151
|
Impaired Wnt signaling in dopamine containing neurons is associated with pathogenesis in a rotenone triggered Drosophila Parkinson's disease model. Sci Rep 2018; 8:2372. [PMID: 29403026 PMCID: PMC5799194 DOI: 10.1038/s41598-018-20836-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/25/2018] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease, which is the one of the most common neurodegenerative movement disorder, is characterized by a progressive loss of dopamine containing neurons. The mechanisms underlying disease initiation and development are not well understood and causative therapies are currently not available. To elucidate the molecular processes during early stages of Parkinson’s disease, we utilized a Drosophila model. To induce Parkinson’s disease-like phenotypes, we treated flies with the pesticide rotenone and isolated dopamine producing neurons of animals that were at an early disease stage. Transcriptomic analyses revealed that gene ontologies associated with regulation of cell death and neuronal functions were significantly enriched. Moreover, the activities of the MAPK/EGFR- and TGF-β signaling pathways were enhanced, while the Wnt pathway was dampened. In order to evaluate the role of Wnt signaling for survival of dopaminergic neurons in the disease model, we rescued the reduced Wnt signaling activity by ectopic overexpression of armadillo/β-catenin. This intervention rescued the rotenone induced movement impairments in the Drosophila model. Taken together, this initial study showed a highly relevant role of Wnt signaling for dopamine producing neurons during pathogenesis in Parkinson’s disease and it implies that interfering with this pathway might by a suitable therapeutic option for the future.
Collapse
|
152
|
Tapia-Rojas C, Inestrosa NC. Wnt signaling loss accelerates the appearance of neuropathological hallmarks of Alzheimer's disease in J20-APP transgenic and wild-type mice. J Neurochem 2018; 144:443-465. [DOI: 10.1111/jnc.14278] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/29/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Cheril Tapia-Rojas
- Centro de Envejecimiento y Regeneración (CARE UC); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
- Centre for Healthy Brain Ageing; School of Psychiatry; Faculty of Medicine; University of New South Wales; Sydney New South Wales Australia
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA); Universidad de Magallanes; Punta Arenas Chile
| |
Collapse
|
153
|
Mastrodonato A, Barbati SA, Leone L, Colussi C, Gironi K, Rinaudo M, Piacentini R, Denny CA, Grassi C. Olfactory memory is enhanced in mice exposed to extremely low-frequency electromagnetic fields via Wnt/β-catenin dependent modulation of subventricular zone neurogenesis. Sci Rep 2018; 8:262. [PMID: 29321633 PMCID: PMC5762682 DOI: 10.1038/s41598-017-18676-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/15/2017] [Indexed: 12/03/2022] Open
Abstract
Exposure to extremely low-frequency electromagnetic fields (ELFEF) influences the expression of key target genes controlling adult neurogenesis and modulates hippocampus-dependent memory. Here, we assayed whether ELFEF stimulation affects olfactory memory by modulating neurogenesis in the subventricular zone (SVZ) of the lateral ventricle, and investigated the underlying molecular mechanisms. We found that 30 days after the completion of an ELFEF stimulation protocol (1 mT; 50 Hz; 3.5 h/day for 12 days), mice showed enhanced olfactory memory and increased SVZ neurogenesis. These effects were associated with upregulated expression of mRNAs encoding for key regulators of adult neurogenesis and were mainly dependent on the activation of the Wnt pathway. Indeed, ELFEF stimulation increased Wnt3 mRNA expression and nuclear localization of its downstream target β-catenin. Conversely, inhibition of Wnt3 by Dkk-1 prevented ELFEF-induced upregulation of neurogenic genes and abolished ELFEF’s effects on olfactory memory. Collectively, our findings suggest that ELFEF stimulation increases olfactory memory via enhanced Wnt/β-catenin signaling in the SVZ and point to ELFEF as a promising tool for enhancing SVZ neurogenesis and olfactory function.
Collapse
Affiliation(s)
- Alessia Mastrodonato
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy.,Columbia University, Department of Psychiatry, New York, NY, 10032, USA.,Research Foundation for Mental Hygiene Inc. (RFMH), Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | | | - Lucia Leone
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy
| | - Claudia Colussi
- CNR, Institute of Cell Biology and Neurobiology, Monterotondo (RM), 00015, Italy
| | - Katia Gironi
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy
| | - Marco Rinaudo
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy
| | - Roberto Piacentini
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy
| | - Christine A Denny
- Columbia University, Department of Psychiatry, New York, NY, 10032, USA.,Research Foundation for Mental Hygiene Inc. (RFMH), Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Claudio Grassi
- Università Cattolica del Sacro Cuore, Institute of Human Physiology, Rome, 00168, Italy. .,Fondazione Policlinico Universitario A. Gemelli, Rome, 00168, Italy.
| |
Collapse
|
154
|
Corrêa-Velloso JC, Gonçalves MC, Naaldijk Y, Oliveira-Giacomelli Á, Pillat MM, Ulrich H. Pathophysiology in the comorbidity of Bipolar Disorder and Alzheimer's Disease: pharmacological and stem cell approaches. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:34-53. [PMID: 28476640 DOI: 10.1016/j.pnpbp.2017.04.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022]
Abstract
Neuropsychiatric disorders involve various pathological mechanisms, resulting in neurodegeneration and brain atrophy. Neurodevelopmental processes have shown to be critical for the progression of those disorders, which are based on genetic and epigenetic mechanisms as well as on extrinsic factors. We review here common mechanisms underlying the comorbidity of Bipolar Disorders and Alzheimer's Disease, such as aberrant neurogenesis and neurotoxicity, reporting current therapeutic approaches. The understanding of these mechanisms precedes stem cell-based strategies as a new therapeutic possibility for treatment and prevention of Bipolar and Alzheimer's Disease progression. Taking into account the difficulty of studying the molecular basis of disease progression directly in patients, we also discuss the importance of stem cells for effective drug screening, modeling and treating psychiatric diseases, once in vitro differentiation of patient-induced pluripotent stem cells provides relevant information about embryonic origins, intracellular pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Juliana C Corrêa-Velloso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Maria Cb Gonçalves
- Departamento de Neurologia e Neurociências, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, São Paulo, SP 04039-032, Brazil
| | - Yahaira Naaldijk
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Ágatha Oliveira-Giacomelli
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
155
|
Amyloid β synaptotoxicity is Wnt-PCP dependent and blocked by fasudil. Alzheimers Dement 2017; 14:306-317. [PMID: 29055813 PMCID: PMC5869054 DOI: 10.1016/j.jalz.2017.09.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/13/2017] [Accepted: 09/07/2017] [Indexed: 01/18/2023]
Abstract
Introduction Synapse loss is the structural correlate of the cognitive decline indicative of dementia. In the brains of Alzheimer's disease sufferers, amyloid β (Aβ) peptides aggregate to form senile plaques but as soluble peptides are toxic to synapses. We previously demonstrated that Aβ induces Dickkopf-1 (Dkk1), which in turn activates the Wnt–planar cell polarity (Wnt-PCP) pathway to drive tau pathology and neuronal death. Methods We compared the effects of Aβ and of Dkk1 on synapse morphology and memory impairment while inhibiting or silencing key elements of the Wnt-PCP pathway. Results We demonstrate that Aβ synaptotoxicity is also Dkk1 and Wnt-PCP dependent, mediated by the arm of Wnt-PCP regulating actin cytoskeletal dynamics via Daam1, RhoA and ROCK, and can be blocked by the drug fasudil. Discussion Our data add to the importance of aberrant Wnt signaling in Alzheimer's disease neuropathology and indicate that fasudil could be repurposed as a treatment for the disease. Aβ synaptotoxicity is Dickkopf-1 and Wnt-PCP dependent. The Wnt-PCP pathway drives Aβ-driven synapse loss via RhoA and ROCK. ROCK inhibitor fasudil blocks Aβ-driven synapse loss and cognitive impairment. Fasudil should be assessed for repurposing for Alzheimer's disease.
Collapse
|
156
|
McLeod F, Marzo A, Podpolny M, Galli S, Salinas P. Evaluation of Synapse Density in Hippocampal Rodent Brain Slices. J Vis Exp 2017. [PMID: 29053699 PMCID: PMC5752395 DOI: 10.3791/56153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In the brain, synapses are specialized junctions between neurons, determining the strength and spread of neuronal signaling. The number of synapses is tightly regulated during development and neuronal maturation. Importantly, deficits in synapse number can lead to cognitive dysfunction. Therefore, the evaluation of synapse number is an integral part of neurobiology. However, as synapses are small and highly compact in the intact brain, the assessment of absolute number is challenging. This protocol describes a method to easily identify and evaluate synapses in hippocampal rodent slices using immunofluorescence microscopy. It includes a three-step procedure to evaluate synapses in high-quality confocal microscopy images by analyzing the co-localization of pre- and postsynaptic proteins in hippocampal slices. It also explains how the analysis is performed and gives representative examples from both excitatory and inhibitory synapses. This protocol provides a solid foundation for the analysis of synapses and can be applied to any research investigating the structure and function of the brain.
