extensively for memory or motor impairments, though reports of amygdala-dependent behaviors are lacking. To this end, we tested the rTg4510 mouse model on a behavioral battery that included amygdala-dependent tasks of exploration. As expected, rTg4510 mice exhibit profound impairments in hippocampal-dependent learning and memory tests, including contextual fear conditioning. However, rTg4510 mice also display an abnormal hyperexploratory phenotype in the open-field assay, elevated plus maze, light-dark exploration, and cued fear conditioning, indicative of amygdala dysfunction. Furthermore, significant tau burden is detected in the amygdala of both rTg4510 mice and human FTDP-17 patients, suggesting that the rTg4510 mouse model recapitulates the behavioral disturbances and neurodegeneration of the amygdala characteristic of FTDP-17.
Collapse
Affiliation(s)
- Casey Cook
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Judy H Dunmore
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Kristyn Scheffel
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Nawsheen Shukoor
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Jimei Tong
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | | | - Virginia Phillips
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Linda Rousseau
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Michael S Penuliar
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA; Neurobiology of Disease Program, Mayo Graduate School, Rochester, MN, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA; Neurobiology of Disease Program, Mayo Graduate School, Rochester, MN, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA; Neurobiology of Disease Program, Mayo Graduate School, Rochester, MN, USA.
| |
Collapse
|
21
|
Penke Z, Morice E, Veyrac A, Gros A, Chagneau C, LeBlanc P, Samson N, Baumgärtel K, Mansuy IM, Davis S, Laroche S. Zif268/Egr1 gain of function facilitates hippocampal synaptic plasticity and long-term spatial recognition memory. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130159. [PMID: 24298160 DOI: 10.1098/rstb.2013.0159] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It is well established that Zif268/Egr1, a member of the Egr family of transcription factors, is critical for the consolidation of several forms of memory; however, it is as yet uncertain whether increasing expression of Zif268 in neurons can facilitate memory formation. Here, we used an inducible transgenic mouse model to specifically induce Zif268 overexpression in forebrain neurons and examined the effect on recognition memory and hippocampal synaptic transmission and plasticity. We found that Zif268 overexpression during the establishment of memory for objects did not change the ability to form a long-term memory of objects, but enhanced the capacity to form a long-term memory of the spatial location of objects. This enhancement was paralleled by increased long-term potentiation in the dentate gyrus of the hippocampus and by increased activity-dependent expression of Zif268 and selected Zif268 target genes. These results provide novel evidence that transcriptional mechanisms engaging Zif268 contribute to determining the strength of newly encoded memories.
Collapse
Affiliation(s)
- Zsuzsa Penke
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, , Orsay 91405, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chao JS, Chao CC, Chang CL, Chiu YR, Yuan CJ. Development of single-vector Tet-on inducible systems with high sensitivity to doxycycline. Mol Biotechnol 2012; 51:240-6. [PMID: 22002194 DOI: 10.1007/s12033-011-9461-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Single-vector Tet-on systems were developed to enable the tight regulation of transgenes in mammalian cells with a low dosage of doxycycline. Both the regulatory and the responsive units were integrated in a single vector and separated by a short DNA segment (214 bp). In the developed single-vector Tet-on systems, a high level of expression of the transgene can be induced by doxycycline at a concentration of as low as 1 ng/ml, which is 500-1,000 times lower than that usually utilized in other Tet-on systems. The single-vector Tet-on system developed here exhibited 3.5-10.8 times greater inducibility of the transgene in response to doxycycline than did a dual-vector system from a commercial source. Further studies indicate that the basal activity of Tet-on systems depends greatly on the strength of the promoter that controls the transactivator. The basal activity of Tet-on systems was high when the transactivator that was directed by the human cytomegalovirus promoter, and it was almost undetectable when the transactivator was placed under the control of a moderate strength mouse mammary tumor virus promoter. Moreover, the introduction of selectable markers allows the developed single-vector Tet-on systems to facilitate the generation of conditional transgenic cells and animals with high inducibility, low basal activity and detrimental effects of the long-term administration of doxycycline.
Collapse
Affiliation(s)
- Jiun-Shuan Chao
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 300, Taiwan, ROC
| | | | | | | | | |
Collapse
|
23
|
Kozlova EN, Berens C. Guiding Differentiation of Stem Cells in Vivo by Tetracycline-Controlled Expression of Key Transcription Factors. Cell Transplant 2012; 21:2537-54. [DOI: 10.3727/096368911x637407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplantation of stem or progenitor cells is an attractive strategy for cell replacement therapy. However, poor long-term survival and insufficiently reproducible differentiation to functionally appropriate cells in vivo still present major obstacles for translation of this methodology to clinical applications. Numerous experimental studies have revealed that the expression of just a few transcription factors can be sufficient to drive stem cell differentiation toward a specific cell type, to transdifferentiate cells from one fate to another, or to dedifferentiate mature cells to pluripotent stem/progenitor cells (iPSCs). We thus propose here to apply the strategy of expressing the relevant key transcription factors to guide the differentiation of transplanted cells to the desired cell fate in vivo. To achieve this requires tools allowing us to control the expression of these genes in the transplant. Here, we describe drug-inducible systems that allow us to sequentially and timely activate gene expression from the outside, with a particular emphasis on the Tet system, which has been widely and successfully used in stem cells. These regulatory systems offer a tool for strictly limiting gene expression to the respective optimal stage after transplantation. This approach will direct the differentiation of the immature stem/progenitor cells in vivo to the desired cell type.
