1
|
Hadi F, Mortaja M, Hadi Z. Calcium (Ca 2+) hemostasis, mitochondria, autophagy, and mitophagy contribute to Alzheimer's disease as early moderators. Cell Biochem Funct 2024; 42:e4085. [PMID: 38951992 DOI: 10.1002/cbf.4085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
This review rigorously investigates the early cerebral changes associated with Alzheimer's disease, which manifest long before clinical symptoms arise. It presents evidence that the dysregulation of calcium (Ca2+) homeostasis, along with mitochondrial dysfunction and aberrant autophagic processes, may drive the disease's progression during its asymptomatic, preclinical stage. Understanding the intricate molecular interplay that unfolds during this critical period offers a window into identifying novel therapeutic targets, thereby advancing the treatment of neurodegenerative disorders. The review delves into both established and emerging insights into the molecular alterations precipitated by the disruption of Ca2+ balance, setting the stage for cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Fatemeh Hadi
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Mahsa Mortaja
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California, USA
| | - Zahra Hadi
- Department of Chemistry, Faculty of Physics and Chemistry, Alzahra University, Tehran, Iran
| |
Collapse
|
2
|
Desbois M, Opperman KJ, Amezquita J, Gaglio G, Crawley O, Grill B. Ubiquitin ligase activity inhibits Cdk5 to control axon termination. PLoS Genet 2022; 18:e1010152. [PMID: 35421092 PMCID: PMC9041834 DOI: 10.1371/journal.pgen.1010152] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/26/2022] [Accepted: 03/17/2022] [Indexed: 01/29/2023] Open
Abstract
The Cdk5 kinase plays prominent roles in nervous system development, plasticity, behavior and disease. It also has important, non-neuronal functions in cancer, the immune system and insulin secretion. At present, we do not fully understand negative regulatory mechanisms that restrict Cdk5. Here, we use Caenorhabditis elegans to show that CDK-5 is inhibited by the RPM-1/FSN-1 ubiquitin ligase complex. This atypical RING ubiquitin ligase is conserved from C. elegans through mammals. Our finding originated from unbiased, in vivo affinity purification proteomics, which identified CDK-5 as a putative RPM-1 substrate. CRISPR-based, native biochemistry showed that CDK-5 interacts with the RPM-1/FSN-1 ubiquitin ligase complex. A CRISPR engineered RPM-1 substrate ‘trap’ enriched CDK-5 binding, which was mediated by the FSN-1 substrate recognition module. To test the functional genetic relationship between the RPM-1/FSN-1 ubiquitin ligase complex and CDK-5, we evaluated axon termination in mechanosensory neurons and motor neurons. Our results indicate that RPM-1/FSN-1 ubiquitin ligase activity restricts CDK-5 to control axon termination. Collectively, these proteomic, biochemical and genetic results increase our understanding of mechanisms that restrain Cdk5 in the nervous system. Cdk5 is an atypical cyclin dependent kinase and an important player in nervous system development, plasticity, and disease. Decades of research has focused on understanding how Cdk5 is activated. In contrast, we know much less about the genetic and molecular mechanisms that restrict Cdk5 activity. Here, we examined how Cdk5 is inhibited in the nervous system using the model organism C. elegans. Our results indicate that the RPM-1/FSN-1 E3 ubiquitin ligase complex inhibits Cdk5 to control termination of axon growth. Our finding that ubiquitin ligase activity restricts Cdk5 in the nervous system in vivo now opens up the interesting possibility that ubiquitin ligase activity might regulate Cdk5 in other cellular contexts and disease settings.
Collapse
Affiliation(s)
- Muriel Desbois
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Karla J. Opperman
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Jonathan Amezquita
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, United States of America
| | - Gabriel Gaglio
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Oliver Crawley
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington M1-A303/305 Behnke Conference Room, Arnold building, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
3
|
Taccola C, Barneoud P, Cartot-Cotton S, Valente D, Schussler N, Saubaméa B, Chasseigneaux S, Cochois V, Mignon V, Curis E, Lochus M, Nicolic S, Dodacki A, Cisternino S, Declèves X, Bourasset F. Modifications of physical and functional integrity of the blood-brain barrier in an inducible mouse model of neurodegeneration. Neuropharmacology 2021; 191:108588. [PMID: 33940010 DOI: 10.1016/j.neuropharm.2021.108588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
The inducible p25 overexpression mouse model recapitulate many hallmark features of Alzheimer's disase including progressive neuronal loss, elevated Aβ, tau pathology, cognitive dysfunction, and impaired synaptic plasticity. We chose p25 mice to evaluate the physical and functional integrity of the blood-brain barrier (BBB) in a context of Tau pathology (pTau) and severe neurodegeneration, at an early (3 weeks ON) and a late (6 weeks ON) stage of the pathology. Using in situ brain perfusion and confocal imaging, we found that the brain vascular surface area and the physical integrity of the BBB were unaltered in p25 mice. However, there was a significant 14% decrease in cerebrovascular volume in 6 weeks ON mice, possibly explained by a significant 27% increase of collagen IV in the basement membrane of brain capillaries. The function of the BBB transporters GLUT1 and LAT1 was evaluated by measuring brain uptake of d-glucose and phenylalanine, respectively. In 6 weeks ON p25 mice, d-glucose brain uptake was significantly reduced by about 17% compared with WT, without any change in the levels of GLUT1 protein or mRNA in brain capillaries. The brain uptake of phenylalanine was not significantly reduced in p25 mice compared with WT. Lack of BBB integrity, impaired BBB d-glucose transport have been observed in several mouse models of AD. In contrast, reduced cerebrovascular volume and an increased basement membrane thickness may be more specifically associated with pTau in mouse models of neurodegeneration.
Collapse
Affiliation(s)
- Camille Taccola
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine & Early Development, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France; INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Pascal Barneoud
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, 1 Avenue Pierre Brossolette, 91380, Chilly-Mazarin, France
| | - Sylvaine Cartot-Cotton
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine & Early Development, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France
| | - Delphine Valente
- Drug Metabolism & Pharmacokinetics, Research platform, Sanofi, 3 Digue d'Alfortville, 94140, Alfortville, France
| | - Nathalie Schussler
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, 1 Avenue Pierre Brossolette, 91380, Chilly-Mazarin, France
| | - Bruno Saubaméa
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Stéphanie Chasseigneaux
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Véronique Cochois
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Virginie Mignon
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Emmanuel Curis
- Laboratoire de biomathématiques, plateau iB(2), EA 7537 « BioSTM », UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France; Service de bioinformatique et statistique médicale, hôpital Saint-Louis, APHP, 1, avenue Claude Vellefaux, 75010, Paris, France
| | - Murielle Lochus
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Sophie Nicolic
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Agnès Dodacki
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Salvatore Cisternino
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Xavier Declèves
- INSERM UMR-S 1144, UFR de Pharmacie, Faculté de Santé, Université de Paris, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Fanchon Bourasset
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive, Université Bourgogne Franche-Comté, 19 rue Ambroise Paré, 25000, Besançon, France.
| |
Collapse
|
4
|
Gao G, He J, Luo Y, Sun Y, Zhou Y, Zhang J, Xing Y, Dai J. Axonopathy Likely Initiates Neuropathological Processes Via a Mechanism of Axonal Leakage in Alzheimer's Mouse Models. Curr Mol Med 2020; 19:183-195. [PMID: 30961496 DOI: 10.2174/1566524019666190405174908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND The formation of hyperphosphorylated tau and the production of β-amyloid are thought to be critical steps contributing to the pathological mechanisms in Alzheimer's disease (AD). However, there has been a long-lasting debate over their importance in the onset of AD. Recent studies have demonstrated that axonopathy is considered as an early neuropathological change of AD. However, the exact relationship between the development of axonopathy and the classic neuropathological changes such as senile plaques (SPs) and neurofibrillary tangles (NFTs) is unclear. OBJECTIVE The aim of this study was to investigate whether the formation of SPs and NFTs is associated with the development of axonal leakage. METHOD AND RESULTS Here we show that the formation and development of axonal leakage - a novel axonopathy is an age-dependent process, accompanied by swellings of axons and varicosities and associated with chronic oxidative stress induced by thiamine deficient (TD) diet in Kunming mice. In an APP/PS1 transgenic mouse model of AD, axonal leakage appears at 3 months, becomes more obvious at 6 months and severe, beyond 1 year. We also show that slight axonal leakage is related to the formation of hyperphosphorylated tau, but not plaques, and that only severe axonal leakage accompanied by the extensive swollen axons and varicosities, and overproduction of β-amyloid leads to the formation of SPs and hyperphosphorylated tau. CONCLUSION These data provide an explanation of the common origin and development of SPs and NFTs, and suggest that axonal leakage might be a key event in the development of the neuropathological processes in AD.
Collapse
Affiliation(s)
- Ge Gao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing He
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China.,Clinical Laboratory of the Second Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Luo
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China.,Clinical Laboratory of Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China.,The College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China
| | - Yanping Zhou
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China.,Department of Pathophysiology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Junxia Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Department of Anatomy, Histology and Embryology, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Ying Xing
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China.,The College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China
| |
Collapse
|
5
|
Wang X, Fei F, Qu J, Li C, Li Y, Zhang S. The role of septin 7 in physiology and pathological disease: A systematic review of current status. J Cell Mol Med 2018; 22:3298-3307. [PMID: 29602250 PMCID: PMC6010854 DOI: 10.1111/jcmm.13623] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/05/2018] [Indexed: 12/22/2022] Open
Abstract
Septins are a conserved family of cytoskeletal GTPases present in different organisms, including yeast, drosophila, Caenorhabditis elegans and humans. In humans, septins are involved in various cellular processes, including exocytosis, apoptosis, leukemogenesis, carcinogenesis and neurodegeneration. Septin 7 is unique out of 13 human septins. Mammalian septin 6, septin 7, septin 2 and septin 9 coisolate together in complexes to form the core unit for the generation of the septin filaments. Physiological septin filaments are hetero-oligomeric complexes consisting of core septin hexamers and octamers. Furthermore, septin 7 plays a crucial role in cytokinesis and mitosis. Septin 7 is localized to the filopodia and branches of developing hippocampal neurons, and is the most abundant septin in the adult rat forebrain as well as a structural component of the human and mouse sperm annuli. Septin 7 is crucial to the spine morphogenesis and dendrite growth in neurons, and is also a structural constituent of the annulus in human and mouse sperm. It can suppress growth of some tumours such as glioma and papillary thyroid carcinoma. However, the molecular mechanisms of involvement of septin 7 in human disease, especially in the development of cancer, remain unclear. This review focuses on the structure, function and mechanism of septin 7 in vivo, and summarizes the role of septin 7 in cell proliferation, cytokinesis, nervous and reproductive systems, as well as the underlying molecular events linking septin 7 to various diseases, such as Alzheimer's disease, schizophrenia, neuropsychiatric systemic lupus erythematosus, tumour and so on.