Collapse
Affiliation(s)
- Faye McLeod
- Department of Cell and Developmental Biology, University College London;
| | - Aude Marzo
- Department of Cell and Developmental Biology, University College London;
| | - Marina Podpolny
- Department of Cell and Developmental Biology, University College London
| | - Soledad Galli
- Department of Cell and Developmental Biology, University College London
| | - Patricia Salinas
- Department of Cell and Developmental Biology, University College London
| |
Collapse
|
157
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Effects of cannabidiol interactions with Wnt/β-catenin pathway and PPARγ on oxidative stress and neuroinflammation in Alzheimer's disease. Acta Biochim Biophys Sin (Shanghai) 2017; 49:853-866. [PMID: 28981597 DOI: 10.1093/abbs/gmx073] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, in which the primary etiology remains unknown. AD presents amyloid beta (Aβ) protein aggregation and neurofibrillary plaque deposits. AD shows oxidative stress and chronic inflammation. In AD, canonical Wingless-Int (Wnt)/β-catenin pathway is downregulated, whereas peroxisome proliferator-activated receptor γ (PPARγ) is increased. Downregulation of Wnt/β-catenin, through activation of glycogen synthase kinase-3β (GSK-3β) by Aβ, and inactivation of phosphatidylinositol 3-kinase/Akt signaling involve oxidative stress in AD. Cannabidiol (CBD) is a non-psychotomimetic phytocannabinoid from Cannabis sativa plant. In PC12 cells, Aβ-induced tau protein hyperphosphorylation is inhibited by CBD. This inhibition is associated with a downregulation of p-GSK-3β, an inhibitor of Wnt pathway. CBD may also increase Wnt/β-catenin by stimulation of PPARγ, inhibition of Aβ and ubiquitination of amyloid precursor protein. CBD attenuates oxidative stress and diminishes mitochondrial dysfunction and reactive oxygen species generation. CBD suppresses, through activation of PPARγ, pro-inflammatory signaling and may be a potential new candidate for AD therapy.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France
| | | | - Rémy Guillevin
- Université de Poitiers et CHU de Poitiers, DACTIM, Laboratoire de Mathématiques et Applications, UMR CNRS 7348, SP2MI, Futuroscope, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France
- CHU Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
158
|
Gunn‐Moore D, Kaidanovich‐Beilin O, Iradi MCG, Gunn‐Moore F, Lovestone S. Alzheimer's disease in humans and other animals: A consequence of postreproductive life span and longevity rather than aging. Alzheimers Dement 2017; 14:195-204. [DOI: 10.1016/j.jalz.2017.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/19/2017] [Accepted: 08/19/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Danièlle Gunn‐Moore
- University of Edinburgh Royal (Dick) School of Veterinary Studies and The Roslin Institute Easter Bush Campus Roslin UK
| | | | - María Carolina Gallego Iradi
- University of Florida, College of Medicine Department of Neuroscience, Center for Translational Research in Neurodegenerative Diseases Gainesville FL USA
| | | | | |
Collapse
|
159
|
Zhou L, Chen D, Huang XM, Long F, Cai H, Yao WX, Chen ZC, Liao ZJ, Deng ZZ, Tan S, Shan YL, Cai W, Wang YG, Yang RH, Jiang N, Peng T, Hong MF, Lu ZQ. Wnt5a Promotes Cortical Neuron Survival by Inhibiting Cell-Cycle Activation. Front Cell Neurosci 2017; 11:281. [PMID: 29033786 PMCID: PMC5626855 DOI: 10.3389/fncel.2017.00281] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/30/2017] [Indexed: 01/11/2023] Open
Abstract
β-Amyloid protein (Aβ) is thought to cause neuronal loss in Alzheimer’s disease (AD). Aβ treatment promotes the re-activation of a mitotic cycle and induces rapid apoptotic death of neurons. However, the signaling pathways mediating cell-cycle activation during neuron apoptosis have not been determined. We find that Wnt5a acts as a mediator of cortical neuron survival, and Aβ42 promotes cortical neuron apoptosis by downregulating the expression of Wnt5a. Cell-cycle activation is mediated by the reduced inhibitory effect of Wnt5a in Aβ42 treated cortical neurons. Furthermore, Wnt5a signals through the non-canonical Wnt/Ca2+ pathway to suppress cyclin D1 expression and negatively regulate neuronal cell-cycle activation in a cell-autonomous manner. Together, aberrant downregulation of Wnt5a signaling is a crucial step during Aβ42 induced cortical neuron apoptosis and might contribute to AD-related neurodegeneration.
Collapse
Affiliation(s)
- Li Zhou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Rehabilitation, The First Affiliated Hospital of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Di Chen
- Laboratory of Viral Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Sino-French Hoffmann Institute of Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xu-Ming Huang
- Department of Rehabilitation, The First Affiliated Hospital of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Fei Long
- Laboratory of Viral Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Sino-French Hoffmann Institute of Immunology, Guangzhou Medical University, Guangzhou, China
| | - Hua Cai
- Laboratory of Viral Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Sino-French Hoffmann Institute of Immunology, Guangzhou Medical University, Guangzhou, China
| | - Wen-Xia Yao
- Laboratory of Viral Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Sino-French Hoffmann Institute of Immunology, Guangzhou Medical University, Guangzhou, China
| | - Zhong-Cheng Chen
- Department of Laboratory, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Zhe-Zhi Deng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sha Tan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi-Long Shan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Cai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu-Ge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ri-Hong Yang
- Department of Pathology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Jiang
- Department of Hepatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tao Peng
- Laboratory of Viral Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Sino-French Hoffmann Institute of Immunology, Guangzhou Medical University, Guangzhou, China
| | - Ming-Fan Hong
- Department of Neurology, The First Affiliated Hospital of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zheng-Qi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
160
|
Jenwitheesuk A, Park S, Wongchitrat P, Tocharus J, Mukda S, Shimokawa I, Govitrapong P. Comparing the Effects of Melatonin with Caloric Restriction in the Hippocampus of Aging Mice: Involvement of Sirtuin1 and the FOXOs Pathway. Neurochem Res 2017; 43:153-161. [PMID: 28770437 DOI: 10.1007/s11064-017-2369-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
Abstract
It has been suggested that age-related neurodegeneration might be associated with neuropeptide Y (NPY); sirtuin1 (SIRT1) and forkhead box transcription factors O subfamily (FOXOs) pathways. Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent associated with the SIRT1-FOXOs pathway. This study aimed to compare the effects of melatonin (Mel) and caloric restriction (CR) on the expression of Sirt1, FoxO1, FoxO3a and FOXOs target genes in the aging mouse hippocampus. Neuropeptide Y-knockout (NpyKO) and wild-type (WT) male mice aged 19 months were previously treated either with food ad libitum or CR for 16 months. WT old animals were divided into four groups: control, CR, Mel and CR+Mel treated groups. The Mel and CR+Mel were treated with melatonin 10 mg/kg, daily, subcutaneously for 7 consecutive days. Mel treatment upregulated the mRNA expression of Sirt1, FOXOs (FoxO1 and FoxO3a) target genes that regulated the cell cycle [e.g., cyclin-dependent kinase inhibitor 1B (p27)], Wingless and INT-1 (Wnt1) and inducible signaling pathway protein 1 (Wisp1) in the aged mouse hippocampus. CR treatment also showed the similar actions. However, the mRNA expression of Sirt1, FoxO1, FoxO3a, p27 or Wisp1 did not alter in the CR+Mel group when compared with CR or Mel group. Melatonin could not produce any additive effect on the CR treatment group, suggesting that both treatments mimicked the effect, possibly via the same pathway. NPY which mediates physiological adaptations to energy deficits is an essential link between CR and longevity in mice. In order to focus on the role of Npy in mediating the effects of melatonin, the gene expression between NpyKO and WT male mice were compared. Our data showed that, in the absence of Npy, melatonin could not mediate effects on those gene expressions, suggesting that Npy was required for melatonin to mediate the effect, possibly, on life extension.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Seongjoon Park
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Isao Shimokawa
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan.