Collapse
Affiliation(s)
- Elena N Kozlova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
24
|
Dynamic histone marks in the hippocampus and cortex facilitate memory consolidation. Nat Commun 2012; 3:991. [PMID: 22871810 DOI: 10.1038/ncomms1997] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/10/2012] [Indexed: 01/14/2023] Open
Abstract
Memory consolidation requires a timely controlled interplay between the hippocampus, a brain region important for memory formation, and the cortex, a region recruited for memory storage. Here we show that memory consolidation is associated with specific epigenetic modifications on histone proteins that have a distinct dynamic in these brain areas. While in the hippocampus, histone post-translational modifications (PTMs) are rapidly and transiently activated after learning, in the cortex they are induced with delay but persist over time. When these histone PTMs are increased in vivo by transgenic intervention or intense training, they facilitate memory consolidation. Conversely, when they are pharmacologically blocked, memory consolidation is impaired. These histone PTMs are further associated with the expression of the immediate early gene zif268, a transcription factor that favours memory consolidation. These findings reveal the spatiotemporal dynamics of histone marks during memory consolidation, and demonstrate their inherent 'mnemonic' property.
Collapse
|
25
|
Harper KM, Hiramoto T, Tanigaki K, Kang G, Suzuki G, Trimble W, Hiroi N. Alterations of social interaction through genetic and environmental manipulation of the 22q11.2 gene Sept5 in the mouse brain. Hum Mol Genet 2012; 21:3489-99. [PMID: 22589251 DOI: 10.1093/hmg/dds180] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Social behavior dysfunction is a symptomatic element of schizophrenia and autism spectrum disorder (ASD). Although altered activities in numerous brain regions are associated with defective social cognition and perception, the causative relationship between these altered activities and social cognition and perception-and their genetic underpinnings-are not known in humans. To address these issues, we took advantage of the link between hemizygous deletion of human chromosome 22q11.2 and high rates of social behavior dysfunction, schizophrenia and ASD. We genetically manipulated Sept5, a 22q11.2 gene, and evaluated its role in social interaction in mice. Sept5 deficiency, against a high degree of homogeneity in a congenic genetic background, selectively impaired active affiliative social interaction in mice. Conversely, virally guided overexpression of Sept5 in the hippocampus or, to a lesser extent, the amygdala elevated levels of active affiliative social interaction in C57BL/6J mice. Congenic knockout mice and mice overexpressing Sept5 in the hippocampus or amygdala were indistinguishable from control mice in novelty and olfactory responses, anxiety or motor activity. Moreover, post-weaning individual housing, an environmental condition designed to reduce stress in male mice, selectively raised levels of Sept5 protein in the amygdala and increased active affiliative social interaction in C57BL/6J mice. These findings identify this 22q11.2 gene in the hippocampus and amygdala as a determinant of social interaction and suggest that defective social interaction seen in 22q11.2-associated schizophrenia and ASD can be genetically and environmentally modified by altering this 22q11.2 gene.
Collapse
Affiliation(s)
- Kathryn M Harper
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Burger A, Koesters R, Schäfer BW, Niggli FK. Generation of a novel rtTA transgenic mouse to induce time-controlled, tissue-specific alterations in Pax2-expressing cells. Genesis 2011; 49:797-802. [PMID: 21157934 DOI: 10.1002/dvg.20701] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 11/23/2010] [Accepted: 12/04/2010] [Indexed: 11/07/2022]
Abstract
The paired-box transcription factor Pax2 plays a major role in early development of the kidney and the central nervous system. It is expressed in the metanephric mesenchyme of the developing kidney, at the midbrain-hindbrain boundary and the anlagen of the inner ear. The early expression of Pax2, especially in the developing kidney, prompted us to use this locus as a novel genetic tool to introduce temporally-controlled expression of transgenes. We generated a transgenic Pax2-rtTA mouse strain through genetic recombineering using a large BAC clone which drives expression of TetO-controlled transgenes upon doxycycline treatment in natively Pax2-expressing tissues. We show that expression of a TetO-responsive lacZ gene is tightly regulated by addition of doxycycline and can be detected in all Pax2-expressing tissues. Our transgenic Pax2-rtTA mouse thus represents a suitable tool to study the cell fates and molecular pathways in Pax2-positive tissues during development, such as the kidney. We further propose that the Pax2-rtTA tool has great potential to induce time-controlled, tissue-specific alterations for tumorigenic transformation of Pax2-expressing cells for generating in vivo tumor models, such as Wilms tumor.
Collapse
Affiliation(s)
- Alexa Burger
- Department of Oncology, University Children's Hospital, Zurich, Switzerland
| | | | | | | |
Collapse
|
27
|
Protein phosphatase 1 (PP1) is a post-translational regulator of the mammalian circadian clock. PLoS One 2011; 6:e21325. [PMID: 21712997 PMCID: PMC3119686 DOI: 10.1371/journal.pone.0021325] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 05/25/2011] [Indexed: 01/25/2023] Open
Abstract
Circadian clocks coordinate the timing of important biological processes. Interconnected transcriptional and post-translational feedback loops based on a set of clock genes generate and maintain these rhythms with a period of about 24 hours. Many clock proteins undergo circadian cycles of post-translational modifications. Among these modifications, protein phosphorylation plays an important role in regulating activity, stability and intracellular localization of clock components. Several protein kinases were characterized as regulators of the circadian clock. However, the function of protein phosphatases, which balance phosphorylation events, in the mammalian clock mechanism is less well understood. Here, we identify protein phosphatase 1 (PP1) as regulator of period and light-induced resetting of the mammalian circadian clock. Down-regulation of PP1 activity in cells by RNA interference and in vivo by expression of a specific inhibitor in the brain of mice tended to lengthen circadian period. Moreover, reduction of PP1 activity in the brain altered light-mediated clock resetting behavior in mice, enhancing the phase shifts in either direction. At the molecular level, diminished PP1 activity increased nuclear accumulation of the clock component PER2 in neurons. Hence, PP1, may reduce PER2 phosphorylation thereby influencing nuclear localization of this protein. This may at least partially influence period and phase shifting properties of the mammalian circadian clock.