Collapse
Affiliation(s)
- Xinlu Wang
- Graduate SchoolTianjin University of Traditional Chinese MedicineTianjinChina
- Department of PathologyTianjin Union Medical CenterTianjinChina
| | - Fei Fei
- Department of PathologyTianjin Union Medical CenterTianjinChina
- Nankai University School of MedicineNankai UniversityTianjinChina
| | - Jie Qu
- Department of PathologyTianjin Union Medical CenterTianjinChina
- Nankai University School of MedicineNankai UniversityTianjinChina
| | - Chunyuan Li
- Department of PathologyTianjin Union Medical CenterTianjinChina
- Nankai University School of MedicineNankai UniversityTianjinChina
| | - Yuwei Li
- Department of Colorectal SurgeryTianjin Union Medical CenterTianjinChina
| | - Shiwu Zhang
- Department of PathologyTianjin Union Medical CenterTianjinChina
| |
Collapse
|
6
|
Uddin MS, Stachowiak A, Mamun AA, Tzvetkov NT, Takeda S, Atanasov AG, Bergantin LB, Abdel-Daim MM, Stankiewicz AM. Autophagy and Alzheimer's Disease: From Molecular Mechanisms to Therapeutic Implications. Front Aging Neurosci 2018; 10:04. [PMID: 29441009 PMCID: PMC5797541 DOI: 10.3389/fnagi.2018.00004] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/08/2018] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of progressive dementia in the elderly. It is characterized by a progressive and irreversible loss of cognitive abilities and formation of senile plaques, composed mainly of amyloid β (Aβ), and neurofibrillary tangles (NFTs), composed of tau protein, in the hippocampus and cortex of afflicted humans. In brains of AD patients the metabolism of Aβ is dysregulated, which leads to the accumulation and aggregation of Aβ. Metabolism of Aβ and tau proteins is crucially influenced by autophagy. Autophagy is a lysosome-dependent, homeostatic process, in which organelles and proteins are degraded and recycled into energy. Thus, dysfunction of autophagy is suggested to lead to the accretion of noxious proteins in the AD brain. In the present review, we describe the process of autophagy and its importance in AD. Additionally, we discuss mechanisms and genes linking autophagy and AD, i.e., the mTOR pathway, neuroinflammation, endocannabinoid system, ATG7, BCL2, BECN1, CDK5, CLU, CTSD, FOXO1, GFAP, ITPR1, MAPT, PSEN1, SNCA, UBQLN1, and UCHL1. We also present pharmacological agents acting via modulation of autophagy that may show promise in AD therapy. This review updates our knowledge on autophagy mechanisms proposing novel therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Anna Stachowiak
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| | | | - Nikolay T. Tzvetkov
- Department of Molecular Biology and Biochemical Pharmacology, Institute of Molecular Biology “Roumen Tsanev”, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Shinya Takeda
- Department of Clinical Psychology, Tottori University Graduate School of Medical Sciences, Tottori, Japan
| | - Atanas G. Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | | | - Mohamed M. Abdel-Daim
- Department of Pharmacology, Suez Canal University, Ismailia, Egypt
- Department of Ophthalmology and Micro-technology, Yokohama City University, Yokohama, Japan
| | - Adrian M. Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| |
Collapse
|
7
|
He Y, Pan S, Xu M, He R, Huang W, Song P, Huang J, Zhang H, Hu Y. Adeno‐associated virus 9–mediated Cdk5 inhibitory peptide reverses pathologic changes and behavioral deficits in the Alzheimer's disease mouse model. FASEB J 2017; 31:3383-3392. [PMID: 28420695 DOI: 10.1096/fj.201700064r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/05/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Yong He
- Department of NeurologyNanfang HospitalSouthern Medical University Guangzhou China
- Department of NeurologyFirst People's Hospital of Chenzhou Chenzhou China
| | - Suyue Pan
- Department of NeurologyNanfang HospitalSouthern Medical University Guangzhou China
| | - Miaojing Xu
- Department of NeurologyNanfang HospitalSouthern Medical University Guangzhou China
| | - Rongni He
- Department of NeurologyZhujiang HospitalSouthern Medical University Guangzhou China
| | - Wei Huang
- Department of NeurologyZhujiang HospitalSouthern Medical University Guangzhou China
| | - Pingping Song
- Department of NeurologyNanfang HospitalSouthern Medical University Guangzhou China
| | - Jianou Huang
- Department of Neurology421 Hospital Guangzhou China
| | - Han‐Ting Zhang
- Department of Behavioral Medicine and PsychiatryWest Virginia University Health Sciences Center Morgantown West Virginia USA
- Department of Physiology and PharmacologyWest Virginia University Health Sciences Center Morgantown West Virginia USA
| | - Yafang Hu
- Department of NeurologyNanfang HospitalSouthern Medical University Guangzhou China
| |
Collapse
|
8
|
De Vos KJ, Hafezparast M. Neurobiology of axonal transport defects in motor neuron diseases: Opportunities for translational research? Neurobiol Dis 2017; 105:283-299. [PMID: 28235672 PMCID: PMC5536153 DOI: 10.1016/j.nbd.2017.02.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/20/2017] [Indexed: 12/12/2022] Open
Abstract
Intracellular trafficking of cargoes is an essential process to maintain the structure and function of all mammalian cell types, but especially of neurons because of their extreme axon/dendrite polarisation. Axonal transport mediates the movement of cargoes such as proteins, mRNA, lipids, membrane-bound vesicles and organelles that are mostly synthesised in the cell body and in doing so is responsible for their correct spatiotemporal distribution in the axon, for example at specialised sites such as nodes of Ranvier and synaptic terminals. In addition, axonal transport maintains the essential long-distance communication between the cell body and synaptic terminals that allows neurons to react to their surroundings via trafficking of for example signalling endosomes. Axonal transport defects are a common observation in a variety of neurodegenerative diseases, and mutations in components of the axonal transport machinery have unequivocally shown that impaired axonal transport can cause neurodegeneration (reviewed in El-Kadi et al., 2007, De Vos et al., 2008; Millecamps and Julien, 2013). Here we review our current understanding of axonal transport defects and the role they play in motor neuron diseases (MNDs) with a specific focus on the most common form of MND, amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Kurt J De Vos
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Majid Hafezparast
- Neuroscience, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
9
|
Shukla V, Seo J, Binukumar B, Amin ND, Reddy P, Grant P, Kuntz S, Kesavapany S, Steiner J, Mishra SK, Tsai LH, Pant HC. TFP5, a Peptide Inhibitor of Aberrant and Hyperactive Cdk5/p25, Attenuates Pathological Phenotypes and Restores Synaptic Function in CK-p25Tg Mice. J Alzheimers Dis 2017; 56:335-349. [PMID: 28085018 PMCID: PMC10020940 DOI: 10.3233/jad-160916] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It has been reported that cyclin-dependent kinase 5 (cdk5), a critical neuronal kinase, is hyperactivated in Alzheimer's disease (AD) and may be, in part, responsible for the hallmark pathology of amyloid plaques and neurofibrillary tangles (NFTs). It has been proposed by several laboratories that hyperactive cdk5 results from the overexpression of p25 (a truncated fragment of p35, the normal cdk5 regulator), which, when complexed to cdk5, induces hyperactivity, hyperphosphorylated tau/NFTs, amyloid-β plaques, and neuronal death. It has previously been shown that intraperitoneal (i.p.) injections of a modified truncated 24-aa peptide (TFP5), derived from the cdk5 activator p35, penetrated the blood-brain barrier and significantly rescued AD-like pathology in 5XFAD model mice. The principal pathology in the 5XFAD mutant, however, is extensive amyloid plaques; hence, as a proof of concept, we believe it is essential to demonstrate the peptide's efficacy in a mouse model expressing high levels of p25, such as the inducible CK-p25Tg model mouse that overexpresses p25 in CamKII positive neurons. Using a modified TFP5 treatment, here we show that peptide i.p. injections in these mice decrease cdk5 hyperactivity, tau, neurofilament-M/H hyperphosphorylation, and restore synaptic function and behavior (i.e., spatial working memory, motor deficit using Rota-rod). It is noteworthy that TFP5 does not inhibit endogenous cdk5/p35 activity, nor other cdks in vivo suggesting it might have no toxic side effects, and may serve as an excellent therapeutic candidate for neurodegenerative disorders expressing abnormally high brain levels of p25 and hyperactive cdk5.
Collapse
Affiliation(s)
- Varsha Shukla
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jinsoo Seo
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - B.K. Binukumar
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Niranjana D. Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Preethi Reddy
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Philip Grant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Susan Kuntz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Joseph Steiner
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Santosh K. Mishra
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MA, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harish C. Pant
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Correspondence to: Dr. Harish C. Pant, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Zhou X, Huang J, Pan S, Xu M, He R, Ji Z, Hu Y. Neurodegeneration-Like Pathological and Behavioral Changes in an AAV9-Mediated p25 Overexpression Mouse Model. J Alzheimers Dis 2016; 53:843-55. [PMID: 27258419 DOI: 10.3233/jad-160191] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Xiao Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianou Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- 421 Hospital, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Miaojing Xu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rongni He
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong Ji
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Ciesielski J, Su TP, Tsai SY. Myristic acid hitchhiking on sigma-1 receptor to fend off neurodegeneration. ACTA ACUST UNITED AC 2016; 3. [PMID: 27077074 PMCID: PMC4827442 DOI: 10.14800/rci.1114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurodegenerative diseases are linked to tauopathy as a result of cyclin dependent kinase 5 (cdk5) binding to its p25 activator instead of its p35 activator and becoming over-activated. The overactive complex stimulates the hyperphosphorylation of tau proteins, leading to neurofibrillary tangles (NFTs) and stunting axon growth and development. It is known that the sigma-1 receptor (Sig-1R), an endoplasmic reticulum chaperone, can be involved in axon growth by promoting neurite sprouting through nerve growth factor (NGF) and tropomyosin receptor kinase B (TrkB)[1, 2]. It has also been previously demonstrated that a Sig-1R deficiency impairs the process of neurogenesis by causing a down-regulation of N-methyl-D-aspartate receptors (NMDARs)[3]. The recent study by Tsai et al. sought to understand the relationship between Sig-1R and tauopathy[4]. It was discovered that the Sig-1R helps maintain proper tau phosphorylation and axon development by facilitating p35 myristoylation and promoting p35 turnover. Neurons that had the Sig-1R knocked down exhibited shortened axons and higher levels of phosphorylated tau proteins compared to control neurons. Here we discuss these recent findings on the role of Sig-1R in tauopathy and highlight the newly presented physiological consequences of the Sig-1R-lipid interaction, helping to understand the close relationship between lipids and neurodegeneration.