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand. .,Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Salaya, Thailand. .,Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Lak Si, Bangkok, 10210, Thailand.
| |
Collapse
|
161
|
García-Velázquez L, Arias C. The emerging role of Wnt signaling dysregulation in the understanding and modification of age-associated diseases. Ageing Res Rev 2017. [PMID: 28624530 DOI: 10.1016/j.arr.2017.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wnt signaling is a highly conserved pathway that participates in multiple aspects of cellular function during development and in adults. In particular, this pathway has been implicated in cell fate determination, proliferation and cell polarity establishment. In the brain, it contributes to synapse formation, axonal remodeling, dendrite outgrowth, synaptic activity, neurogenesis and behavioral plasticity. The expression and distribution of Wnt components in different organs vary with age, which may have important implications for preserving tissue homeostasis. The dysregulation of Wnt signaling has been implicated in age-associated diseases, such as cancer and some neurodegenerative conditions. This is a relevant research topic, as an important research avenue for therapeutic targeting of the Wnt pathway in regenerative medicine has recently been opened. In this review, we discuss the recent findings on the regulation of Wnt components during aging, particularly in brain functioning, and the implications of Wnt signaling in age-related diseases.
Collapse
|
162
|
Sodium selenate activated Wnt/β-catenin signaling and repressed amyloid-β formation in a triple transgenic mouse model of Alzheimer's disease. Exp Neurol 2017; 297:36-49. [PMID: 28711506 DOI: 10.1016/j.expneurol.2017.07.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/21/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
Abstract
Accumulating evidences show that selenium dietary intake is inversely associated with the mortality of Alzheimer's disease (AD). Sodium selenate has been reported to reduce neurofibrillary tangles (NFT) in the tauopathic mouse models, but its effects on the Wnt/β-catenin signaling pathway and APP processing remain unknown during AD formation. In this paper, triple transgenic AD mice (3×Tg-AD) had been treated with sodium selenate in drinking water for 10month before the detection of hippocampal pathology. Increased Aβ generation, tau hyperphosphorylation and neuronal apoptosis were found in the hippocampus of AD model mouse. Down-regulation of Wnt/β-catenin signaling is closely associated with the alteration of AD pathology. Treatment with sodium selenate significantly promoted the activity of protein phosphatases of type 2A (PP2A) and repressed the hallmarks of AD. Activation of PP2A by sodium selenate could increase active β-catenin level and inhibit GSK3β activity in the hippocampal tissue and primarily cultured neurons of AD model mouse, leading to activation of Wnt/β-catenin signaling and transactivation of target genes, including positively-regulated genes c-myc, survivin, TXNRD2 and negatively-regulated gene BACE1. Meanwhile, APP phosphorylation was also reduced on the Thr668 residue after selenate treatment, causing the decreases of APP cleavage and Aβ generation. These findings reveal that the Wnt/β-catenin signaling is a potential target for prevention of AD and sodium selenate may be developed as a new drug for AD treatment.
Collapse
|
163
|
Abstract
Wnt signals regulate cell proliferation, migration and differentiation during development, as well as synaptic transmission and plasticity in the adult brain. Abnormal Wnt signaling is central to a number of brain pathologies. We review here, the significance of this pathway focused in the contribution of the most frequent alterations in receptors, secretable modulators and downstream targets in Alzheimer's disease (AD) and Glioblastoma (GBM). β-catenin and GSK3 levels are pivotal in the neurodegeneration associated to AD contributing to memory deficits, tau phosphorylation, increased β-amyloid production and modulation of Apolipoprotein E in the brain. In consequence, β-catenin and GSK3 are targets for potential treatments in AD. Also, Wnt pathway components and secreted molecules interfering with this signaling contribute to the progression of tumoral cells. Wnt pathway activation is a bad prognosis in brain cancer; however, mutations in WNT or Frizzled (FZD) genes do not account for the cases of GBM. Instead, recent studies indicate that epigenetic modifications contribute to the development of GBMs opening novel strategies to study GBM progression.
Collapse
|
164
|
Plasma dickkopf-related protein 1, an antagonist of the Wnt pathway, is associated with HIV-associated neurocognitive impairment. AIDS 2017; 31:1379-1385. [PMID: 28358733 DOI: 10.1097/qad.0000000000001481] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Dickkopf-related protein 1 (DKK1) is a soluble antagonist of the Wningless (Wnt) pathway. It binds to and sequesters low-density lipoprotein receptor-related proteins 5/6 away from Wnts. Because the Wnt pathway regulates synaptic transmission and plasticity, we hypothesized that increased DKK1 would increase the risk for neurocognitive impairment (NCI) in HIV-positive (HIV) individuals. We evaluated, here, the relationship between plasma DKK1 and global NCI. METHODS Plasma samples and data from 41 HIV to 42 HIV adults were obtained from the University of California, San Diego, California, USA. Concentrations of DKK1 and a comparator protein, monocyte chemoattractant protein-1 (MCP-1), were quantified in plasma by immunoassay. All study participants completed a standardized comprehensive neuropsychological test battery and their performance was summarized using the global deficit score method. RESULTS A higher DKK1 level was associated with NCI among HIV participants (d = 0.63, P = 0.05), particularly among the 26 participants whose plasma HIV RNA level was suppressed (d = 0.74, P = 0.08). DKK1 level was not associated with NCI among HIV participants (P = 0.98). was not associated with NCI in either group. In HIV adults with suppressed plasma HIV RNA, a receiver operator characteristic curve identified that a DKK1 level of at least 735 pg/ml had a positive predictive value of 83.3% for a diagnosis of NCI. This association did not weaken after accounting for the effect of AIDS, nadir CD4 T-cell count, addictive drug use, or demographic characteristics. CONCLUSION DKK1 is a specific biomarker for NCI in HIV adults, implicating the Wnt pathway in HIV neuropathogenesis.
Collapse
|
165
|
Tapia-Rojas C, Burgos PV, Inestrosa NC. Inhibition of Wnt signaling induces amyloidogenic processing of amyloid precursor protein and the production and aggregation of Amyloid-β (Aβ) 42 peptides. J Neurochem 2017; 139:1175-1191. [PMID: 27778356 DOI: 10.1111/jnc.13873] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/05/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and the most frequent cause of dementia in the aged population. According to the amyloid hypothesis, the amyloid-β (Aβ) peptide plays a key role in the pathogenesis of AD. Aβ is generated from the amyloidogenic processing of amyloid precursor protein and can aggregate to form oligomers, which have been described as a major synaptotoxic agent in neurons. Dysfunction of Wnt signaling has been linked to increased Aβ formation; however, several other studies have argued against this possibility. Herein, we use multiple experimental approaches to confirm that the inhibition of Wnt signaling promoted the amyloidogenic proteolytic processing of amyloid precursor protein. We also demonstrate that inhibiting Wnt signaling increases the production of the Aβ42 peptide, the Aβ42 /Aβ40 ratio, and the levels of Aβ oligomers such as trimers and tetramers. Moreover, we show that activating Wnt signaling reduces the levels of Aβ42 and its aggregates, increases Aβ40 levels, and reduces the Aβ42 /Aβ40 ratio. Finally, we show that the protective effects observed in response to activation of the Wnt pathway rely on β-catenin-dependent transcription, which is demonstrated experimentally via the expression of various 'mutant forms of β-catenin'. Together, our findings indicate that loss of the Wnt signaling pathway may contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Cheril Tapia-Rojas
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia V Burgos
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Sydney, Australia
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
166
|
Wang J, Gu J, Wu H, Zhu G, Feng D, Li Y, Guo W, Tian K, Gao G, Gao L. Pentazocine Protects SN4741 Cells Against MPP +-Induced Cell Damage via Up-Regulation of the Canonical Wnt/β-Catenin Signaling Pathway. Front Aging Neurosci 2017; 9:196. [PMID: 28659791 PMCID: PMC5469889 DOI: 10.3389/fnagi.2017.00196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/31/2017] [Indexed: 12/04/2022] Open
Abstract
The Wnt/β-catenin signaling pathway has been linked to many neurodegenerative diseases including Parkinson’s disease (PD). A glycoprotein named Dickkopf-1 (Dkk1) can combine with the receptor complex on cell membrane to inhibit Wnt/β-catenin signaling. Opioids, a series of compounds including morphine, fentanyl and pentazocine, have been reported to contribute to the up-regulation of Wnt/β-catenin signaling. Naloxone is an antagonist that has been used as an antidote to opioids through mu-opioid receptor. 1-methyl-4-phenylpyridinium (MPP+), which serves as a selective toxin for dopaminergic neurons, has been used to create experimental models of PD. In our study, we examined the protective effects of pentazocine against MPP+-induced cell death in the nigral dopaminergic cell line, SN4741 and tried to elucidate the molecular mechanisms underlying such protective effects. The data showed that pretreatment with pentazocine significantly rescued the SN4741 cell against MPP+. Moreover, the MPP+-exposed SN4741 cells exhibited a down-regulation of β-catenin, which could be restored by treatment with pentazocine. However, Dkk1 but not naloxonewas associated with the abrogation of protective effect of pentazocine. These results suggest that pentazocine alleviates MPP+-induced SN4741 cells apoptosis via the up-regulation of canonical Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Jintao Gu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical UniversityXi'an, China
| | - Hao Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Yuqian Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Keyong Tian
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical UniversityXi'an, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| |
Collapse
|
167
|
Prenatal high sucrose intake affected learning and memory of aged rat offspring with abnormal oxidative stress and NMDARs/Wnt signaling in the hippocampus. Brain Res 2017; 1669:114-121. [PMID: 28532855 DOI: 10.1016/j.brainres.2017.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/29/2017] [Accepted: 05/19/2017] [Indexed: 01/07/2023]
Abstract
Maternal over-nutrition may predispose offspring to obesity, type 2 diabetes and other adult diseases. The present study investigated long-term impact of prenatal high sucrose (HS) diets on cognitive capabilities in aged rat offspring. The fasting plasma glucose concentration did not differ between the control and HS groups. However, the fasting plasma insulin and insulin resistance index values were significantly increased in HS offspring that showed abnormal glucose tolerance test. HS offspring exhibited increased escape latency and swimming path length to the platform, and reduced time in the target quadrant and the number of crossing the platform, as compared with the control group. The expression of Grin2b/NR2B, Wnt2, Wnt3a and active form of β-catenin protein were decreased, and Dickkopf-related protein 1 was increased in the HS group. In addition, the levels of lipid peroxidation biomarker thiobarbituricacid reactive substance, nicotinamide adenine dinucleotide phosphate oxidases 2 and superoxide dismutase 1 were significantly increased, and the activity of catalase was decreased in the hippocampus in the HS group. The results demonstrate that prenatal HS-induced metabolic changes cause cognitive deficits in aged rat offspring, probably due to altered N-methyl-d-aspartate receptors/Wnt signaling and oxidative stress in the hippocampus.