Collapse
|
28
|
Modeling the impact of mitochondrial DNA damage in forebrain neurons and beyond. Mech Ageing Dev 2011; 132:424-8. [PMID: 21354441 DOI: 10.1016/j.mad.2011.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 02/15/2011] [Accepted: 02/18/2011] [Indexed: 01/30/2023]
Abstract
We have generated an inducible transgenic mouse model, which expresses a mutated version of UNG1 (mutUNG1) that removes thymine, in addition to uracil from mitochondrial DNA. The abasic-sites (AP-sites) generated by removal of thymine or uracil are a threat to genomic integrity, and are particularly harmful in mitochondria due to inhibition of mitochondrial DNA polymerase. MutUNG1, accompanied by a luciferase reporter-gene, is controlled by the Tet-on system. Transgene expression is spatially regulated by the forebrain specific CaMKIIα-promoter, and temporally by the addition of doxycycline. Mice harboring this transgene develop compromised mitochondrial dynamics, neurodegeneration and impaired behavior.
Collapse
|
29
|
Atlas of transgenic Tet-Off Ca2+/calmodulin-dependent protein kinase II and prion protein promoter activity in the mouse brain. Neuroimage 2010; 54:2603-11. [PMID: 21093594 DOI: 10.1016/j.neuroimage.2010.11.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 10/31/2010] [Accepted: 11/08/2010] [Indexed: 01/20/2023] Open
Abstract
Conditional transgenic mouse models are important tools for investigations of neurodegenerative diseases and evaluation of potential therapeutic interventions. A popular conditional transgenic system is the binary tetracycline-responsive gene (Tet-Off) system, in which the expression of the gene of interest depends on a tetracycline-regulatable transactivator (tTA) under the control of a specific promoter construct. The most frequently used Tet-Off promoter mouse lines are the Ca(2+)/calmodulin-dependent protein kinase II (CamKII) and prion protein (PrP) promoter lines, respectively. To target the regulated gene of interest to relevant brain regions, a priori knowledge about the spatial distribution of the regulated gene expression in the brain is important. Such distribution patterns can be investigated using double transgenic mice in which the promoter construct regulates a LacZ reporter gene encoding the marker β-galactosidase which can be histologically detected using its substrate X-gal. We have previously published an atlas showing the brain-wide expression mediated by the Tet-Off PrP promoter mouse line, but the distribution of activity in the Tet-Off CamKII promoter mouse line is less well known. To compare promoter activity distributions in these two Tet-Off mouse lines, we have developed an online digital atlas tailored for side-by-side comparison of histological section images. The atlas provides a comprehensive list of brain regions containing X-gal labeling and an interactive dual image viewer tool for panning and zooming of corresponding section images. Comparison of spatial expression patterns between the two lines show considerable regional and cellular differences, relevant in context of generation and analysis of inducible models based on these two tetracycline responsive promoter mouse lines.
Collapse
|
30
|
Gräff J, Koshibu K, Jouvenceau A, Dutar P, Mansuy IM. Protein phosphatase 1-dependent transcriptional programs for long-term memory and plasticity. Learn Mem 2010; 17:355-63. [PMID: 20592054 DOI: 10.1101/lm.1766510] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Gene transcription is essential for the establishment and the maintenance of long-term memory (LTM) and for long-lasting forms of synaptic plasticity. The molecular mechanisms that control gene transcription in neuronal cells are complex and recruit multiple signaling pathways in the cytoplasm and the nucleus. Protein kinases (PKs) and phosphatases (PPs) are important players in these mechanisms. Protein serine/threonine phosphatase 1 (PP1), in particular, was recently shown to be important for transcription-dependent memory by regulating chromatin remodeling. However, the impact of PP1 on gene transcription in adult neurons remains not fully delineated. Here, we demonstrate that the nuclear pool of PP1 is associated with transcriptional events involving molecular components of signaling cascades acting as positive and negative regulators of memory and brain plasticity. The data show that inhibiting this pool selectively in forebrain neurons improves memory performance, enhances long-term potentiation (LTP), and modulates gene transcription. These findings highlight an important role for PP1 in the regulation of gene transcription in LTM and synaptic plasticity in the adult brain.
Collapse
Affiliation(s)
- Johannes Gräff
- Brain Research Institute, Medical Faculty of University Zürich and Department of Biology of Swiss Federal Institute of Technology, CH-8057 Zürich, Switzerland
| | | | | | | | | |
Collapse
|
31
|
Zhou H, Liu Y, He F, Mo L, Sun TT, Wu XR. Temporally and spatially controllable gene expression and knockout in mouse urothelium. Am J Physiol Renal Physiol 2010; 299:F387-95. [PMID: 20427471 DOI: 10.1152/ajprenal.00185.2010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Urothelium that lines almost the entire urinary tract performs important functions and is prone to assaults by urinary microbials, metabolites, and carcinogens. To improve our understanding of urothelial physiology and disease pathogenesis, we sought to develop two novel transgenic systems, one that would allow inducible and urothelium-specific gene expression, and another that would allow inducible and urothelium-specific knockout. Toward this end, we combined the ability of the mouse uroplakin II promoter (mUPII) to drive urothelium-specific gene expression with a versatile tetracycline-mediated inducible system. We found that, when constructed under the control of mUPII, only a modified, reverse tetracycline trans-activator (rtTA-M2), but not its original version (rtTA), could efficiently trans-activate reporter gene expression in mouse urothelium on doxycycline (Dox) induction. The mUPII/rtTA-M2-inducible system retained its strict urothelial specificity, had no background activity in the absence of Dox, and responded rapidly to Dox administration. Using a reporter gene whose expression was secondarily controlled by histone remodeling, we were able to identify, colocalize with 5-bromo-2-deoxyuridine incorporation, and semiquantify newly divided urothelial cells. Finally, we established that, when combined with a Cre recombinase under the control of the tetracycline operon, the mUPII-driven rtTA-M2 could inducibly inactivate any gene of interest in mouse urothelium. The establishment of these two new transgenic mouse systems enables the manipulation of gene expression and/or inactivation in adult mouse urothelium at any given time, thus minimizing potential compensatory effects due to gene overexpression or loss and allowing more accurate modeling of urothelial diseases than previously reported constitutive systems.