Collapse
Affiliation(s)
- Jenna Ciesielski
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, Maryland 21224, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, Maryland 21224, USA
| | - Shang-Yi Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, DHHS, Baltimore, Maryland 21224, USA
| |
Collapse
|
12
|
Tsai SYA, Pokrass MJ, Klauer NR, Nohara H, Su TP. Sigma-1 receptor regulates Tau phosphorylation and axon extension by shaping p35 turnover via myristic acid. Proc Natl Acad Sci U S A 2015; 112:6742-7. [PMID: 25964330 PMCID: PMC4450430 DOI: 10.1073/pnas.1422001112] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dysregulation of cyclin-dependent kinase 5 (cdk5) per relative concentrations of its activators p35 and p25 is implicated in neurodegenerative diseases. P35 has a short t½ and undergoes rapid proteasomal degradation in its membrane-bound myristoylated form. P35 is converted by calpain to p25, which, along with an extended t½, promotes aberrant activation of cdk5 and causes abnormal hyperphosphorylation of tau, thus leading to the formation of neurofibrillary tangles. The sigma-1 receptor (Sig-1R) is an endoplasmic reticulum chaperone that is implicated in neuronal survival. However, the specific role of the Sig-1R in neurodegeneration is unclear. Here we found that Sig-1Rs regulate proper tau phosphorylation and axon extension by promoting p35 turnover through the receptor's interaction with myristic acid. In Sig-1R-KO neurons, a greater accumulation of p35 is seen, which results from neither elevated transcription of p35 nor disrupted calpain activity, but rather to the slower degradation of p35. In contrast, Sig-1R overexpression causes a decrease of p35. Sig-1R-KO neurons exhibit shorter axons with lower densities. Myristic acid is found here to bind Sig-1R as an agonist that causes the dissociation of Sig-1R from its cognate partner binding immunoglobulin protein. Remarkably, treatment of Sig-1R-KO neurons with exogenous myristic acid mitigates p35 accumulation, diminishes tau phosphorylation, and restores axon elongation. Our results define the involvement of Sig-1Rs in neurodegeneration and provide a mechanistic explanation that Sig-1Rs help maintain proper tau phosphorylation by potentially carrying and providing myristic acid to p35 for enhanced p35 degradation to circumvent the formation of overreactive cdk5/p25.
Collapse
Affiliation(s)
- Shang-Yi A Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD 21224
| | - Michael J Pokrass
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD 21224
| | - Neal R Klauer
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD 21224
| | - Hiroshi Nohara
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD 21224
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, US Department of Health and Human Services, Baltimore, MD 21224
| |
Collapse
|
13
|
Analysis of Cdk5-related phosphoproteomics in growth cones. J Mol Neurosci 2014; 52:384-91. [PMID: 24234032 DOI: 10.1007/s12031-013-0162-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Neurons establish interactions with target cells via elongation and guidance of axons, and the growth cone plays pivotal roles in this process. Cyclin-dependent kinase 5 (Cdk5)is a key regulator of nervous system development. Cdk5 regulates several significant events by phosphorylating substrates that are involved in neurogenesis, and previous studies of Cdk5 have typically focused on single substrates. Here, we took anew approach to investigate Cdk5 substrates using mass spectrometry and bioinformatics analyses. Axonal growth cones were isolated and analyzed by HPLC-MALDI-MS/MS. In total, 178,617 MS/MS spectra were detected. Candidates were analyzed by GPS 2.1 and Scansite 3, which predicted that 2,664 and 275 sites, respectively, were potential phosphorylation sites of Cdk5. There were 190 overlapped phosphorylation sites, corresponding to 89 proteins. Those proteins correlated with axonal functions were classified, and two of them were verified using a classic site-specific mutation strategy. This is the first study in which the phosphoproteome of axonal growth cones was identified. The systematic examination of Cdk5 substrates could provide a reference for further study of molecular mechanisms of axonal growth cones, and new insights into treatments of neuronal disorders.
Collapse
|
14
|
D'Alton S, Lewis J. Therapeutic and diagnostic challenges for frontotemporal dementia. Front Aging Neurosci 2014; 6:204. [PMID: 25191265 PMCID: PMC4137452 DOI: 10.3389/fnagi.2014.00204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/25/2014] [Indexed: 12/12/2022] Open
Abstract
In the search for therapeutic modifiers, frontotemporal dementia (FTD) has traditionally been overshadowed by other conditions such as Alzheimer's disease (AD). A clinically and pathologically diverse condition, FTD has been galvanized by a number of recent discoveries such as novel genetic variants in familial and sporadic forms of disease and the identification of TAR DNA binding protein of 43 kDa (TDP-43) as the defining constituent of inclusions in more than half of cases. In combination with an ever-expanding knowledge of the function and dysfunction of tau-a protein which is pathologically aggregated in the majority of the remaining cases-there exists a greater understanding of FTD than ever before. These advances may indicate potential approaches for the development of hypothetical therapeutics, but FTD remains highly complex and the roles of tau and TDP-43 in neurodegeneration are still wholly unclear. Here the challenges facing potential therapeutic strategies are discussed, which include sufficiently accurate disease diagnosis and sophisticated technology to deliver effective therapies.
Collapse
Affiliation(s)
- Simon D'Alton
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida Gainesville, FL, USA
| | - Jada Lewis
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida Gainesville, FL, USA
| |
Collapse
|
15
|
Zimmer ER, Leuzy A, Bhat V, Gauthier S, Rosa-Neto P. In vivo tracking of tau pathology using positron emission tomography (PET) molecular imaging in small animals. Transl Neurodegener 2014; 3:6. [PMID: 24628994 PMCID: PMC3995516 DOI: 10.1186/2047-9158-3-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/07/2014] [Indexed: 12/21/2022] Open
Abstract
Hyperphosphorylation of the tau protein leading to the formation of neurofibrillary tangles (NFTs) is a common feature in a wide range of neurodegenerative diseases known as tauopathies, which include Alzheimer's disease (AD) and the frontotemporal dementias (FTDs). Although heavily investigated, the mechanisms underlying the pathogenesis and progression of tauopathies have yet to be fully understood. In this context, several rodent models have been developed that successfully recapitulate the behavioral and neurochemical features of tau pathology, aiming to achieve a better understanding of the link between tau and neurodegeneration. To date, behavioral and biochemical parameters assessed using these models have been conducted using a combination of memory tasks and invasive methods such as cerebrospinal fluid (CSF) sampling or post-mortem analysis. Recently, several novel positron emission tomography (PET) radiopharmaceuticals targeting tau tangles have been developed, allowing for non-invasive in vivo quantification of tau pathology. Combined with tau transgenic models and microPET, these tracers hold the promise of advancing the development of theoretical models and advancing our understanding of the natural history of AD and non-AD tauopathies. In this review, we briefly describe some of the most important insights for understanding the biological basis of tau pathology, and shed light on the opportunity for improved modeling of tau pathology using a combination of tau-radiopharmaceuticals and animal models.
Collapse
Affiliation(s)
- Eduardo Rigon Zimmer
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging, Douglas Mental Health University Institute, McGill University, 6875 La Salle Blv - FBC room 3149, Montreal, QC, H4H 1R3, Canada.,Montreal Neurological Institute (MNI), Montreal, Canada.,Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Antoine Leuzy
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging, Douglas Mental Health University Institute, McGill University, 6875 La Salle Blv - FBC room 3149, Montreal, QC, H4H 1R3, Canada.,Montreal Neurological Institute (MNI), Montreal, Canada
| | - Venkat Bhat
- Department of Psychiatry, McGill University, Montreal, Brazil
| | - Serge Gauthier
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging, Douglas Mental Health University Institute, McGill University, 6875 La Salle Blv - FBC room 3149, Montreal, QC, H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging, Douglas Mental Health University Institute, McGill University, 6875 La Salle Blv - FBC room 3149, Montreal, QC, H4H 1R3, Canada.,Montreal Neurological Institute (MNI), Montreal, Canada
| |
Collapse
|
16
|
Specific inhibition of p25/Cdk5 activity by the Cdk5 inhibitory peptide reduces neurodegeneration in vivo. J Neurosci 2013; 33:334-43. [PMID: 23283346 DOI: 10.1523/jneurosci.3593-12.2013] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The aberrant hyperactivation of Cyclin-dependent kinase 5 (Cdk5), by the production of its truncated activator p25, results in the formation of hyperphosphorylated tau, neuroinflammation, amyloid deposition, and neuronal death in vitro and in vivo. Mechanistically, this occurs as a result of a neurotoxic insult that invokes the intracellular elevation of calcium to activate calpain, which cleaves the Cdk5 activator p35 into p25. It has been shown previously that the p25 transgenic mouse as a model to investigate the mechanistic implications of p25 production in the brain, which recapitulates deregulated Cdk5-mediated neuropathological changes, such as hyperphosphorylated tau and neuronal death. To date, strategies to inhibit Cdk5 activity have not been successful in targeting selectively aberrant activity without affecting normal Cdk5 activity. Here we show that the selective inhibition of p25/Cdk5 hyperactivation in vivo, through overexpression of the Cdk5 inhibitory peptide (CIP), rescues against the neurodegenerative pathologies caused by p25/Cdk5 hyperactivation without affecting normal neurodevelopment afforded by normal p35/Cdk5 activity. Tau and amyloid pathologies as well as neuroinflammation are significantly reduced in the CIP-p25 tetra transgenic mice, whereas brain atrophy and subsequent cognitive decline are reversed in these mice. The findings reported here represent an important breakthrough in elucidating approaches to selectively inhibit the p25/Cdk5 hyperactivation as a potential therapeutic target to reduce neurodegeneration.