Collapse
|
168
|
Jenwitheesuk A, Boontem P, Wongchitrat P, Tocharus J, Mukda S, Govitrapong P. Melatonin regulates the aging mouse hippocampal homeostasis via the sirtuin1-FOXO1 pathway. EXCLI JOURNAL 2017; 16:340-353. [PMID: 28507478 PMCID: PMC5427465 DOI: 10.17179/excli2016-852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 12/15/2022]
Abstract
Sirtuin1 (SIRT1) and forkhead box transcription factor O subfamily 1 (FOXO1) play vital roles in the maintenance of hippocampal neuronal homeostasis during aging. Our previous study showed that melatonin, a hormone mainly secreted by the pineal gland, restored the impaired memory of aged mice. Age-related neuronal energy deficits contribute to the pathogenesis of several neurodegenerative disorders. An attempt has been made to determine whether the effect of melatonin is mediated through the SIRT1-FOXO1 pathways. The present results showed that aged mice (22 months old) exhibited significantly downregulated SIRT1, FOXO1, and melatonin receptors MT1 and MT2 protein expression but upregulated tumor suppressor protein 53 (p53), acetyl-p53 protein (Ac-p53), mouse double minute 2 homolog (MDM2), Dickkopf-1 (DKK1) protein expression in mouse hippocampus compared with the young group. Melatonin treatment (10 mg/kg, daily in drinking water for 6 months) in aged mice significantly attenuated the age-induced downregulation of SIRT1, FOXO1, MT1 and MT2 protein expression and attenuated the age-induced increase in p53, ac-p53, MDM2, and DKK1 protein and mRNA expression. Melatonin decreased p53 and MDM2 expression, which led to a decrease in FOXO1 degradation. These present results suggest that melatonin may help the hippocampal neuronal homeostasis by increasing SIRT1, FOXO1 and melatonin receptors expression while decreasing DKK1 expression in the aging hippocampus. DKK1 can be induced by the accumulation of amyloid β (Aβ) which is the major hallmark of Alzheimer's disease.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand
| | - Parichart Boontem
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom 73170, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom, Thailand.,Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand.,Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
169
|
Neurosteroids Involvement in the Epigenetic Control of Memory Formation and Storage. Neural Plast 2016; 2016:5985021. [PMID: 28090360 PMCID: PMC5206442 DOI: 10.1155/2016/5985021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022] Open
Abstract
Memory is our ability to store and remember past experiences; it is the result of changes in neuronal circuits of specific brain areas as the hippocampus. During memory formation, neurons integrate their functions and increase the strength of their connections, so that synaptic plasticity is improved and consolidated. All these processes recruit several proteins at the synapses, whose expression is highly regulated by DNA methylation and histone tails posttranslational modifications. Steroids are known to influence memory process, and, among them, neurosteroids are implicated in neurodegenerative disease related to memory loss and cognitive impairment. The epigenetic control of neurosteroids involvement in memory formation and maintenance could represent the basis for neuroregenerative therapies.
Collapse
|
170
|
The age factor in axonal repair after spinal cord injury: A focus on neuron-intrinsic mechanisms. Neurosci Lett 2016; 652:41-49. [PMID: 27818358 DOI: 10.1016/j.neulet.2016.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 11/27/2022]
Abstract
Age is an important consideration for recovery and repair after spinal cord injury. Spinal cord injury is increasingly affecting the middle-aged and aging populations. Despite rapid progress in research to promote axonal regeneration and repair, our understanding of how age can modulate this repair is rather limited. In this review, we discuss the literature supporting the notion of an age-dependent decline in axonal growth after central nervous system (CNS) injury. While both neuron-intrinsic and extrinsic factors are involved in the control of axon growth after injury, here we focus on possible intrinsic mechanisms for this age-dependent decline.
Collapse
|
171
|
Bradburn S, McPhee JS, Bagley L, Sipila S, Stenroth L, Narici MV, Pääsuke M, Gapeyeva H, Osborne G, Sassano L, Meskers CGM, Maier AB, Hogrel JY, Barnouin Y, Butler-Browne G, Murgatroyd C. Association between osteocalcin and cognitive performance in healthy older adults. Age Ageing 2016; 45:844-849. [PMID: 27515675 PMCID: PMC5105824 DOI: 10.1093/ageing/afw137] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/23/2016] [Indexed: 01/25/2023] Open
Abstract
Introduction cognitive deterioration and reductions of bone health coincide with increasing age. We examine the relationship between bone composition and plasma markers of bone remodelling with measures of cognitive performance in healthy adults. Methods this cross-sectional study included 225 old (52% women, mean age: 74.4 ± 3.3 years) and 134 young (52% women, mean age: 23.4 ± 2.7 years) adult participants from the MyoAge project. Whole body bone mineral density was measured by dual-energy X-ray absorptiometry. Blood analyses included a panel of bone-related peptides (dickkopf-1, osteoprotegerin, osteocalcin (OC), osteopontin, sclerostin, parathyroid hormone and fibroblast growth factor 23), as well as serum calcium and 25-hydroxy vitamin D assays. A selection of cognitive domains (working memory capacity, episodic memory, executive functioning and global cognition) was assessed with a standardised neuropsychological test battery. Results adjusting for covariates and multiple testing revealed that plasma OC levels were positively associated with measures of executive functioning (β = 0.444, P < 0.001) and global cognition (β = 0.381, P = 0.001) in the older women. Discussion these correlative results demonstrate a positive association between OC, a factor known to regulate bone remodelling, with cognitive performance in older non-demented women. Further work should address possible mechanistic interpretations in humans.