Collapse
Affiliation(s)
- Haiping Zhou
- Departments of Urology, New York Univ. School of Medicine, New York, 10010, USA
| | | | | | | | | | | |
Collapse
|
32
|
Mitochondrial DNA toxicity in forebrain neurons causes apoptosis, neurodegeneration, and impaired behavior. Mol Cell Biol 2010; 30:1357-67. [PMID: 20065039 DOI: 10.1128/mcb.01149-09] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial dysfunction underlying changes in neurodegenerative diseases is often associated with apoptosis and a progressive loss of neurons, and damage to the mitochondrial genome is proposed to be involved in such pathologies. In the present study we designed a mouse model that allows us to specifically induce mitochondrial DNA toxicity in the forebrain neurons of adult mice. This is achieved by CaMKIIalpha-regulated inducible expression of a mutated version of the mitochondrial UNG DNA repair enzyme (mutUNG1). This enzyme is capable of removing thymine from the mitochondrial genome. We demonstrate that a continual generation of apyrimidinic sites causes apoptosis and neuronal death. These defects are associated with behavioral alterations characterized by increased locomotor activity, impaired cognitive abilities, and lack of anxietylike responses. In summary, whereas mitochondrial base substitution and deletions previously have been shown to correlate with premature and natural aging, respectively, we show that a high level of apyrimidinic sites lead to mitochondrial DNA cytotoxicity, which causes apoptosis, followed by neurodegeneration.
Collapse
|
33
|
The power of reversibility regulating gene activities via tetracycline-controlled transcription. Methods Enzymol 2010; 477:429-53. [PMID: 20699154 DOI: 10.1016/s0076-6879(10)77022-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tetracycline-controlled transcriptional activation systems are widely used to control gene expression in transgenic animals in a truly conditional manner. By this we refer to the capability of these expression systems to control gene activities not only in a tissue specific and temporal defined but also reversible manner. This versatility has made the Tet regulatory systems to a preeminent tool in reverse mouse genetics. The development of the technology in the past 15 years will be reviewed and guidelines will be given for its implementation in creating transgenic rodents. Finally, we highlight some recent exciting applications of the Tet technology as well as its foreseeable combination with other emerging technologies in mouse transgenesis.
Collapse
|
34
|
Abstract
Chromatin remodeling through histone posttranslational modifications (PTMs) and DNA methylation has recently been implicated in cognitive functions, but the mechanisms involved in such epigenetic regulation remain poorly understood. Here, we show that protein phosphatase 1 (PP1) is a critical regulator of chromatin remodeling in the mammalian brain that controls histone PTMs and gene transcription associated with long-term memory. Our data show that PP1 is present at the chromatin in brain cells and interacts with enzymes of the epigenetic machinery including HDAC1 (histone deacetylase 1) and histone demethylase JMJD2A (jumonji domain-containing protein 2A). The selective inhibition of the nuclear pool of PP1 in forebrain neurons in transgenic mice is shown to induce several histone PTMs that include not only phosphorylation but also acetylation and methylation. These PTMs are residue-specific and occur at the promoter of genes important for memory formation like CREB (cAMP response element-binding protein) and NF-kappaB (nuclear factor-kappaB). These histone PTMs further co-occur with selective binding of RNA polymerase II and altered gene transcription, and are associated with improved long-term memory for objects and space. Together, these findings reveal a novel mechanism for the epigenetic control of gene transcription and long-term memory in the adult brain that depends on PP1.
Collapse
|
35
|
NCS-1 in the dentate gyrus promotes exploration, synaptic plasticity, and rapid acquisition of spatial memory. Neuron 2009; 63:643-56. [PMID: 19755107 DOI: 10.1016/j.neuron.2009.08.014] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Revised: 08/21/2009] [Accepted: 08/21/2009] [Indexed: 11/24/2022]
Abstract
The molecular underpinnings of exploration and its link to learning and memory remain poorly understood. Here we show that inducible, modest overexpression of neuronal calcium sensor 1 (Ncs1) selectively in the adult murine dentate gyrus (DG) promotes a specific form of exploratory behavior. The mice also display a selective facilitation of long-term potentiation (LTP) in the medial perforant path and a selective enhancement in rapid-acquisition spatial memory, phenotypes that are reversed by direct application of a cell-permeant peptide (DNIP) designed to interfere with NCS-1 binding to the dopamine type-2 receptor (D2R). Moreover, the DNIP and the D2R-selective antagonist L-741,626 attenuated exploratory behavior, DG LTP, and spatial memory in control mice. These data demonstrate a role for NCS-1 and D2R in DG plasticity and provide insight for understanding how the DG contributes to the origin of exploration and spatial memory acquisition.