Collapse
|
17
|
Boutajangout A, Sigurdsson EM, Krishnamurthy PK. Tau as a therapeutic target for Alzheimer's disease. Curr Alzheimer Res 2012; 8:666-77. [PMID: 21679154 DOI: 10.2174/156720511796717195] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 04/10/2011] [Accepted: 04/15/2011] [Indexed: 01/02/2023]
Abstract
Neurofibrillary tangles (NFTs) are one of the pathological hallmarks of Alzheimer's disease (AD) and are primarily composed of aggregates of hyperphosphorylated forms of the microtubule associated protein tau. It is likely that an imbalance of kinase and phosphatase activities leads to the abnormal phosphorylation of tau and subsequent aggregation. The wide ranging therapeutic approaches that are being developed include to inhibit tau kinases, to enhance phosphatase activity, to promote microtubule stability, and to reduce tau aggregate formation and/or enhance their clearance with small molecule drugs or by immunotherapeutic means. Most of these promising approaches are still in preclinical development whilst some have progressed to Phase II clinical trials. By pursuing these lines of study, a viable therapy for AD and related tauopathies may be obtained.
Collapse
Affiliation(s)
- A Boutajangout
- Departments of Physiology and Neuroscience, New York University School of Medicine, New York, NY 10016, USA.
| | | | | |
Collapse
|
18
|
Pang TYC, Hannan AJ. Enhancement of cognitive function in models of brain disease through environmental enrichment and physical activity. Neuropharmacology 2012; 64:515-28. [PMID: 22766390 DOI: 10.1016/j.neuropharm.2012.06.029] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/06/2012] [Accepted: 06/15/2012] [Indexed: 12/21/2022]
Abstract
This review will provide an overview of the non-drug based approaches that have been demonstrated to enhance cognitive function of the compromised brain, primarily focussed on the two most widely adopted paradigms of environmental enrichment and enhanced physical exercise. Environmental enrichment involves the generation of novelty and complexity in animal housing conditions which facilitates enhanced sensory and cognitive stimulation as well as physical activity. In a wide variety of animal models of brain disorders, environmental enrichment and exercise have been found to have beneficial effects, including cognitive enhancement, delayed disease onset, enhanced cellular plasticity and associated molecular processes. Potential cellular and molecular mechanisms will also be discussed, which have relevance for the future development of 'enviromimetics', drugs which could mimic or enhance the beneficial effects of environmental stimulation. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Terence Y C Pang
- Florey Neuroscience Institutes, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3010, Australia.
| | | |
Collapse
|
19
|
Chadwick L, Gentle L, Strachan J, Layfield R. Review: unchained maladie - a reassessment of the role of Ubb(+1) -capped polyubiquitin chains in Alzheimer's disease. Neuropathol Appl Neurobiol 2012; 38:118-31. [PMID: 22082077 DOI: 10.1111/j.1365-2990.2011.01236.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molecular misreading allows the formation of mutant proteins in the absence of gene mutations. A mechanism has been proposed by which a frameshift mutant of the ubiquitin protein, Ubb(+1) , which accumulates in an age-dependent manner as a result of molecular misreading, contributes to neuropathology in Alzheimer's disease (Lam et al. 2000). Specifically, in the Ubb(+1) -mediated proteasome inhibition hypothesis Ubb(+1) 'caps' unanchored (that is, nonsubstrate linked) polyubiquitin chains, which then act as dominant inhibitors of the 26S proteasome. A review of subsequent literature indicates that this original hypothesis is broadly supported, and offers new insights into the mechanisms accounting for the age-dependent accumulation of Ubb(+1) , and how Ubb(+1) -mediated proteasome inhibition may contribute to Alzheimer's disease. Further, recent studies have highlighted a physiological role for free endogenous unanchored polyubiquitin chains in the direct activation of certain protein kinases. This raises the possibility that Ubb(+1) -capped unanchored polyubiquitin chains could also exert harmful effects through the aberrant activation of tau or other ubiquitin-dependent kinases, neuronal NF-κB activity or NF-κB-mediated neuroinflammatory processes.
Collapse
Affiliation(s)
- L Chadwick
- School of Biomedical Sciences, University of Nottingham, UK
| | | | | | | |
Collapse
|
20
|
Cdk5/p25-induced cytosolic PLA2-mediated lysophosphatidylcholine production regulates neuroinflammation and triggers neurodegeneration. J Neurosci 2012; 32:1020-34. [PMID: 22262900 DOI: 10.1523/jneurosci.5177-11.2012] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The deregulation of cyclin-dependent kinase 5 (Cdk5) by p25 has been shown to contribute to the pathogenesis in a number of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD) and Alzheimer's disease (AD). In particular, p25/Cdk5 has been shown to produce hyperphosphorylated tau, neurofibrillary tangles as well as aberrant amyloid precursor protein processing found in AD. Neuroinflammation has been observed alongside the pathogenic process in these neurodegenerative diseases, however the precise mechanism behind the induction of neuroinflammation and the significance in the AD pathogenesis has not been fully elucidated. In this report, we uncover a novel pathway for p25-induced neuroinflammation where p25 expression induces an early trigger of neuroinflammation in vivo in mice. Lipidomic mass spectrometry, in vitro coculture and conditioned media transfer experiments show that the soluble lipid mediator lysophosphatidylcholine (LPC) is released by p25 overexpressing neurons to initiate astrogliosis, neuroinflammation and subsequent neurodegeneration. Reverse transcriptase PCR and gene silencing experiments show that cytosolic phospholipase 2 (cPLA2) is the key enzyme mediating the p25-induced LPC production and cPLA2 upregulation is critical in triggering the p25-mediated inflammatory and neurodegenerative process. Together, our findings delineate a potential therapeutic target for the reduction of neuroinflammation in neurodegenerative diseases including AD.
Collapse
|
21
|
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a multifaceted serine/threonine kinase protein with important roles in the nervous system. Two related proteins, p35 and p39, activate Cdk5 upon direct binding. Over the past decade, Cdk5 activity has been demonstrated to regulate many events during brain development, including neuronal migration as well as axon and dendrite development. Recent evidence also suggests a pivotal role for Cdk5 in synaptic plasticity, behavior, and cognition. Dysfunction of Cdk5 has been implicated in a number of neurological disorders and neurodegenerative diseases including Alzheimer's disease, amyotrophic lateral sclerosis, Niemann-Pick type C disease, and ischemia. Hyperactivation of Cdk5 due to the conversion of p35 to p25 by the calcium-dependent protease calpain during neurotoxicity also contributes to the pathological state. This review surveys recent literature surrounding Cdk5 in synaptic plasticity and homeostasis, with particular emphasis on Cdk5 kinase activity under neurodegenerative conditions.
Collapse
Affiliation(s)
- Susan C Su
- Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
22
|
Engmann O, Hortobágyi T, Thompson AJ, Guadagno J, Troakes C, Soriano S, Al-Sarraj S, Kim Y, Giese KP. Cyclin-dependent kinase 5 activator p25 is generated during memory formation and is reduced at an early stage in Alzheimer's disease. Biol Psychiatry 2011; 70:159-68. [PMID: 21616478 DOI: 10.1016/j.biopsych.2011.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/23/2011] [Accepted: 04/06/2011] [Indexed: 01/08/2023]
Abstract
BACKGROUND The cyclin-dependent kinase 5 activator p35 can be cleaved into p25. Formation of p25 has been suggested to contribute to neurodegeneration in Alzheimer's disease (AD). However, overexpression of low levels of p25 in mice enhances memory formation. Therefore, it has been suggested that p25 formation might be an event early in AD to compensate for impairments in synaptic plasticity. Ongoing p25 formation has been hypothesized to contribute to neurodegeneration at the later stages of AD. METHODS Here, we tested the early compensation hypothesis by analyzing the levels of p25 and its precursor p35 in AD postmortem samples from different brain regions at different stages of tau pathology, using quantitative Western blots. Furthermore, we studied p35 and p25 during spatial memory formation. By employing quantitative mass spectrometry, we identified proteins downstream of p25, which were then studied in AD samples. RESULTS We found that p25 is generated during spatial memory formation. Furthermore, we demonstrate that overexpression of p25 in the physiological range increases the expression of two proteins implicated in spine formation, septin 7 and optic atrophy 1. We show that the expression of p35 and p25 is reduced as an early event in AD. Moreover, expression of the p25-regulated protein optic atrophy 1 was reduced in a time course similar to p25 expression. CONCLUSIONS Our findings suggest that p25 generation is a mechanism underlying hippocampal memory formation that is impaired in the early stages of AD. Our findings argue against the previously raised early compensation hypothesis and they propose that p25-mediated neurotoxicity does not occur in AD.
Collapse
Affiliation(s)
- Olivia Engmann
- Department of Neuroscience, Medical Research Council Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mungenast AE, Tsai LH. Addressing the complex etiology of Alzheimer’s disease: the role of p25/Cdk5. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder characterized by the progressive loss of forebrain neurons and the deterioration of learning and memory. Therapies for AD have primarily focused upon either the inhibition of amyloid synthesis or its deposition in the brain, but clinical testing to date has not yet found an effective amelioration of cognitive symptoms. Synaptic loss closely correlates with the degree of dementia in AD patients. However, mouse AD models that target the amyloid-β pathway generally do not exhibit a profound loss of synapses, despite extensive synaptic dysfunction. The increased generation of p25, an activator of the cyclin-dependent kinase 5 (Cdk5) has been found in both human patients and mouse models of neurodegeneration. The current work reviews our knowledge, to date, on the role of p25/Cdk5 in Alzheimer’s disease, with a focus upon the interaction of amyloid-β and p25/Cdk5 in synaptic dysfunction and neuronal loss.