Collapse
Affiliation(s)
- Steven Bradburn
- Manchester Metropolitan University, School of Healthcare Science, Manchester, UK
| | - Jamie S McPhee
- Manchester Metropolitan University, School of Healthcare Science, Manchester, UK
| | - Liam Bagley
- Manchester Metropolitan University, School of Healthcare Science, Manchester, UK
| | - Sarianna Sipila
- University of Jyväskylä, Gerontology Research Centre, Health Sciences, 40630 Jyväskylä, Finland
| | - Lauri Stenroth
- University of Jyväskylä, Gerontology Research Centre, Health Sciences, 40630 Jyväskylä, Finland
| | | | - Mati Pääsuke
- University of Tartu, Institute of Exercise Biology and Physiotherapy, Tartu, Estonia
| | - Helena Gapeyeva
- University of Tartu, Institute of Exercise Biology and Physiotherapy, Tartu, Estonia
| | | | | | - Carel G M Meskers
- VU University Medical Center, Rehabilitation Medicine, Amsterdam, The Netherlands
| | - Andrea B Maier
- VU University Medical Center, Faculty of Human Movement Sciences, Amsterdam, The Netherlands
- Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | | | | | | | - Chris Murgatroyd
- Manchester Metropolitan University, School of Healthcare Science, Manchester, UK
| |
Collapse
|
172
|
Caraci F, Tascedda F, Merlo S, Benatti C, Spampinato SF, Munafò A, Leggio GM, Nicoletti F, Brunello N, Drago F, Sortino MA, Copani A. Fluoxetine Prevents Aβ 1-42-Induced Toxicity via a Paracrine Signaling Mediated by Transforming-Growth-Factor-β1. Front Pharmacol 2016; 7:389. [PMID: 27826242 PMCID: PMC5078904 DOI: 10.3389/fphar.2016.00389] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023] Open
Abstract
Selective reuptake inhibitors (SSRIs), such as fluoxetine and sertraline, increase circulating Transforming-Growth-Factor-β1 (TGF-β1) levels in depressed patients, and are currently studied for their neuroprotective properties in Alzheimer’s disease. TGF-β1 is an anti-inflammatory cytokine that exerts neuroprotective effects against β-amyloid (Aβ)-induced neurodegeneration. In the present work, the SSRI, fluoxetine, was tested for the ability to protect cortical neurons against 1 μM oligomeric Aβ1-42-induced toxicity. At therapeutic concentrations (100 nM–1 μM), fluoxetine significantly prevented Aβ-induced toxicity in mixed glia-neuronal cultures, but not in pure neuronal cultures. Though to a lesser extent, also sertraline was neuroprotective in mixed cultures, whereas serotonin (10 nM–10 μM) did not mimick fluoxetine effects. Glia-conditioned medium collected from astrocytes challenged with fluoxetine protected pure cortical neurons against Aβ toxicity. The effect was lost in the presence of a neutralizing antibody against TGF-β1 in the conditioned medium, or when the specific inhibitor of type-1 TGF-β1 receptor, SB431542, was added to pure neuronal cultures. Accordingly, a 24 h treatment of cortical astrocytes with fluoxetine promoted the release of active TGF-β1 in the culture media through the conversion of latent TGF-β1 to mature TGF-β1. Unlike fluoxetine, both serotonin and sertraline did not stimulate the astrocyte release of active TGF-β1. We conclude that fluoxetine is neuroprotective against Aβ toxicity via a paracrine signaling mediated by TGF-β1, which does not result from a simplistic SERT blockade.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Drug Sciences, University of CataniaCatania, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Oasi Maria SantissimaTroina, Italy
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia Modena, Italy
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Cristina Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia Modena, Italy
| | - Simona F Spampinato
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Antonio Munafò
- Department of Drug Sciences, University of Catania Catania, Italy
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Ferdinando Nicoletti
- Istituto di Ricovero e Cura a Carattere Scientifico NeuromedPozzilli, Italy; Department of Physiology and Pharmacology, University of Rome SapienzaRome, Italy
| | - Nicoletta Brunello
- Istituto di Ricovero e Cura a Carattere Scientifico Oasi Maria Santissima Troina, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Agata Copani
- Department of Drug Sciences, University of CataniaCatania, Italy; Institute of Biostructure and Bioimaging, National Research CouncilCatania, Italy
| |
Collapse
|
173
|
Vallée A, Lecarpentier Y. Alzheimer Disease: Crosstalk between the Canonical Wnt/Beta-Catenin Pathway and PPARs Alpha and Gamma. Front Neurosci 2016; 10:459. [PMID: 27807401 PMCID: PMC5069291 DOI: 10.3389/fnins.2016.00459] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
The molecular mechanisms underlying the pathophysiology of Alzheimer's disease (AD) are still not fully understood. In AD, Wnt/beta-catenin signaling has been shown to be downregulated while the peroxisome proliferator-activated receptor (PPAR) gamma (mARN and protein) is upregulated. Certain neurodegenerative diseases share the same Wnt/beta-catenin/PPAR gamma profile, such as bipolar disorder and schizophrenia. Conversely, other NDs share an opposite profile, such as amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, multiple sclerosis, and Friedreich's ataxia. AD is characterized by the deposition of extracellular Abeta plaques and the formation of intracellular neurofibrillary tangles in the central nervous system (CNS). Activation of Wnt signaling or inhibition of both glycogen synthase kinase-3beta and Dickkopf 1, two key negative regulators of the canonical Wnt pathway, are able to protect against Abeta neurotoxicity and to ameliorate cognitive performance in AD patients. Although PPAR gamma is upregulated in AD patients, and despite the fact that it has been shown that the PPAR gamma and Wnt/beta catenin pathway systems work in an opposite manner, PPAR gamma agonists diminish learning and memory deficits, decrease Abeta activation of microglia, and prevent hippocampal and cortical neurons from dying. These beneficial effects observed in AD transgenic mice and patients might be partially due to the anti-inflammatory properties of PPAR gamma agonists. Moreover, activation of PPAR alpha upregulates transcription of the alpha-secretase gene and represents a new therapeutic treatment for AD. This review focuses largely on the behavior of two opposing pathways in AD, namely Wnt/beta-catenin signaling and PPAR gamma. It is hoped that this approach may help to develop novel AD therapeutic strategies integrating PPAR alpha signaling.
Collapse
Affiliation(s)
- Alexandre Vallée
- CHU Amiens Picardie, Université Picardie Jules VerneAmiens, France
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of PoitiersPoitiers, France
- AP-HP, Epidemiology and Clinical Research Department, University Hospital Bichat-Claude BernardParis, France
| | | |
Collapse
|
174
|
Marzo A, Galli S, Lopes D, McLeod F, Podpolny M, Segovia-Roldan M, Ciani L, Purro S, Cacucci F, Gibb A, Salinas PC. Reversal of Synapse Degeneration by Restoring Wnt Signaling in the Adult Hippocampus. Curr Biol 2016; 26:2551-2561. [PMID: 27593374 PMCID: PMC5070786 DOI: 10.1016/j.cub.2016.07.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/12/2016] [Indexed: 11/30/2022]
Abstract
Synapse degeneration occurs early in neurodegenerative diseases and correlates strongly with cognitive decline in Alzheimer’s disease (AD). The molecular mechanisms that trigger synapse vulnerability and those that promote synapse regeneration after substantial synaptic failure remain poorly understood. Increasing evidence suggests a link between a deficiency in Wnt signaling and AD. The secreted Wnt antagonist Dickkopf-1 (Dkk1), which is elevated in AD, contributes to amyloid-β-mediated synaptic failure. However, the impact of Dkk1 at the circuit level and the mechanism by which synapses disassemble have not yet been explored. Using a transgenic mouse model that inducibly expresses Dkk1 in the hippocampus, we demonstrate that Dkk1 triggers synapse loss, impairs long-term potentiation, enhances long-term depression, and induces learning and memory deficits. We decipher the mechanism involved in synapse loss induced by Dkk1 as it can be prevented by combined inhibition of the Gsk3 and RhoA-Rock pathways. Notably, after loss of synaptic connectivity, reactivation of the Wnt pathway by cessation of Dkk1 expression completely restores synapse number, synaptic plasticity, and long-term memory. These findings demonstrate the remarkable capacity of adult neurons to regenerate functional circuits and highlight Wnt signaling as a targetable pathway for neuronal circuit recovery after synapse degeneration. Wnt signaling is required for synapse integrity in the adult hippocampus Dkk1 induces synapse loss and deficits in synaptic plasticity and long-term memory Dkk1 disassembles synapses by activating the Gsk3 and Rock pathways Synapse loss and memory defects are reversible by reactivation of the Wnt pathway
Collapse
Affiliation(s)
- Aude Marzo
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Soledad Galli
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Douglas Lopes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Faye McLeod
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Marina Podpolny
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | | | - Lorenza Ciani
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Silvia Purro
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Francesca Cacucci
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Alasdair Gibb
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
175
|
Pang L, Dong N, Wang D, Zhang N, Xing J. Increased Dickkopf-1 expression is correlated with poisoning severity in carbon monoxide-poisoned humans and rats. Inhal Toxicol 2016; 28:455-62. [PMID: 27353797 DOI: 10.1080/08958378.2016.1198440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
CONTEXT Carbon monoxide (CO) poisoning results in neuronal injury. The expression of Dickkopf-1 (DKK-1) has not been investigated previously after CO poisoning. OBJECTIVE The current study aimed to investigate the DKK-1 expression levels in humans and rats with acute CO poisoning and to analyze their correlation with poisoning severity. MATERIALS AND METHODS We measured serum DKK-1 levels in patients with acute CO poisoning (n = 94) and in healthy controls (n = 90). On admission, a poisoning severity score (PSS) was determined for each patient. In addition, 36 male Sprague-Dawley rats were randomly assigned into three groups: (a) Sham group, (b) Low CO group and (c) High CO group. At 2 h after CO poisoning, DKK-1 expression and histopathological damage in the hippocampal tissues were measured. RESULTS Serum DKK-1 levels were significantly higher in the acute CO-poisoned patients, compared to the healthy controls. Serum DKK-1 levels were significantly higher in the CO-poisoned patients with a lower PSS. In rats, CO poisoning induced significant upregulation of the gene and protein expression of DKK-1 in hippocampal tissues. Moreover, there was a positive correlation between DKK-1 levels and the degree of damage in the hippocampal tissues. DISCUSSION DKK-1 induction in neurons after CO poisoning causes further neuronal injury. The severity of acute CO poisoning in rat models is associated with elevated serum DKK-1 levels and its upregulation in the brain tissue. CONCLUSION DKK-1 appears to have potential utility in providing valuable information for determining the severity and damage of CO poisoning.