Collapse
|
36
|
Baumgärtel K, Tweedie-Cullen RY, Grossmann J, Gehrig P, Livingstone-Zatchej M, Mansuy IM. Changes in the proteome after neuronal zif268 overexpression. J Proteome Res 2009; 8:3298-316. [PMID: 19374395 DOI: 10.1021/pr801000r] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Long-lasting forms of brain plasticity are a cellular basis for long-term memory, and their disturbance underlies pathological conditions such as dementia and cognitive impairment. Neuronal plasticity is a complex process that utilizes molecular cascades in the cytoplasm and the nucleus and involves numerous transcription factors, in particular, immediate early genes (IEGs). The signaling cascades that control IEGs are fairly well described, but the downstream transcriptional response is poorly understood, especially its late components. Here, we investigated the response induced by the IEG Zif268 in the adult brain in relation to long-term memory. Using a mouse model with increased neuronal expression of Zif268 that leads to improved memory, we identified an ensemble of proteins regulated by Zif268 expression and differentiated between direct and indirect targets based on the presence of a consensus binding motif in their promoter. We show that Zif268 regulates numerous substrates with diverse biological functions including protein modification and degradation (proteasome-core complex), phosphorylation, cell division, sensory perception, metabolism, and metal ion transport. The results provide a comprehensive and quantitative data set characterizing the Zif268-dependent proteome in the adult mouse brain and offers biologically important new insight into activity-dependent pathways downstream of IEGs.
Collapse
Affiliation(s)
- Karsten Baumgärtel
- Brain Research Institute, Medical Faculty of the University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
37
|
Konopka W, Duniec K, Klejman A, Wawrzyniak M, Owczarek D, Gawrys L, Maleszewski M, Mallet J, Kaczmarek L. Tet system in the brain: Transgenic rats and lentiviral vectors approach. Genesis 2009; 47:274-80. [DOI: 10.1002/dvg.20487] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
38
|
Czöndör K, Ellwanger K, Fuchs YF, Lutz S, Gulyás M, Mansuy IM, Hausser A, Pfizenmaier K, Schlett K. Protein kinase D controls the integrity of Golgi apparatus and the maintenance of dendritic arborization in hippocampal neurons. Mol Biol Cell 2009; 20:2108-20. [PMID: 19211839 DOI: 10.1091/mbc.e08-09-0957] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Protein kinase D (PKD) is known to participate in various cellular functions, including secretory vesicle fission from the Golgi and plasma membrane-directed transport. Here, we report on expression and function of PKD in hippocampal neurons. Expression of an enhanced green fluorescent protein (EGFP)-tagged PKD activity reporter in mouse embryonal hippocampal neurons revealed high endogenous PKD activity at the Golgi complex and in the dendrites, whereas PKD activity was excluded from the axon in parallel with axonal maturation. Expression of fluorescently tagged wild-type PKD1 and constitutively active PKD1(S738/742E) (caPKD1) in neurons revealed that both proteins were slightly enriched at the trans-Golgi network (TGN) and did not interfere with its thread-like morphology. By contrast, expression of dominant-negative kinase inactive PKD1(K612W) (kdPKD1) led to the disruption of the neuronal Golgi complex, with kdPKD1 strongly localized to the TGN fragments. Similar findings were obtained from transgenic mice with inducible, neuron-specific expression of kdPKD1-EGFP. As a prominent consequence of kdPKD1 expression, the dendritic tree of transfected neurons was reduced, whereas caPKD1 increased dendritic arborization. Our results thus provide direct evidence that PKD activity is selectively involved in the maintenance of dendritic arborization and Golgi structure of hippocampal neurons.
Collapse
Affiliation(s)
- Katalin Czöndör
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary H-1117
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chiu C, Reid CA, Tan HO, Davies PJ, Single FN, Koukoulas I, Berkovic SF, Tan SS, Sprengel R, Jones MV, Petrou S. Developmental impact of a familial GABAA receptor epilepsy mutation. Ann Neurol 2008; 64:284-93. [PMID: 18825662 DOI: 10.1002/ana.21440] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE A major goal of epilepsy research is to understand the molecular and functional basis of seizure genesis. A human GABA(A) gamma2 gene mutation (R43Q) is associated with generalized epilepsy. Introduction of this mutation into a mouse by gene targeting recapitulates the human phenotype demonstrating a strong genotype to phenotype link. GABA(A) receptors play a role in the moment-to-moment control of brain function and also on the long-term wiring of the brain by directing neuronal development. Our objective was to determine whether developmental expression of the mutation alters seizure susceptibility later in life. METHODS A tetracycline-based conditional model for activation of a hypomorphic Q43 disease allele was created and validated. Seizure susceptibility was assessed using the subcutaneous pentylenetetrazole model. RESULTS Seizure susceptibility was significantly reduced in mice where the Q43 allele was suppressed during development. INTERPRETATION These results demonstrate that a human epilepsy-causing mutation impacts network stability during a critical developmental period. These data suggest that identification of presymptomatic children may provide a window for therapeutic intervention before overt symptoms are observed, potentially altering the course of epileptogenesis.