Collapse
Affiliation(s)
- Alison E Mungenast
- Picower Institute for Learning & Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li-Huei Tsai
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
24
|
De Vos A, Anandhakumar J, Van den Brande J, Verduyckt M, Franssens V, Winderickx J, Swinnen E. Yeast as a model system to study tau biology. Int J Alzheimers Dis 2011; 2011:428970. [PMID: 21559193 PMCID: PMC3090044 DOI: 10.4061/2011/428970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/21/2011] [Indexed: 11/20/2022] Open
Abstract
Hyperphosphorylated and aggregated human protein tau constitutes a hallmark of a multitude of neurodegenerative diseases called tauopathies, exemplified by Alzheimer's disease. In spite of an enormous amount of research performed on tau biology, several crucial questions concerning the mechanisms of tau toxicity remain unanswered. In this paper we will highlight some of the processes involved in tau biology and pathology, focusing on tau phosphorylation and the interplay with oxidative stress. In addition, we will introduce the development of a human tau-expressing yeast model, and discuss some crucial results obtained in this model, highlighting its potential in the elucidation of cellular processes leading to tau toxicity.
Collapse
Affiliation(s)
- Ann De Vos
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jayamani Anandhakumar
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jeff Van den Brande
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Mathias Verduyckt
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Vanessa Franssens
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Erwin Swinnen
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| |
Collapse
|
25
|
Gruenenfelder FI, Thomson G, Penderis J, Edgar JM. Axon-glial interaction in the CNS: what we have learned from mouse models of Pelizaeus-Merzbacher disease. J Anat 2011; 219:33-43. [PMID: 21401588 DOI: 10.1111/j.1469-7580.2011.01363.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In the central nervous system (CNS) the majority of axons are surrounded by a myelin sheath, which is produced by oligodendrocytes. Myelin is a lipid-rich insulating material that facilitates the rapid conduction of electrical impulses along the myelinated nerve fibre. Proteolipid protein and its isoform DM20 constitute the most abundant protein component of CNS myelin. Mutations in the PLP1 gene encoding these myelin proteins cause Pelizaeus-Merzbacher disease and the related allelic disorder, spastic paraplegia type 2. Animal models of these diseases, particularly models lacking or overexpressing Plp1, have shed light on the interplay between axons and oligodendrocytes, and how one component influences the other.
Collapse
Affiliation(s)
- Fredrik I Gruenenfelder
- Applied Neurobiology Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | | | | | | |
Collapse
|
26
|
Hanger DP, Seereeram A, Noble W. Mediators of tau phosphorylation in the pathogenesis of Alzheimer's disease. Expert Rev Neurother 2010; 9:1647-66. [PMID: 19903024 DOI: 10.1586/ern.09.104] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The need for disease-modifying drugs for Alzheimer's disease has become increasingly important owing to escalating disease prevalence and the associated socio-economic burden. Until recently, reducing brain amyloid accumulation has been the main therapeutic focus; however, increasing evidence suggests that targeting abnormal tau phosphorylation could be beneficial. Tau is phosphorylated by several protein kinases and this is balanced by dephosphorylation by protein phosphatases. Phosphorylation at specific sites can influence the physiological functions of tau, including its role in binding to and stabilizing the neuronal cytoskeleton. aberrant phosphorylation of tau could render it susceptible to potentially pathogenic alterations, including conformational changes, proteolytic cleavage and aggregation. While strategies that reduce tau phosphorylation in transgenic models of disease have been promising, our understanding of the mechanisms through which tau becomes abnormally phosphorylated in disease is lacking.
Collapse
Affiliation(s)
- Diane P Hanger
- MRC Centre for Neurodegeneration Research, King's College London, Institute of Psychiatry, Department of Neuroscience (P037), De Crespigny Park, London SE5 8AF, UK.
| | | | | |
Collapse
|
27
|
Spatial learning impairment, enhanced CDK5/p35 activity, and downregulation of NMDA receptor expression in transgenic mice expressing tau-tubulin kinase 1. J Neurosci 2009; 28:14511-21. [PMID: 19118186 DOI: 10.1523/jneurosci.3417-08.2008] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Tau-tubulin kinase-1 (TTBK1) is involved in phosphorylation of tau protein at specific Serine/Threonine residues found in paired helical filaments, suggesting its role in tauopathy pathogenesis. We found that TTBK1 levels were upregulated in brains of human Alzheimer' disease (AD) patients compared with age-matched non-AD controls. To understand the effects of TTBK1 activation in vivo, we developed transgenic mice harboring human full-length TTBK1 genomic DNA (TTBK1-Tg). Transgenic TTBK1 is highly expressed in subiculum and cortical pyramidal layers, and induces phosphorylated neurofilament aggregation. TTBK1-Tg mice show significant age-dependent memory impairment as determined by radial arm water maze test, which is associated with enhancement of tau and neurofilament phosphorylation, increased levels of p25 and p35, both activators of cyclin-dependent protein kinase 5 (CDK5), enhanced calpain I activity, and reduced levels of hippocampal NMDA receptor types 2B (NR2B) and D. Enhanced CDK5/p35 complex formation is strongly correlated with dissociation of F-actin from p35, suggesting the inhibitory mechanism of CDK5/p35 complex formation by F-actin. Expression of recombinant TTBK1 in primary mouse cortical neurons significantly downregulated NR2B in a CDK5- and calpain-dependent manner. These data suggest that TTBK1 in AD brain may be one of the underlying mechanisms inducing CDK5 and calpain activation, NR2B downregulation, and subsequent memory dysfunction.
Collapse
|
28
|
Gendron TF, Petrucelli L. The role of tau in neurodegeneration. Mol Neurodegener 2009; 4:13. [PMID: 19284597 PMCID: PMC2663562 DOI: 10.1186/1750-1326-4-13] [Citation(s) in RCA: 333] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 03/11/2009] [Indexed: 01/31/2023] Open
Abstract
Since the identification of tau as the main component of neurofibrillary tangles in Alzheimer's disease and related tauopathies, and the discovery that mutations in the tau gene cause frontotemporal dementia, much effort has been directed towards determining how the aggregation of tau into fibrillar inclusions causes neuronal death. As evidence emerges that tau-mediated neuronal death can occur even in the absence of tangle formation, a growing number of studies are focusing on understanding how abnormalities in tau (e.g. aberrant phosphorylation, glycosylation or truncation) confer toxicity. Though data obtained from experimental models of tauopathies strongly support the involvement of pathologically modified tau and tau aggregates in neurodegeneration, the exact neurotoxic species remain unclear, as do the mechanism(s) by which they cause neuronal death. Nonetheless, it is believed that tau-mediated neurodegeneration is likely to result from a combination of toxic gains of function as well as from the loss of normal tau function. To truly appreciate the detrimental consequences of aberrant tau function, a better understanding of all functions carried out by tau, including but not limited to the role of tau in microtubule assembly and stabilization, is required. This review will summarize what is currently known regarding the involvement of tau in the initiation and development of neurodegeneration in tauopathies, and will also highlight some of the remaining questions in need of further investigation.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida, USA.
| | | |
Collapse
|
29
|
Mlewski EC, Krapacher FA, Ferreras S, Paglini G. Transient enhanced expression of Cdk5 activator p25 after acute and chronic d-amphetamine administration. Ann N Y Acad Sci 2008; 1139:89-102. [PMID: 18991853 DOI: 10.1196/annals.1432.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The cellular and molecular mechanisms of sensitization in the addictive process are still unclear. Recently, chronic treatment with cocaine has been shown to upregulate the expression of cyclin-dependent kinase 5 (cdk5) and its specific activator, p35, in the striatum, as a downstream target gene of DeltaFosB, and has been implicated in compensatory adaptive changes associated with psychostimulants. Cdk5 is a serine/threonine kinase and its activation is achieved through association with a regulatory subunit, either p35 or p39. P35 is cleaved by the protease calpain, which results in the generation of a truncated product termed p25, which contains all elements necessary for cdk5 activation. The cdk5/p35 complex plays an essential role in neuronal development and survival. It has also been involved in neuronal trafficking and transport and in dopaminergic transmission, indicating its role either in presynaptic and postsynaptic signaling. In this study we report that the cdk5/p35 complex participates in acute and chronic d-amphetamine (AMPH)-evoked behavioral events, and we show a surprisingly transient enhanced expression of p25 and a lasting increased expression of p35 in dorsal striatal synaptosomes after acute and chronic AMPH administration. Pak1, a substrate for cdk5, is also enriched in the synaptosomal fraction of acute AMPH-treated rats. Our data suggest that the transient upregulation of p25 may regulate the activity of cdk5 in phosphorylating particular substrates, such as Pak1, implicated in the compensatory adaptive morphophysiologic changes associated with the process of behavioral sensitization to psychostimulants.
Collapse
Affiliation(s)
- Estela Cecilia Mlewski
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Córdoba, Argentina
| | | | | | | |
Collapse
|
30
|
Dopaminergic and glutamatergic signaling crosstalk in Huntington's disease neurodegeneration: the role of p25/cyclin-dependent kinase 5. J Neurosci 2008; 28:10090-101. [PMID: 18829967 DOI: 10.1523/jneurosci.3237-08.2008] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Altered glutamatergic and dopaminergic signaling has been proposed as contributing to the specific striatal cell death observed in Huntington's disease (HD). However, the precise mechanisms by which mutant huntingtin sensitize striatal cells to dopamine and glutamate inputs remain unclear. Here, we demonstrate in knock-in HD striatal cells that mutant huntingtin enhances dopamine-mediated striatal cell death via dopamine D(1) receptors. Moreover, we show that NMDA receptors specifically potentiate the vulnerability of mutant huntingtin striatal cells to dopamine toxicity as pretreatment with NMDA increased D(1)R-induced cell death in mutant but not wild-type cells. As potential underlying mechanism of increased striatal vulnerability, we identified aberrant cyclin-dependent kinase 5 (Cdk5) activation. We demonstrate that enhanced Cdk5 phosphorylation and increased calpain-mediated conversion of the Cdk5 activator p35 into p25 may account for the deregulation of Cdk5 associated to dopamine and glutamate receptor activation in knock-in HD striatal cells. Moreover, supporting a detrimental role of Cdk5 in striatal cell death, neuronal loss can be widely prevented by roscovitine, a potent Cdk5 inhibitor. Significantly, reduced Cdk5 expression together with enhanced Cdk5 phosphorylation and p25 accumulation also occurs in the striatum of mutant Hdh(Q111) mice and HD human brain suggesting the relevance of deregulated Cdk5 pathway in HD pathology. These findings provide new insights into the molecular mechanisms underlying the selective vulnerability of striatal cells in HD and identify p25/Cdk5 as an important mediator of dopamine and glutamate neurotoxicity associated to HD.