Collapse
Affiliation(s)
- Li Pang
- a Department of Emergency , the First Hospital of Jilin University , Changchun , China
| | - Ning Dong
- a Department of Emergency , the First Hospital of Jilin University , Changchun , China
| | - Dawei Wang
- a Department of Emergency , the First Hospital of Jilin University , Changchun , China
| | - Nan Zhang
- a Department of Emergency , the First Hospital of Jilin University , Changchun , China
| | - Jihong Xing
- a Department of Emergency , the First Hospital of Jilin University , Changchun , China
| |
Collapse
|
176
|
Libro R, Bramanti P, Mazzon E. The role of the Wnt canonical signaling in neurodegenerative diseases. Life Sci 2016; 158:78-88. [PMID: 27370940 DOI: 10.1016/j.lfs.2016.06.024] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/20/2016] [Accepted: 06/26/2016] [Indexed: 01/06/2023]
Abstract
The Wnt/β-catenin or Wnt canonical pathway controls multiple biological processes throughout development and adult life. Growing evidences have suggested that deregulation of the Wnt canonical pathway could be involved in the pathogenesis of neurodegenerative diseases. The Wnt canonical signaling is a pathway tightly regulated, which activation results in the inhibition of the Glycogen Synthase Kinase 3β (GSK-3β) function and in increased β-catenin activity, that migrates into the nucleus, activating the transcription of the Wnt target genes. Conversely, when the Wnt canonical pathway is turned off, increased levels of GSK-3β promote β-catenin degradation. Hence, GSK-3β could be considered as a key regulator of the Wnt canonical pathway. Of note, GSK-3β has also been involved in the modulation of inflammation and apoptosis, determining the delicate balance between immune tolerance/inflammation and neuronal survival/neurodegeneration. In this review, we have summarized the current acknowledgements about the role of the Wnt canonical pathway in the pathogenesis of some neurodegenerative diseases including Alzheimer's disease, cerebral ischemia, Parkinson's disease, Huntington's disease, multiple sclerosis and amyotrophic lateral sclerosis, with particular regard to the main in vitro and in vivo studies in this field, by reviewing 85 research articles about.
Collapse
Affiliation(s)
- Rosaliana Libro
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
177
|
sFRP-mediated Wnt sequestration as a potential therapeutic target for Alzheimer’s disease. Int J Biochem Cell Biol 2016; 75:104-11. [DOI: 10.1016/j.biocel.2016.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 01/28/2023]
|
178
|
Abstract
Wnt signaling has emerged in recent years as a major player in both nervous system development and adult synaptic plasticity. Of particular relevance to researchers studying learning and memory, Wnt signaling is critical for normal functioning of the hippocampus, a brain region that is essential for many types of memory formation and whose dysfunction is implicated in numerous neurodegenerative and psychiatric conditions. Impaired hippocampal Wnt signaling is implicated in several of these conditions, however, little is known about how Wnt signaling mediates hippocampal memory formation. This review will provide a general overview of Wnt signaling and discuss evidence demonstrating a key role for Wnt signaling in hippocampal memory formation in both normal and disease states. The regulation of Wnt signaling by ovarian sex steroid hormones will also be highlighted, given that the neuroprotection afforded by Wnt-hormone interactions may have significant implications for cognitive function in aging, neurodegenerative disease, and ischemic injury.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, WI, USA
| |
Collapse
|
179
|
Thysiadis S, Mpousis S, Avramidis N, Katsamakas S, Balomenos A, Remelli R, Efthimiopoulos S, Sarli V. Discovery of novel phenoxazinone derivatives as DKK1/LRP6 interaction inhibitors: Synthesis, biological evaluation and structure–activity relationships. Bioorg Med Chem 2016; 24:1014-22. [DOI: 10.1016/j.bmc.2016.01.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/11/2016] [Accepted: 01/14/2016] [Indexed: 01/22/2023]
|
180
|
The antineoplastic drug flavopiridol reverses memory impairment induced by Amyloid-ß1-42 oligomers in mice. Pharmacol Res 2016; 106:10-20. [PMID: 26875816 DOI: 10.1016/j.phrs.2016.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/06/2023]
Abstract
The ectopic re-activation of cell cycle in neurons is an early event in the pathogenesis of Alzheimer's disease (AD), which could lead to synaptic failure and ensuing cognitive deficits before frank neuronal death. Cytostatic drugs that act as cyclin-dependent kinase (CDK) inhibitors have been poorly investigated in animal models of AD. In the present study, we examined the effects of flavopiridol, an inhibitor of CDKs currently used as antineoplastic drug, against cell cycle reactivation and memory loss induced by intracerebroventricular injection of Aß1-42 oligomers in CD1 mice. Cycling neurons, scored as NeuN-positive cells expressing cyclin A, were found both in the frontal cortex and in the hippocampus of Aβ-injected mice, paralleling memory deficits. Starting from three days after Aβ injection, flavopiridol (0.5, 1 and 3mg/kg) was intraperitoneally injected daily, for eleven days. Here we show that a treatment with flavopiridol (0.5 and 1mg/kg) was able to rescue the loss of memory induced by Aβ1-42, and to prevent the occurrence of ectopic cell-cycle events in the mouse frontal cortex and hippocampus. This is the first evidence that a cytostatic drug can prevent cognitive deficits in a non-transgenic animal model of AD.
Collapse
|
181
|
Stress-induced upregulation of VLDL receptor alters Wnt-signaling in neurons. Exp Cell Res 2016; 340:238-47. [PMID: 26751967 DOI: 10.1016/j.yexcr.2016.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/16/2015] [Accepted: 01/01/2016] [Indexed: 12/12/2022]
Abstract
Lipoprotein receptor family members hold multiple roles in the brain, and alterations in lipoprotein receptor expression and function are implicated in neuronal stress, developmental disorders and neurodegenerative diseases, such as Alzheimer's disease. Berberine (BBR), a nutraceutical shown to have both neuroprotective and neurotoxic properties, is suggested to regulate lipoprotein receptor expression. We show that subtoxic concentration of BBR regulates neuronal lipoprotein receptor expression in a receptor- and time-dependent fashion in cerebellar granule neurons (CGN). Similarly to BBR, subtoxic concentrations of neuronal stressors cobalt chloride, thapsigargin and rotenone increased very-low-density lipoprotein receptor (VLDLR) mRNA and protein expression in CGN suggesting a conserved pathway for stress-induced upregulation of VLDLR in neurons. We also show that VLDLR upregulation is accompanied by transiently increased stabilization of hypoxia-induced factor 1 alpha (HIF-1α) and decreased β-catenin levels affecting the Wnt pathway through GSK3β phosphorylation, a crucial player in neurodegenerative processes. Our results indicate that neuronal stress differentially regulates lipoprotein receptor expression in neurons, with VLDLR upregulation as a common element as a modulator of neuronal Wnt signaling.