Collapse
Affiliation(s)
- Cindy Chiu
- Howard Florey Institute, The University of Melbourne, Parkville, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Havekes R, Nijholt IM, Visser AKD, Eisel ULM, Van der Zee EA. Transgenic inhibition of neuronal calcineurin activity in the forebrain facilitates fear conditioning, but inhibits the extinction of contextual fear memories. Neurobiol Learn Mem 2008; 89:595-8. [PMID: 17884610 DOI: 10.1016/j.nlm.2007.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 08/04/2007] [Accepted: 08/06/2007] [Indexed: 11/16/2022]
Abstract
It is unclear whether protein phosphatases, which counteract the actions of protein kinases, play a beneficial role in the formation and extinction of previously acquired fear memories. In this study, we investigated the role of the calcium/calmodulin dependent phosphatase 2B, also known as calcineurin (CaN) in the formation of contextual fear memory and extinction of previously acquired contextual fear. We used a temporally regulated transgenic approach, that allowed us to selectively inhibit neuronal CaN activity in the forebrain either during conditioning or only during extinction training leaving the conditioning undisturbed. Reducing CaN activity through the expression of a CaN inhibitor facilitated contextual fear conditioning, while it impaired the extinction of previously formed contextual fear memory. These findings give the first genetic evidence that neuronal CaN plays an opposite role in the formation of contextual fear memories and the extinction of previously formed contextual fear memories.
Collapse
Affiliation(s)
- Robbert Havekes
- Department of Molecular Neurobiology, University of Groningen, P.O. Box 14, Kerklaan 30 9750 AA, Haren, The Netherlands.
| | | | | | | | | |
Collapse
|
41
|
Control of the establishment of aversive memory by calcineurin and Zif268. Nat Neurosci 2008; 11:572-8. [PMID: 18425121 DOI: 10.1038/nn.2113] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 03/24/2008] [Indexed: 11/08/2022]
Abstract
Emotional memory is a rapidly acquired and persistent form of memory, and its robustness is in part determined by the initial strength of the memory. Here, we provide new evidence that the protein phosphatase calcineurin (CaN), a potent negative regulator of neuronal signaling that is known to constrain learning and memory, critically regulates the establishment of emotional memory through mechanisms involving the immediate early gene Zif268 (also known as Egr1). We found that CaN is inhibited in the amygdala during the establishment of aversive memory, but Zif268 is activated. Using inducible transgenesis in mice, we further saw that CaN inhibition and Zif268 overexpression during memory establishment strengthen the memory trace and enhance its resistance to extinction. We found that CaN inhibition correlates with increased Zif268 expression and that a common pool of proteins is regulated in the amygdala after CaN inhibition and Zif268 overexpression. Together, these findings reveal a previously unknown mechanism for the control of emotional memory that depends on CaN and Zif268.
Collapse
|
42
|
Protein phosphatase 1-dependent bidirectional synaptic plasticity controls ischemic recovery in the adult brain. J Neurosci 2008; 28:154-62. [PMID: 18171933 DOI: 10.1523/jneurosci.4109-07.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Protein kinases and phosphatases can alter the impact of excitotoxicity resulting from ischemia by concurrently modulating apoptotic/survival pathways. Here, we show that protein phosphatase 1 (PP1), known to constrain neuronal signaling and synaptic strength (Mansuy et al., 1998; Morishita et al., 2001), critically regulates neuroprotective pathways in the adult brain. When PP1 is inhibited pharmacologically or genetically, recovery from oxygen/glucose deprivation (OGD) in vitro, or ischemia in vivo is impaired. Furthermore, in vitro, inducing LTP shortly before OGD similarly impairs recovery, an effect that correlates with strong PP1 inhibition. Conversely, inducing LTD before OGD elicits full recovery by preserving PP1 activity, an effect that is abolished by PP1 inhibition. The mechanisms of action of PP1 appear to be coupled with several components of apoptotic pathways, in particular ERK1/2 (extracellular signal-regulated kinase 1/2) whose activation is increased by PP1 inhibition both in vitro and in vivo. Together, these results reveal that the mechanisms of recovery in the adult brain critically involve PP1, and highlight a novel physiological function for long-term potentiation and long-term depression in the control of brain damage and repair.
Collapse
|
43
|
Magnuson MA, Burlison JS. Caveats and considerations for performing pancreas-specific gene manipulations in the mouse. Diabetes Obes Metab 2007; 9 Suppl 2:5-13. [PMID: 17919173 DOI: 10.1111/j.1463-1326.2007.00771.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conditional gene targeting using the Cre/loxP strategy has proven to be very useful for studies of glucose homeostasis, tissue function and dysfunction in diabetes, and pancreas development. However, use of this strategy over the past decade has revealed a variety of experimental caveats, many of which are a direct consequence of the procedures used to generate Cre-driver lines. We discuss frequently encountered experimental artefacts, the advantages of using bacterial artificial chromosome-derived transgenes or performing a Cre knockin for improving the specificity of expression, and systems for regulating Cre activity. In addition, recent studies indicate that high amounts of Cre in the pancreatic beta-cell may cause glucose intolerance and impaired insulin secretion. However, these findings, while serving as a reminder for simple experimental controls, are unlikely to diminish utilization of this very powerful and useful technology.