Collapse
|
31
|
Barten DM, Albright CF. Therapeutic strategies for Alzheimer's disease. Mol Neurobiol 2008; 37:171-86. [PMID: 18581273 DOI: 10.1007/s12035-008-8031-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 05/27/2008] [Indexed: 12/22/2022]
Abstract
Therapeutic approaches for Alzheimer's disease (AD) are guided by four disease characteristics: amyloid plaques, neurofibrillar tangles (NFT), neurodegeneration, and dementia. Amyloid plaques are composed largely of 4 kDa beta-amyloid (Abeta) peptides, with the more amyloidogenic, 42 amino acid form (Abeta42) as the primary species. Because multiple, rare mutations that cause early-onset, familial AD lead to increased production or aggregation of Abeta42, amyloid therapeutics aim to reduce the amount of toxic Abeta42 aggregates. Amyloid-based therapies include gamma-secretase inhibitors and modulators, BACE inhibitors, aggregation blockers, catabolism inducers, and anti-Abeta biologics. Tangles are composed of paired helical filaments of hyperphosphorylated tau protein. Tau-based therapeutics include kinase inhibitors, microtubule stabilizers, and catabolism inducers. Therapeutic strategies for neurodegeneration target multiple mechanisms, including excitotoxicity, mitochondrial dysfunction, oxidative damage, and inflammation or stimulation of neuronal viability. Although not disease modifying, cognition enhancers are important to treat the symptom of dementia. Strategies for cognition enhancement include cholinesterase inhibitors, and other approaches to enhance the signaling of cholinergic and glutamatergic neurons. In summary, plaques, tangles, neurodegeneration and dementia guide the development of multiple therapeutic approaches for AD and are the subject of this review.
Collapse
Affiliation(s)
- Donna M Barten
- Bristol Myers Squibb, Neuroscience Drug Discovery, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | |
Collapse
|
32
|
Wirths O, Bayer TA. Motor impairment in Alzheimer's disease and transgenic Alzheimer's disease mouse models. GENES BRAIN AND BEHAVIOR 2008; 7 Suppl 1:1-5. [PMID: 18184365 DOI: 10.1111/j.1601-183x.2007.00373.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In this commentary, we accent the accumulating evidence for motor impairment as a common feature of early Alzheimer's disease (AD) pathology. In addition, we summarize the state of knowledge on this phenotype in experimental mouse models, expressing AD-associated genes like tau or amyloid precursor protein.
Collapse
Affiliation(s)
- O Wirths
- Division of Molecular Psychiatry, Department of Psychiatry, University of Goettingen, Goettingen, Germany.
| | | |
Collapse
|
33
|
Muyllaert D, Terwel D, Kremer A, Sennvik K, Borghgraef P, Devijver H, Dewachter I, Van Leuven F. Neurodegeneration and neuroinflammation in cdk5/p25-inducible mice: a model for hippocampal sclerosis and neocortical degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:470-85. [PMID: 18202185 DOI: 10.2353/ajpath.2008.070693] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The cyclin-dependent kinase cdk5 is atypically active in postmitotic neurons and enigmatic among the kinases proposed as molecular actors in neurodegeneration. We generated transgenic mice to express p25, the N-terminally truncated p35 activator of cdk5, in forebrain under tetracycline control (TET-off). Neuronal expression of p25 (p25(ON)) caused high mortality postnatally and early in life. Mortality was completely prevented by administration of doxycycline in the drinking water of pregnant dams and litters until P42, allowing us to study the action of p25 in adult mouse forebrain. Neuronal p25 triggered neurodegeneration and also microgliosis, rapidly and intensely in hippocampus and cortex. Progressive neurodegeneration was severe with marked neuron loss, causing brain atrophy (40% loss at age 5 months) with nearly complete elimination of the hippocampus. Neurodegeneration did not involve phosphorylation of protein tau or generation of amyloid peptide. Degenerating neurons did not stain for terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling or activated caspase-3 but were marked by FluoroJadeB in early stages. Diseased neurons were always closely associated with activated microglia already very early in the disease process. Primary neurons derived from p25 embryos were more prone to apoptosis than wild-type neurons, and they activated microglial cells in co-culture. The inducible p25 mice present as a model for neurodegeneration in hippocampal sclerosis and neocortical degeneration, with important contributions of activated microglia.
Collapse
Affiliation(s)
- David Muyllaert
- Department of Human Genetics, KULeuven-Campus Gasthuisberg ON1-06.602, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Roder HM, Hutton ML. Microtubule-associated protein tau as a therapeutic target in neurodegenerative disease. Expert Opin Ther Targets 2007; 11:435-42. [PMID: 17373874 DOI: 10.1517/14728222.11.4.435] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Interest in the biology of the microtubule-associated protein tau, not only as a pathologic marker, but as a therapeutic target has surged considerably over the last few years. This is due, in part, to the discovery of mutations in tau causing a group of aggressively degenerative neurologic disorders characterized by abnormalities of tau very similar to what is seen in Alzheimer's disease where mutations in tau are absent. As these same mutations also precipitate authentic forms of neurofibrillary degeneration in tau transgenic mice, the gateways to testing therapeutic ideas preclinically have opened. Other Alzheimer's disease animal models have been notoriously bare of this feature, limiting their predictive power for clinical success. In this review, the authors discuss some of the main therapeutic ideas presently advanced in the field and their molecular rationales.
Collapse
Affiliation(s)
- Hanno M Roder
- Mayo Clinic, Department of Neuroscience, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | | |
Collapse
|
35
|
Gopinath S, Blair IP, Kennerson ML, Durnall JC, Nicholson GA. A novel locus for distal motor neuron degeneration maps to chromosome 7q34-q36. Hum Genet 2007; 121:559-64. [PMID: 17354000 DOI: 10.1007/s00439-007-0348-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Accepted: 02/23/2007] [Indexed: 12/13/2022]
Abstract
The motor neuron diseases (MND) are a group of related neurodegenerative diseases that cause the relative selective progressive death of motor neurons. These diseases range from slowly progressive forms including hereditary motor neuropathy (HMN), to the rapidly progressive disorder amyotrophic lateral sclerosis (ALS). There is clinical and genetic overlap among these MNDs, implicating shared pathogenic mechanisms. We recruited a large family with a MND that was previously described as juvenile ALS and distal HMN. We identified a novel MND/HMN locus on chromosome 7q34-q36 following a genome-wide scan for linkage in this family. The disease causing mutation maps to a 26.2 cM (12.3 Mb) interval flanked by D7S2513 and D7S637 on chromosome 7q34-q36. Recombinant haplotype analysis including unaffected individuals suggests that the refined candidate interval spans 14.3 cM (6.3 Mb) flanked by D7S2511 and D7S798. One gene in the candidate interval, CDK5, was selected for immediate mutation analysis based upon its known association with an ALS-like phenotype in mice however, no mutations were identified. Identification of genes causing familial MND will lead to a greater understanding of the biological basis of both familial and sporadic motor neuron degeneration including ALS.
Collapse
Affiliation(s)
- Sumana Gopinath
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord Hospital, and Faculty of Medicine, University of Sydney, NSW 2139, Australia
| | | | | | | | | |
Collapse
|
36
|
Steinhilb ML, Dias-Santagata D, Mulkearns EE, Shulman JM, Biernat J, Mandelkow EM, Feany MB. S/P and T/P phosphorylation is critical for tau neurotoxicity inDrosophila. J Neurosci Res 2007; 85:1271-8. [PMID: 17335084 DOI: 10.1002/jnr.21232] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The microtubule-associated protein tau is hyperphosphorylated abnormally in AD and related neurodegenerative disorders. Many phospho epitopes created by proline directed kinases (SP/TP sites) show relative specificity for disease states. To test whether phosphorylation at the disease-associated SP/TP sites affects tau toxicity in vivo, we expressed a form of tau in Drosophila in which all SP/TP sites are mutated to alanine. We find that blocking phosphorylation at SP/TP motifs markedly reduces tau toxicity in vivo. Using phosphorylation-specific antibodies, we identify a positive correlation between increased phosphorylation at disease-associated sites and neurotoxicity. We use the phosphorylation-incompetent version of tau to show that kinase and phosphatase modifiers of tau neurotoxicity, including cdk5/p35, the JNK kinase hemipterous and PP2A act via SP/TP phosphorylation sites. We provide direct evidence in an animal model system to support the role of phosphorylation at SP/TP sites in playing a critical role in tau neurotoxicity.
Collapse
Affiliation(s)
- Michelle L Steinhilb
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Julien JP, Kriz J. Chapter 6 Animal models of motor neuron death. HANDBOOK OF CLINICAL NEUROLOGY 2007; 82:121-138. [PMID: 18808891 DOI: 10.1016/s0072-9752(07)80009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
38
|
Kuan WL, Lin R, Tyers P, Barker RA. The importance of A9 dopaminergic neurons in mediating the functional benefits of fetal ventral mesencephalon transplants and levodopa-induced dyskinesias. Neurobiol Dis 2006; 25:594-608. [PMID: 17188499 DOI: 10.1016/j.nbd.2006.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Revised: 11/02/2006] [Accepted: 11/03/2006] [Indexed: 10/23/2022] Open
Abstract
Intrastriatal transplantation of fetal ventral mesencephalon (VM) tissue provides the potential to alleviate motor symptoms of Parkinson's disease (PD) and levodopa-induced dyskinesia (LID). However, the degree of recovery varies among individuals with an incidence of "off-phase", graft-induced dyskinesia (GID) in some patients. We hypothesised that this variability is due to the heterogeneous nature of dopaminergic neurons in the transplant. We therefore investigated this in the unilateral 6-hydroxydopamine-lesioned rat model of PD. These animals were primed to develop LID and then transplanted with fetal VM into the caudal aspects of the striatum. No GID was observed but in a significant number of animals the transplants ameliorated LID. There was a correlation between the degree of behavioural and LID recovery with the number of A9 dopaminergic neurons in the transplant, based on their expression of a G-protein-coupled inward rectifying current potassium channel (Girk2). Furthermore, we showed that LID development is related to an abnormal expression profile of cyclin-dependent kinase 5 (Cdk5) and dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) in the striatum and that intrastriatal VM transplants normalised both Cdk5 expression and DARPP-32 phosphorylation in animals exhibiting functional improvement. These results suggest that an A9 dopaminergic neuron-enriched transplant may be the key to an effective PD cell replacement therapy through normalisation of the altered striatal expression of Cdk5/DARPP-32.