Collapse
|
182
|
Mpousis S, Thysiadis S, Avramidis N, Katsamakas S, Efthimiopoulos S, Sarli V. Synthesis and evaluation of gallocyanine dyes as potential agents for the treatment of Alzheimer's disease and related neurodegenerative tauopathies. Eur J Med Chem 2016; 108:28-38. [DOI: 10.1016/j.ejmech.2015.11.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/27/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022]
|
183
|
Wnt/Ryk signaling contributes to neuropathic pain by regulating sensory neuron excitability and spinal synaptic plasticity in rats. Pain 2015; 156:2572-2584. [DOI: 10.1097/j.pain.0000000000000366] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
184
|
Sun J, Wang H, Chen LN, Wang J, Lv Y, Yang XD, Zhang BY, Tian C, Shi Q, Dong XP. Remarkable impairment of Wnt/β-catenin signaling in the brains of the mice infected with scrapie agents. J Neurochem 2015; 136:731-740. [PMID: 26526056 DOI: 10.1111/jnc.13416] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/02/2015] [Accepted: 10/02/2015] [Indexed: 01/06/2023]
Abstract
Prion diseases are a group of neurodegenerative diseases characterized by neuronal loss and spongiform degeneration, astrogliosis and aggregation of scrapie prion protein (PrPSc ) in the central nervous system (CNS). The Wnt signaling pathway is a highly evolutionarily conserved pathway in eukaryotes that regulates cell proliferation, differentiation and survival. Impairment of Wnt/β-catenin signaling has been reported in the CNS of various neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. To investigate the functional state of Wnt/β-catenin signaling in the CNS tissues during the progression of prion disease, the components of Wnt/β-catenin signaling in the brains of the scrapie agents 139A- and ME7-infected mice were evaluated. Compared with the normal controls, the brain levels of phosphor-β-catenin (Ser33,37 and Thr41 ) in 139A- and ME7-infected mice were significantly increased, while those of cyclin D1, which is one of the target genes of Wnt signaling, were decreased. The levels of phosphor-glycogen synthase kinase-3β (GSK-3β) Ser9 were markedly reduced, representing an enhanced GSK-3β activity in scrapie-infected mice. Both western blot and immunohistochemical assays revealed a remarkable increase of Dickkopf-1, the antagonist of Wnt/β-catenin signaling, in the brains of scrapie-infected anim-als, which co-localized well with the remaining neurons in the immunofluorescent tests. We also observed slightly decreased Wnt-3 and unchanged disheveled-3 (Dvl-3) in the brains of the infected mice. Our data, here, strongly indicate an impairment of Wnt/β-catenin pathway in the brains of prion disease, which shows a time-dependent progression along with the incubation period. Schematic for the impairment of canonical Wnt signaling during prion infection. The left and right parts represent the normal and prion-infected situations, respectively. Prion infection or PrPSc accumulation triggers the over-expression of Dickkopf WNT signaling pathway inhibitor 1 (DKK-1) and the enhancement of glycogen synthase kinase 3β (GSK-3β) activity, which subsequently promotes the phosphorylation and degradation of β-catenin. As a result, the impairment of β-catenin signaling leads to the down-regulation of Wnt target genes.
Collapse
Affiliation(s)
- Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Wang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.,Department of Immunology and the key Laboratory of laboratory medicine of Jiangsu province, Jiangsu University Medical School, Zhenjiang, Jiangsu, China
| | - Li-Na Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Wang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Dong Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bao-Yun Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chan Tian
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University, Hangzhou 310003), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.,Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
185
|
Tapia-Rojas C, Lindsay CB, Montecinos-Oliva C, Arrazola MS, Retamales RM, Bunout D, Hirsch S, Inestrosa NC. Is L-methionine a trigger factor for Alzheimer's-like neurodegeneration?: Changes in Aβ oligomers, tau phosphorylation, synaptic proteins, Wnt signaling and behavioral impairment in wild-type mice. Mol Neurodegener 2015; 10:62. [PMID: 26590557 PMCID: PMC4654847 DOI: 10.1186/s13024-015-0057-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 11/02/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND L-methionine, the principal sulfur-containing amino acid in proteins, plays critical roles in cell physiology as an antioxidant and in the breakdown of fats and heavy metals. Previous studies suggesting the use of L-methionine as a treatment for depression and other diseases indicate that it might also improve memory and propose a role in brain function. However, some evidence indicates that an excess of methionine can be harmful and can increase the risk of developing Type-2 diabetes, heart diseases, certain types of cancer, brain alterations such as schizophrenia, and memory impairment. RESULTS Here, we report the effects of an L-methionine-enriched diet in wild-type mice and emphasize changes in brain structure and function. The animals in our study presented 1) higher levels of phosphorylated tau protein, 2) increased levels of amyloid-β (Aβ)-peptides, including the formation of Aβ oligomers, 3) increased levels of inflammatory response,4) increased oxidative stress, 5) decreased level of synaptic proteins, and 6) memory impairment and loss. We also observed dysfunction of the Wnt signaling pathway. CONCLUSION Taken together, the results of our study indicate that an L-methionine-enriched diet causes neurotoxic effects in vivo and might contribute to the appearance of Alzheimer's-like neurodegeneration.
Collapse
Affiliation(s)
- Cheril Tapia-Rojas
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina B Lindsay
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carla Montecinos-Oliva
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena S Arrazola
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rocio M Retamales
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Bunout
- Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Sandra Hirsch
- Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, P. Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia. .,Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,CARE Biomedical Center, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile.
| |
Collapse
|
186
|
Maslinic acid promotes synaptogenesis and axon growth via Akt/GSK-3β activation in cerebral ischemia model. Eur J Pharmacol 2015; 764:298-305. [DOI: 10.1016/j.ejphar.2015.07.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 12/13/2022]
|
187
|
Salim H, Zong D, Hååg P, Novak M, Mörk B, Lewensohn R, Lundholm L, Viktorsson K. DKK1 is a potential novel mediator of cisplatin-refractoriness in non-small cell lung cancer cell lines. BMC Cancer 2015; 15:628. [PMID: 26353782 PMCID: PMC4565013 DOI: 10.1186/s12885-015-1635-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/01/2015] [Indexed: 12/30/2022] Open
Abstract
Background Platinum compounds are the mainstay of chemotherapy for lung cancer. Unfortunately treatment failure remains a critical issue since about 60 % of all non-small cell lung cancer (NSCLC) patients display intrinsic platinum resistance. Methods We analyzed global gene expression profiles of NSCLC clones surviving a pulse treatment with cisplatin and mapped deregulated signaling networks in silico by Ingenuity Pathway Analysis (IPA). Further validation was done using siRNA. Results The pooled cisplatin-surviving NSCLC clones from each of the biological replicates demonstrated heterogeneous gene expression patterns both in terms of the number and the identity of the altered genes. Genes involved in Wnt signaling pathway (Dickkopf-1, DKK1), DNA repair machinery (XRCC2) and cell-cell/cell-matrix interaction (FMN1, LGALS9) were among the top deregulated genes by microarray in these replicates and were validated by q-RT-PCR. We focused on DKK1 which previously was reported to be overexpressed in NSCLC patients. IPA network analysis revealed coordinate up-regulation of several DKK1 transcriptional regulators (TCF4, EZH2, DNAJB6 and HDAC2) in cisplatin-surviving clones from that biological replicate. Knockdown of DKK1 by siRNA sensitized for cisplatin in two different NSCLC cell lines and in ovarian A2780 cells, but not in the A2780 cis subline made resistant to cisplatin by chronic exposure, suggesting a role of DKK1 in intrinsic but not acquired platinum refractoriness. Conclusions We identified DKK1 as a possible marker of a cisplatin-refractory phenotype and as a potential novel therapeutic target to improve platinum response of NSCLC cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1635-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hogir Salim
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Dali Zong
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Petra Hååg
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Metka Novak
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Birgitta Mörk
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Rolf Lewensohn
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Lovisa Lundholm
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Kristina Viktorsson
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
188
|
Vossel KA, Xu JC, Fomenko V, Miyamoto T, Suberbielle E, Knox JA, Ho K, Kim DH, Yu GQ, Mucke L. Tau reduction prevents Aβ-induced axonal transport deficits by blocking activation of GSK3β. ACTA ACUST UNITED AC 2015; 209:419-33. [PMID: 25963821 PMCID: PMC4427789 DOI: 10.1083/jcb.201407065] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tau ablation, knockdown, and reconstitution studies in primary mouse neurons show that tau enables amyloid β oligomers to inhibit axonal transport through activation of GSK3β and through functions of tau that do not depend on its microtubule binding activity. Axonal transport deficits in Alzheimer’s disease (AD) are attributed to amyloid β (Aβ) peptides and pathological forms of the microtubule-associated protein tau. Genetic ablation of tau prevents neuronal overexcitation and axonal transport deficits caused by recombinant Aβ oligomers. Relevance of these findings to naturally secreted Aβ and mechanisms underlying tau’s enabling effect are unknown. Here we demonstrate deficits in anterograde axonal transport of mitochondria in primary neurons from transgenic mice expressing familial AD-linked forms of human amyloid precursor protein. We show that these deficits depend on Aβ1–42 production and are prevented by tau reduction. The copathogenic effect of tau did not depend on its microtubule binding, interactions with Fyn, or potential role in neuronal development. Inhibition of neuronal activity, N-methyl-d-aspartate receptor function, or glycogen synthase kinase 3β (GSK3β) activity or expression also abolished Aβ-induced transport deficits. Tau ablation prevented Aβ-induced GSK3β activation. Thus, tau allows Aβ oligomers to inhibit axonal transport through activation of GSK3β, possibly by facilitating aberrant neuronal activity.