Collapse
Affiliation(s)
- M A Magnuson
- Department of Molecular Physiology, Biophysics Vanderbilt University School of Medicine, Center for Stem Cell Biology, Nashville, TN 37232-0255, USA.
| | | |
Collapse
|
44
|
Katsantoni EZ, Anghelescu NE, Rottier R, Moerland M, Antoniou M, de Crom R, Grosveld F, Strouboulis J. Ubiquitous expression of the rtTA2S-M2 inducible system in transgenic mice driven by the human hnRNPA2B1/CBX3 CpG island. BMC DEVELOPMENTAL BIOLOGY 2007; 7:108. [PMID: 17900353 PMCID: PMC2080639 DOI: 10.1186/1471-213x-7-108] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 09/27/2007] [Indexed: 11/30/2022]
Abstract
Background A sensitive, ubiquitously expressed tetracycline inducible system would be a valuable tool in mouse transgenesis. However, this has been difficult to obtain due to position effects observed at different chromosomal sites of transgene integration, which negatively affect expression in many tissues. The aim of this study was to test the utility of a mammalian methylation-free CpG island to drive ubiquitous expression of the sensitive doxycycline (Dox) inducible rtTA2S-M2 Tet-transactivator in transgenic mice. Results An 8 kb genomic fragment from the methylation-free CpG island of the human hnRNPA2B1-CBX3 housekeeping gene locus was tested. In a number of transgenic mouse lines obtained, rtTA2S-M2 expression was detected in many tissues examined. Characterisation of the highest expressing rtTA2S-M2 transgenic mouse line demonstrated Dox-inducible GFP transgene expression in many tissues. Using this line we also show highly sensitive quantitative induction with low doses of Dox of an assayable plasma protein transgene under the control of a Tet Responsive Element (TRE). The utility of this rtTA2S-M2 line for inducible expression in mouse embryos was also demonstrated using a GATA-6 Tet-inducible transgene to show specific phenotypes in the embryonic lung, as well as broader effects resulting from the inducible widespread overexpression of the transgene. Conclusion The ubiquitously expressing rtTA2S-M2 transgenic mouse line described here provides a very useful tool for studying the effects of the widespread, inducible overexpression of genes during embryonic development and in adult mice.
Collapse
Affiliation(s)
- Eleni Z Katsantoni
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Hematology Division, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 115 27 Athens, Greece
| | - Nora E Anghelescu
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Gene Controls Mechanism and Disease, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Robbert Rottier
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Matthijs Moerland
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Michael Antoniou
- Nuclear Biology Group, Division of Medical and Molecular Genetics, GKT School of Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Rini de Crom
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Frank Grosveld
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - John Strouboulis
- Department of Cell Biology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Institute of Molecular Oncology, BSRC "Alexander Fleming", PO Box 74145, 166 02 Varkiza, Greece
| |
Collapse
|
45
|
Knobloch M, Farinelli M, Konietzko U, Nitsch RM, Mansuy IM. Abeta oligomer-mediated long-term potentiation impairment involves protein phosphatase 1-dependent mechanisms. J Neurosci 2007; 27:7648-53. [PMID: 17634359 PMCID: PMC6672892 DOI: 10.1523/jneurosci.0395-07.2007] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Amyloid beta (Abeta) oligomers are derived from proteolytic cleavage of amyloid precursor protein (APP) and can impair memory and hippocampal long-term potentiation (LTP) in vivo and in vitro. They are recognized as the primary neurotoxic agents in Alzheimer's disease. The mechanisms underlying such toxicity on synaptic functions are complex and not fully understood. Here, we provide the first evidence that these mechanisms involve protein phosphatase 1 (PP1). Using a novel transgenic mouse model expressing human APP with the Swedish and Arctic mutations that render Abeta more prone to form oligomers (arcAbeta mice), we show that the LTP impairment induced by Abeta oligomers can be fully reversed by PP1 inhibition in vitro. We further demonstrate that the genetic inhibition of endogenous PP1 in vivo confers resistance to Abeta oligomer-mediated toxicity and preserves LTP. Overall, these results reveal that PP1 is a key player in the mechanisms of AD pathology.
Collapse
Affiliation(s)
- Marlen Knobloch
- Division of Psychiatry Research, University of Zurich, 8008 Zurich, Switzerland, and
| | - Mélissa Farinelli
- Brain Research Institute, University of Zurich and Department of Biology, Swiss Federal Institute of Technology, 8057 Zurich, Switzerland
| | - Uwe Konietzko
- Division of Psychiatry Research, University of Zurich, 8008 Zurich, Switzerland, and
| | - Roger M. Nitsch
- Division of Psychiatry Research, University of Zurich, 8008 Zurich, Switzerland, and
| | - Isabelle M. Mansuy
- Brain Research Institute, University of Zurich and Department of Biology, Swiss Federal Institute of Technology, 8057 Zurich, Switzerland
| |
Collapse
|
46
|
Zhu P, Aller MI, Baron U, Cambridge S, Bausen M, Herb J, Sawinski J, Cetin A, Osten P, Nelson ML, Kügler S, Seeburg PH, Sprengel R, Hasan MT. Silencing and un-silencing of tetracycline-controlled genes in neurons. PLoS One 2007; 2:e533. [PMID: 17579707 PMCID: PMC1888723 DOI: 10.1371/journal.pone.0000533] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 05/14/2007] [Indexed: 11/19/2022] Open
Abstract
To identify the underlying reason for the controversial performance of tetracycline (Tet)-controlled regulated gene expression in mammalian neurons, we investigated each of the three components that comprise the Tet inducible systems, namely tetracyclines as inducers, tetracycline-transactivator (tTA) and reverse tTA (rtTA), and tTA-responsive promoters (Ptets). We have discovered that stably integrated Ptet becomes functionally silenced in the majority of neurons when it is inactive during development. Ptet silencing can be avoided when it is either not integrated in the genome or stably-integrated with basal activity. Moreover, long-term, high transactivator levels in neurons can often overcome integration-induced Ptet gene silencing, possibly by inducing promoter accessibility.