Collapse
Affiliation(s)
- Wei-Li Kuan
- Cambridge Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge CB2 2PY, UK.
| | | | | | | |
Collapse
|
39
|
Lin WL, Zehr C, Lewis J, Hutton M, Yen SH, Dickson DW. Progressive white matter pathology in the spinal cord of transgenic mice expressing mutant (P301L) human tau. ACTA ACUST UNITED AC 2006; 34:397-410. [PMID: 16902761 DOI: 10.1007/s11068-006-8726-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Revised: 09/15/2004] [Accepted: 02/25/2005] [Indexed: 11/28/2022]
Abstract
Transgenic mice expressing mutant (P301L) tau develop paresis, neurofibrillary tangles and neuronal loss in spinal motor neurons beginning at 4 to 6 months of age. Astrocytes and oligodendrocytes acquire filamentous tau inclusions at later ages. Here we report pathology in the spinal white matter of these animals. Progressive white matter pathology, detected as early as 2 months of age, was most marked in lateral and anterior columns, with sparing of posterior columns until late in the disease. Early changes in Luxol fast blue/periodic acid Schiff (LFB/PAS) and toluidine blue stained sections were vacuolation of myelin followed by accumulation of myelin figures within previous axonal tubes and finally influx of PAS-positive macrophages. Myelin debris and vacuoles were found in macrophages. At the ultrastructural level, myelinated axons showed extensive vacuolation of myelin sheaths formed by splitting of myelin lamellae at the intra-period line, while axons were atrophic and contained densely packed neurofilaments. Other axons were lost completely, resulting in collapse and phagocytosis of myelin sheaths. Also present were spheroids derived from swollen axons with thin myelin sheaths containing neurofilaments, tau filaments and degenerating organelles. Many oligodendrocytes had membrane-bound cytoplasmic bodies composed of tightly stacked lamellae capped by dense material. The vacuolar myelopathy in this model to some extent resembles that reported in acquired immune deficiency syndrome and vitamin B12 deficiency. The progressive axonal pathology is most consistent with a dying-back process caused by abnormal accumulation of tau in upstream neurons, while vacuolar myelinopathy may be a secondary manifestation of neuroinflammation.
Collapse
Affiliation(s)
- Wen-Lang Lin
- Mayo Clinic College of Medicine, Jacksonville, Florida, 32224, USA
| | | | | | | | | | | |
Collapse
|
40
|
Neve RL, McPhie DL. The cell cycle as a therapeutic target for Alzheimer's disease. Pharmacol Ther 2006; 111:99-113. [PMID: 16274748 DOI: 10.1016/j.pharmthera.2005.09.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 09/21/2005] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide. It is a progressive, incurable disease whose predominant clinical manifestation is memory loss, and which always ends in death. The classic neuropathological diagnostic markers for AD are amyloid plaques and neurofibrillary tangles, but our understanding of the role that these features of AD play in the etiology and progression of the disease remains incomplete. Research over the last decade has revealed that cell cycle abnormalities also represent a major neuropathological feature of AD. These abnormalities appear very early in the disease process, prior to the appearance of plaques and tangles. Growing evidence suggests that neuronal cell cycle regulatory failure, leading to apoptosis, may be a significant component of the pathogenesis of AD. A number of signaling pathways with the potential to activate aberrant cell cycle re-entry in AD have been described. The relationships among these signaling cascades, which involve the amyloid precursor protein (APP), cyclin-dependent kinases (cdks), and the cell cycle protein Pin1, have not yet been fully elucidated, but details of the individual pathways are beginning to emerge. This review summarizes the current state of knowledge with respect to specific neuronal signaling events that are thought to underlie cell cycle regulatory failure in AD brain. The elements of these pathways that represent potential new therapeutic targets for AD are described. Drugs and peptides that can inhibit molecular steps leading to AD neurodegeneration by intervening in the activation of cell cycle re-entry in neurons represent an entirely new approach to the development of treatments for AD.
Collapse
Affiliation(s)
- Rachael L Neve
- Department of Psychiatry, MRC 223, Harvard Medical School and McLean Hospital, Belmont, MA 02478, USA.
| | | |
Collapse
|
41
|
Julien JP, Kriz J. Transgenic mouse models of amyotrophic lateral sclerosis. Biochim Biophys Acta Mol Basis Dis 2006; 1762:1013-24. [PMID: 16675207 DOI: 10.1016/j.bbadis.2006.03.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 03/21/2006] [Accepted: 03/21/2006] [Indexed: 12/11/2022]
Abstract
The discovery of missense mutations in the gene coding for the Cu/Zn superoxide dismutase 1 (SOD1) in subsets of familial cases was rapidly followed by the generation of transgenic mice expressing various forms of SOD1 mutants. The mice overexpressing high levels of mutant SOD1 mRNAs do develop motor neuron disease but unraveling the mechanisms of pathogenesis has been very challenging. Studies with mouse lines suggest that the toxicity of mutant SOD1 is unrelated to copper-mediated catalysis but rather to propensity of a subfraction of mutant SOD1 proteins to form misfolded protein species and aggregates. However, the mechanism of toxicity of SOD1 mutants remains to be elucidated. Involvement of cytoskeletal components in ALS pathogenesis is supported by several mouse models of motor neuron disease with neurofilament abnormalities and with genetic defects in microtubule-based transport. Here, we describe how transgenic mouse models have been used for understanding pathogenic pathways of motor neuron disease and for pre-clinical drug testing.
Collapse
Affiliation(s)
- Jean-Pierre Julien
- Research Centre of CHUL, Department of Anatomy and Physiology of Laval University, 2705 Boulevard Laurier, Quebec, QC, Canada G1V 4G2.
| | | |
Collapse
|
42
|
Hallows JL, Iosif RE, Biasell RD, Vincent I. p35/p25 is not essential for tau and cytoskeletal pathology or neuronal loss in Niemann-Pick type C disease. J Neurosci 2006; 26:2738-44. [PMID: 16525053 PMCID: PMC6675168 DOI: 10.1523/jneurosci.4834-05.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hyperactivation of the cyclin-dependent kinase 5 (cdk5), triggered by proteolytic conversion of its neuronal activator, p35, to a more potent byproduct, p25, has been implicated in Alzheimer's disease (AD), amyotrophic lateral sclerosis, and Niemann-Pick type C disease (NPC). This mechanism is thought to lead to the development of neuropathological hallmarks, i.e., hyperphosphorylated cytoskeletal proteins, neuronal inclusions, and neurodegeneration, that are common to all three diseases. This pathological ensemble is recapitulated in a single model, the npc-1 (npc(-/-)) mutant mouse. Previously, we showed that pharmacological cdk inhibitors dramatically reduced hyperphosphorylation, lesion formation, and locomotor defects in npc(-/-) mice, suggesting that cdk activity is required for NPC pathogenesis. Here, we used genetic ablation of the p35 gene to examine the specific involvement of p35, p25, and hence cdk5 activation in NPC neuropathogenesis. We found that lack of p35/p25 does not slow the onset or progression or improve the neuropathology of NPC. Our results provide direct evidence that p35/p25-mediated cdk5 deregulation is not essential for NPC pathology and suggest that similar pathology in AD may also be cdk5 independent.
Collapse
|
43
|
Sen A, Thom M, Martinian L, Jacobs T, Nikolic M, Sisodiya SM. Deregulation of cdk5 in Hippocampal sclerosis. J Neuropathol Exp Neurol 2006; 65:55-66. [PMID: 16410749 DOI: 10.1097/01.jnen.0000195940.48033.a2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hippocampal sclerosis (HS) is the most common cause of chronic medically refractory epilepsy in adults. Histologically, HS is characterized by segmental neuronal loss and gliosis. Although neuronal loss is important to the pathophysiology of HS, the molecular mechanisms underlying the neuronal loss remain uncertain. Recently, it has been appreciated that proteins important in neurodevelopment may also have a role in neurodegeneration. Cyclin-dependent kinase 5 (cdk5), known to be crucial in development of the normal cerebral cortex, has now been shown as pivotal in several cell death paradigms, including apoptosis and necrosis. Deregulation of cdk5 by p25 causes hyperphosphorylation of tau and may contribute to pathology in several neurodegenerative conditions. Furthermore, it has been shown that after transient forebrain ischemia, cdk5 causes specific death of CA1 neurons in the rat hippocampus by direct phosphorylation of the NR2A subunit of the NMDA receptor and subsequent excitotoxicity. Because apoptosis, necrosis, and excitotoxicity are all thought to contribute to neuronal loss in HS, we hypothesized that abnormalities of the cdk5 pathway would accompany this disorder. Surgically resected cases of HS with adjacent histologically normal lateral temporal cortex were examined for cdk5 and its activator p35/p25. We consistently found increased immunoreactivity for p35/p25 in surviving neurons within areas of neuronal loss compared with areas where neurons were preserved. Western blots showed the ratio of p25 to p35 to be greater in diseased hippocampi than in the adjacent histologically normal temporal lobe. Histone-based kinase assays demonstrated increased activity of the p25-cdk5 complex in HS compared with the temporal lobe despite neuronal loss in the hippocampal samples. Our results suggest that p25 is pathologically increased in HS and that deregulation of cdk5 by p25 might contribute to neuronal death in this condition.
Collapse
Affiliation(s)
- Arjune Sen
- Department of Clinical and Experimental Epilepsy, The Institute of Neurology, University College London, Queen Square, London, U.K
| | | | | | | | | | | |
Collapse
|
44
|
Giese KP, Ris L, Plattner F. Is there a role of the cyclin-dependent kinase 5 activator p25 in Alzheimer's disease? Neuroreport 2006; 16:1725-30. [PMID: 16237316 DOI: 10.1097/01.wnr.0000185019.67434.d2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sporadic Alzheimer's disease is the leading cause of dementia, but the underlying molecular processes are still unknown. Several studies have observed an accumulation of the protein fragment p25 in sporadic Alzheimer's disease brain. p25 derives from proteolysis of p35, and overactivates the tau kinase cyclin-dependent kinase 5. Transgenic mice expressing high levels of p25 exhibit hyperphosphorylation of tau as seen in Alzheimer's disease, and neurodegeneration. In contrast, low-level p25 expression, less than half of endogenous p35 expression, has a sex-specific effect on hippocampal synaptic plasticity and improves spatial learning in female but not in male mice. Therefore, p25 formation may initially be a compensatory response for early learning deficits in Alzheimer's disease, but continued formation could contribute to detrimental changes in Alzheimer's disease.
Collapse
Affiliation(s)
- K Peter Giese
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| | | | | |
Collapse
|
45
|
Affiliation(s)
- Jonathan C Cruz
- Department of Pathology, Howard Hughes Medical Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | | |
Collapse
|
46
|
Song MS, Saavedra L, de Chaves EIP. Apoptosis is secondary to non-apoptotic axonal degeneration in neurons exposed to Abeta in distal axons. Neurobiol Aging 2005; 27:1224-38. [PMID: 16122841 DOI: 10.1016/j.neurobiolaging.2005.06.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Revised: 06/09/2005] [Accepted: 06/10/2005] [Indexed: 11/26/2022]
Abstract
The goal of this study was to assess if neurons exposed to amyloid-beta peptide (Abeta) exclusively in distal axons, undergo apoptosis. This is relevant to the loss of cholinergic neurons in Alzheimer's disease. Using a three-compartmented culture system for rat sympathetic neurons, we demonstrate that exposure of axons to Abeta1-42 activates an independent destruction program in axons, which leads to nuclear apoptosis. Abeta-induced axonal degeneration does not involve local caspase activation, but causes caspase activation in cell bodies. Accordingly, inhibition of caspase activation blocks Abeta-induced apoptosis but not axonal degeneration. In agreement with previous suggestions that disruption of nerve growth factor (NGF)-mediated signaling might contribute to the loss of cholinergic neurons, we found that provision of NGF to cell bodies protects sympathetic neurons from Abeta-induced apoptosis. However, our data indicate that Abeta-induced axonal degeneration follows a mechanism different than that activated by NGF withdrawal. Only Abeta-induced axonal degeneration is prevented by the calpain inhibitor calpastatin and is insensitive to the inhibitor of the ubiquitin-proteasome system MG132. Importantly, inhibition of Abeta-induced axonal degeneration by calpastatin prevents nuclear apoptosis.
Collapse
Affiliation(s)
- Mee-Sook Song
- Centre for Alzheimer and Neurodegenerative Research, Signal Transduction Research Group and Department of Pharmacology, 928 Medical Science Building, Faculty of Medicine, University of Alberta, Edmonton, Alta., Canada T6G 2H7
| | | | | |
Collapse
|
47
|
Ris L, Angelo M, Plattner F, Capron B, Errington ML, Bliss TVP, Godaux E, Giese KP. Sexual dimorphisms in the effect of low-level p25 expression on synaptic plasticity and memory. Eur J Neurosci 2005; 21:3023-33. [PMID: 15978013 DOI: 10.1111/j.1460-9568.2005.04137.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
p25, a degradation product of p35, has been reported to accumulate in the forebrain of patients with Alzheimer's disease. p25 as well as p35 are activators of cyclin-dependent kinase 5 (Cdk5) although p25/Cdk5 and p35/Cdk5 complexes have distinct properties. Several mouse models with high levels of p25 expression exhibit signs of neurodegeneration. On the contrary, we have shown that low levels of p25 expression do not cause neurodegeneration and are even beneficial for particular types of learning and memory [Angelo et al., (2003) Eur J. Neurosci., 18, 423-431]. Here, we have studied the influence of low-level p25 expression in hippocampal synaptic plasticity and in learning and memory for each sex separately in two different genetic backgrounds (129B6F1 and C57BL/6). Surprisingly, we found that low-level p25 expression had different consequences in male and female mutants. In the two genetic backgrounds LTP induced by a strong stimulation of the Schaffer's collaterals (four trains, 1-s duration, 5-min interval) was severely impaired in male, but not in female, p25 mutants. Furthermore, in the two genetic backgrounds spatial learning in the Morris water maze was faster in female p25 mutants than in male transgenic mice. These results suggest that, in women, the production of p25 in Alzheimer's disease could be a compensation for some early learning and memory deficits.
Collapse
Affiliation(s)
- L Ris
- Laboratory of Neurosciences, University of Mons-Hainaut, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hamdane M, Bretteville A, Sambo AV, Schindowski K, Bégard S, Delacourte A, Bertrand P, Buée L. p25/Cdk5-mediated retinoblastoma phosphorylation is an early event in neuronal cell death. J Cell Sci 2005; 118:1291-8. [PMID: 15741232 DOI: 10.1242/jcs.01724] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
In large models of neuronal cell death, there is a tight correlation between Cdk5 deregulation and cell-cycle dysfunction. However, pathways that link Cdk5 to the cell cycle during neuronal death are still unclear. We have investigated the molecular events that precede p25/Cdk5-triggered neuronal death using a neuronal cell line that allows inducible p25 expression. In this system, no sign of apoptosis was seen before 24 hours of p25 induction. Thus, at that time, cell-cycle-regulatory proteins were analysed by immunoblotting and some of them showed a significant deregulation. Interestingly, after time-course experiments, the earliest feature correlated with p25 expression was the phosphorylation of the retinoblastoma protein (Rb). Indeed, this phosphorylation was observed 6 hours after p25 induction and was abolished in the presence of a Cdk5 inhibitor, roscovitine, which does not inhibit the usual Rb cyclin-D kinases Cdk4 and Cdk6. Furthermore, analyses of levels and subcellular localization of Cdk-related cyclins did not reveal any change following Cdk5 activation, arguing for a direct effect of Cdk5 activity on Rb protein. This latter result was clearly demonstrated by in vitro kinase assays showing that the p25-Cdk5 complex in our cell system phosphorylates Rb directly without the need for any intermediary kinase activity. Hence, Rb might be an appropriate candidate that connects Cdk5 to cell-cycle deregulation during neuronal cell death.
Collapse
Affiliation(s)
- Malika Hamdane
- INSERM U422, Institut de Médecine Prédictive et Recherche Thérapeutique, Université de Lille 2, Place de Verdun, 59045 Lille Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Stoothoff WH, Johnson GVW. Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta Mol Basis Dis 2005; 1739:280-97. [PMID: 15615646 DOI: 10.1016/j.bbadis.2004.06.017] [Citation(s) in RCA: 320] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Accepted: 06/02/2004] [Indexed: 11/24/2022]
Abstract
The microtubule-associated protein tau, abundant in neurons, has gained notoriety due to the fact that it is deposited in cells as fibrillar lesions in numerous neurodegenerative diseases, and most notably Alzheimer's disease. Regulation of microtubule dynamics is the most well-recognized function of tau, but it is becoming increasingly evident that tau plays additional roles in the cell. The functions of tau are regulated by site-specific phosphorylation events, which if dysregulated, as they are in the disease state, result in tau dysfunction and mislocalization, which is potentially followed by tau polymerization, neuronal dysfunction and death. Given the increasing evidence that a disruption in the normal phosphorylation state of tau plays a key role in the pathogenic events that occur in Alzheimer's disease and other neurodegenerative conditions, it is of crucial importance that the protein kinases and phosphatases that regulate tau phosphorylation in vivo as well as the signaling cascades that regulate them be identified. This review focuses on recent literature pertaining to the regulation of tau phosphorylation and function in cell culture and animal model systems, and the role that a dysregulation of tau phosphorylation may play in the neuronal dysfunction and death that occur in neurodegenerative diseases that have tau pathology.
Collapse
Affiliation(s)
- William H Stoothoff
- Department of Psychiatry, School of Medicine, University of Alabama at Birmingham, 1061 Sparks Center, 1720 7th Avenue South, Birmingham, AL 35294-0017, USA
| | | |
Collapse
|
50
|
Zhang M, Li J, Chakrabarty P, Bu B, Vincent I. Cyclin-dependent kinase inhibitors attenuate protein hyperphosphorylation, cytoskeletal lesion formation, and motor defects in Niemann-Pick Type C mice. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:843-53. [PMID: 15331409 PMCID: PMC1618588 DOI: 10.1016/s0002-9440(10)63347-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dysregulation of cyclin-dependent kinases (cdks) and cytoskeletal protein hyperphosphorylation characterizes a subset of human neurodegenerative diseases, including Alzheimer's disease, amyotrophic lateral sclerosis, and Niemann-Pick Type C (NPC). It is thought that these cytoskeletal changes lead eventually to development of hallmark cytoskeletal lesions such as neurofibrillary tangles and axonal spheroids. Although many studies support an involvement of cdks in these neurodegenerative cascades, it is not known whether cdk activity is essential. The naturally occurring npc-1 mutant mouse mimics human NPC, in displaying activation of cdk5, mitotic cdc2, and cdk4, with concomitant cytoskeletal pathology and neurodegeneration. We availed of this model and specific pharmacological inhibitors of cdk activity, to determine whether cdks are necessary for NPC neuropathology. The inhibitors were infused intracerebroventricularly for a 2-week period, initiated at a pathologically incipient stage. While an inactive stereoisomer, iso-olomoucine, was ineffective, two potent inhibitors, roscovitine and olomoucine, attenuated significantly the hyperphosphorylation of neurofilament, tau, and mitotic proteins, reduced the number of spheroids, modulated Purkinje neuron death, and ameliorated motor defects in npc mice. These results suggest that cdk activity is required for neuropathology and subsequent motor impairment in NPC. Studies aimed at knocking down individual cdks in these mice will help identify the specific cdk(s) that are essential, and delineate their precise roles in the neurodegenerative process.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | | | | | | | | |
Collapse
|