Collapse
Affiliation(s)
- Peixin Zhu
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - M. Isabel Aller
- Department of Clinical Neurobiology, University of Heidelberg, Heidelberg, Germany
| | | | - Sidney Cambridge
- Max Planck Institute of Neurobiology, Munich-Martinsried, Germany
| | - Melanie Bausen
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Jan Herb
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Jürgen Sawinski
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Ali Cetin
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Pavel Osten
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Mark L. Nelson
- Paratek Pharmaceuticals Inc., Boston, Massachusetts, United States of America
| | - Sebastian Kügler
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | | | - Rolf Sprengel
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Mazahir T. Hasan
- Max Planck Institute for Medical Research, Heidelberg, Germany
- Max Planck Institute of Neurobiology, Munich-Martinsried, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
47
|
Moerland M, Anghelescu N, Samyn H, van Haperen R, van Gent T, Strouboulis J, van Tol A, Grosveld F, de Crom R. Inducible expression of phospholipid transfer protein (PLTP) in transgenic mice: acute effects of PLTP on lipoprotein metabolism. Transgenic Res 2007; 16:503-13. [PMID: 17437182 DOI: 10.1007/s11248-007-9094-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 03/19/2007] [Indexed: 10/23/2022]
Abstract
One main determinant in high-density lipoprotein (HDL) metabolism is phospholipid transfer protein (PLTP), a plasma protein that is associated with HDL. In transgenic mice overexpressing human PLTP we found that elevated plasma PLTP levels dose-dependently increased the susceptibility to diet-induced atherosclerosis. This could be mainly due to the fact that most functions of PLTP are potentially atherogenic, such as decreasing plasma HDL levels. To further elucidate the role of PLTP in lipoprotein metabolism and atherosclerosis we generated a novel transgenic mouse model that allows conditional expression of human PLTP. In this mouse model a human PLTP encoding sequence is controlled by a Tet-On system. Upon induction of PLTP expression, our mouse model showed a strongly increased PLTP activity (from 3.0 +/- 0.6 to 11.4 +/- 2.8 AU, p < 0.001). The increase in PLTP activity resulted in an acute decrease in plasma cholesterol of 33% and a comparable decrease in phospholipids. The decrease in total plasma cholesterol and phospholipids was caused by a 35% decrease in HDL-cholesterol level and a 41% decrease in HDL-phospholipid level. These results demonstrate the feasibility of our mouse model to induce an acute elevation of PLTP activity, which is easily reversible. As a direct consequence of an increase in PLTP activity, HDL-cholesterol and HDL-phospholipid levels strongly decrease. Using this mouse model, it will be possible to study the effects of acute elevation of PLTP activity on lipoprotein metabolism and pre-existing atherosclerosis.
Collapse
Affiliation(s)
- Matthijs Moerland
- Department of Cell Biology and Genetics, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sun Y, Chen X, Xiao D. Tetracycline-inducible expression systems: new strategies and practices in the transgenic mouse modeling. Acta Biochim Biophys Sin (Shanghai) 2007; 39:235-46. [PMID: 17417678 DOI: 10.1111/j.1745-7270.2007.00258.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
To accurately analyze the function of transgene(s) of interest in transgenic mice, and to generate credible transgenic animal models for multifarious human diseases to precisely mimic human disease states, it is critical to tightly regulate gene expression in the animals in a conditional manner. The ability to turn gene expression on or off in the restricted cells or tissues at specific time permits unprecedented flexibility in dissecting gene functions in health and disease. Pioneering studies in conditional transgene expression have brought about the development of a wide variety of controlled gene expression systems, which meet this criterion. Among them, the tetracycline-controlled expression systems (e.g. Tet-off system and Tet-on system) have been used extensively in vitro and in vivo. In recent years, some strategies derived from tetracycline-inducible system alone, as well as the combined use of Tet-based systems and Cre/lox P switching gene expression system, have been newly developed to allow more flexibility for exploring gene functions in health and disease, and produce credible transgenic animal models for various human diseases. In this review these newly developed strategies are discussed.
Collapse
Affiliation(s)
- Yan Sun
- Center of Experimental Animals, Sun Yat-Sen University, Guangzhou 510080, China
| | | | | |
Collapse
|
49
|
Baumgärtel K, Fernández C, Johansson T, Mansuy IM. Conditional transgenesis and recombination to study the molecular mechanisms of brain plasticity and memory. Handb Exp Pharmacol 2007:315-45. [PMID: 17203661 DOI: 10.1007/978-3-540-35109-2_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In the postgenomic era, a primary focus of mouse genetics is to elucidate the role of individual genes in vivo. However, in the nervous system, studying the contribution of specific genes to brain functions is difficult because the brain is a highly complex organ with multiple neuroanatomical structures, orchestrating virtually every function in the body. Further, higher-order brain functions such as learning and memory simultaneously recruit several signaling cascades in different subcellular compartments and have highly fine-tuned spatial and temporal components. Conditional transgenic and gene targeting methodologies, however, now offer valuable tools with improved spatial and temporal resolution for appropriate studies of these functions. This chapter provides an overview of these tools and describes how they have helped gain better understanding of the role of candidate genes such as the NMDA receptor, the protein kinase CaMKIIIalpha, the protein phosphatases calcineurin and PP1, or the transcription factor CREB, in the processes of learning and memory. This review illustrates the broad and innovative applicability of these methodologies to the study of brain plasticity and cognitive functions.
Collapse
Affiliation(s)
- K Baumgärtel
- Department of Biology, Swiss Federal Institute of Technology, Medical Faculty, University Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | | | | | | |
Collapse
|
50
|
Berens C, Lochner S, Löber S, Usai I, Schmidt A, Drueppel L, Hillen W, Gmeiner P. Subtype selective tetracycline agonists and their application for a two-stage regulatory system. Chembiochem 2006; 7:1320-4. [PMID: 16871602 DOI: 10.1002/cbic.200600226] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Christian Berens
- Department of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstrasse 5, 91058 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|