Collapse
Affiliation(s)
- Keith A Vossel
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Jordan C Xu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Vira Fomenko
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Takashi Miyamoto
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Elsa Suberbielle
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| | - Joseph A Knox
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Daniel H Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 Department of Neurology, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
189
|
Bruggink KA, Kuiperij HB, Gloerich J, Otte-Höller I, Rozemuller AJM, Claassen JAHR, Küsters B, Verbeek MM. Dickkopf-related protein 3 is a potential Aβ-associated protein in Alzheimer's Disease. J Neurochem 2015; 134:1152-62. [PMID: 26119087 DOI: 10.1111/jnc.13216] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/27/2022]
Abstract
Amyloid-β (Aβ) is the most prominent protein in Alzheimer's disease (AD) senile plaques. In addition, Aβ interacts with a variety of Aβ-associated proteins (AAPs), some of which can form complexes with Aβ and influence its clearance, aggregation or toxicity. Identification of novel AAPs may shed new light on the pathophysiology of AD and the metabolic fate of Aβ. In this study, we aimed to identify new AAPs by searching for proteins that may form soluble complexes with Aβ in CSF, using a proteomics approach. We identified the secreted Wnt pathway protein Dickkopf-related protein 3 (Dkk-3) as a potential Aβ-associated protein. Using immunohistochemistry on human AD brain tissue, we observed that (i) Dkk-3 co-localizes with Aβ in the brain, both in diffuse and classic plaques. (ii) Dkk-3 is expressed in neurons and in blood vessel walls in the brain and (iii) is secreted by leptomeningeal smooth muscle cells in vitro. Finally, measurements using ELISA revealed that (iv) Dkk-3 protein is abundantly present in both cerebrospinal fluid and serum, but its levels are similar in non-demented controls and patients with AD, Lewy body dementia, and frontotemporal dementia. Our study demonstrates that Dkk-3 is a hitherto unidentified Aβ-associated protein which, given its relatively high cerebral concentrations and co-localization with Aβ, is potentially involved in AD pathology. In this study, we propose that Dickkopf-related protein-3 (Dkk-3) might be a novel Amyloid-β (Aβ) associated protein. We demonstrate that Dkk-3 is expressed in the brain, especially in vessel walls, and co-localizes with Aβ in senile plaques. Furthermore, Dkk-3 levels in cerebrospinal fluid strongly correlate with Aβ40 levels, but were not suitable to discriminate non-demented controls and patients with dementia.
Collapse
Affiliation(s)
- Kim A Bruggink
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Department of Laboratory Medicine, Radboud Proteomics Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Irene Otte-Höller
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology and Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pathology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| |
Collapse
|
190
|
Zhang L, Bahety P, Ee PLR. Wnt co-receptor LRP5/6 overexpression confers protection against hydrogen peroxide-induced neurotoxicity and reduces tau phosphorylation in SH-SY5Y cells. Neurochem Int 2015; 87:13-21. [PMID: 25959626 DOI: 10.1016/j.neuint.2015.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 04/14/2015] [Accepted: 05/04/2015] [Indexed: 02/03/2023]
Abstract
Emerging studies have suggested the involvement of dysregulated Wnt/β-catenin cascade in the etiology of Alzheimer's disease (AD). Recently, genetic variations in Wnt co-receptor low density lipoprotein receptor-related protein (LRP) 6 causing reduced Wnt signaling has been linked to late-onset AD. Here, we hypothesized that overexpression of Wnt co-receptors LRP5 and LRP6 would serve as an effective new approach in reducing neurotoxicity induced by oxidative stress and decreasing tau phosphorylation in SH-SY5Y human neuroblastoma cells. Our results showed that overexpression of LRP5 and LRP6 in SH-SY5Y cells activates Wnt signaling and downstream proliferation genes, whereas knockdown of the co-receptors represses Wnt signaling and the transcription of proliferative markers. We further demonstrated that overexpression of LRP5 and LRP6 protects SH-SY5Y from cell death caused by hydrogen peroxide-induced oxidative stress, inhibits GSK3β activity and subsequently reduces tau phosphorylation. Together, our findings suggest that rescuing LRP5/6-mediated Wnt signaling improves neuronal cell survival and reduces tau phosphorylation, which support the hypothesis that Wnt signaling might be an attractive therapeutic strategy for managing AD.
Collapse
Affiliation(s)
- Luqi Zhang
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Priti Bahety
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Pui Lai Rachel Ee
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
191
|
N-butylidenephthalide attenuates Alzheimer's disease-like cytopathy in Down syndrome induced pluripotent stem cell-derived neurons. Sci Rep 2015; 5:8744. [PMID: 25735452 PMCID: PMC4348654 DOI: 10.1038/srep08744] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/02/2015] [Indexed: 01/07/2023] Open
Abstract
Down syndrome (DS) patients with early-onset dementia share similar neurodegenerative features with Alzheimer's disease (AD). To recapitulate the AD cell model, DS induced pluripotent stem cells (DS-iPSCs), reprogrammed from mesenchymal stem cells in amniotic fluid, were directed toward a neuronal lineage. Neuroepithelial precursor cells with high purity and forebrain characteristics were robustly generated on day 10 (D10) of differentiation. Accumulated amyloid deposits, Tau protein hyperphosphorylation and Tau intracellular redistribution emerged rapidly in DS neurons within 45 days but not in normal embryonic stem cell-derived neurons. N-butylidenephthalide (Bdph), a major phthalide ingredient of Angelica sinensis, was emulsified by pluronic F127 to reduce its cellular toxicity and promote canonical Wnt signaling. Interestingly, we found that F127-Bdph showed significant therapeutic effects in reducing secreted Aβ40 deposits, the total Tau level and the hyperphosphorylated status of Tau in DS neurons. Taken together, DS-iPSC derived neural cells can serve as an ideal cellular model of DS and AD and have potential for high-throughput screening of candidate drugs. We also suggest that Bdph may benefit DS or AD treatment by scavenging Aβ aggregates and neurofibrillary tangles.
Collapse
|
192
|
Aberrant Wnt signaling pathway in medial temporal lobe structures of Alzheimer’s disease. J Neural Transm (Vienna) 2015; 122:1303-18. [PMID: 25680440 DOI: 10.1007/s00702-015-1375-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/03/2015] [Indexed: 01/13/2023]
|
193
|
Stolz A, Neufeld K, Ertych N, Bastians H. Wnt-mediated protein stabilization ensures proper mitotic microtubule assembly and chromosome segregation. EMBO Rep 2015; 16:490-9. [PMID: 25656539 DOI: 10.15252/embr.201439410] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/13/2015] [Indexed: 12/12/2022] Open
Abstract
Wnt signaling stimulates cell proliferation by promoting the G1/S transition of the cell cycle through β-catenin/TCF4-mediated gene transcription. However, Wnt signaling peaks in mitosis and contributes to the stabilization of proteins other than β-catenin, a pathway recently introduced as Wnt-dependent stabilization of proteins (Wnt/STOP). Here, we show that Wnt/STOP regulated by basal Wnt signaling during a normal cell cycle is required for proper spindle microtubule assembly and for faithful chromosome segregation during mitosis. Consequently, inhibition of basal Wnt signaling results in increased microtubule assembly rates, abnormal mitotic spindle formation and the induction of aneuploidy in human somatic cells.
Collapse
Affiliation(s)
- Ailine Stolz
- Section for Cellular Oncology, Institute of Molecular Oncology, Georg-August University Göttingen Göttingen Center for Molecular Biosciences (GZMB) and University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Kim Neufeld
- Section for Cellular Oncology, Institute of Molecular Oncology, Georg-August University Göttingen Göttingen Center for Molecular Biosciences (GZMB) and University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Norman Ertych
- Section for Cellular Oncology, Institute of Molecular Oncology, Georg-August University Göttingen Göttingen Center for Molecular Biosciences (GZMB) and University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Holger Bastians
- Section for Cellular Oncology, Institute of Molecular Oncology, Georg-August University Göttingen Göttingen Center for Molecular Biosciences (GZMB) and University Medical Center Göttingen (UMG), Göttingen, Germany
| |
Collapse
|
194
|
Vargas JY, Ahumada J, Arrázola MS, Fuenzalida M, Inestrosa NC. WASP-1, a canonical Wnt signaling potentiator, rescues hippocampal synaptic impairments induced by Aβ oligomers. Exp Neurol 2015; 264:14-25. [DOI: 10.1016/j.expneurol.2014.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 12/12/2022]
|
195
|
Patel AK, Surapaneni K, Yi H, Nakamura REI, Karli SZ, Syeda S, Lee T, Hackam AS. Activation of Wnt/β-catenin signaling in Muller glia protects photoreceptors in a mouse model of inherited retinal degeneration. Neuropharmacology 2014; 91:1-12. [PMID: 25486619 DOI: 10.1016/j.neuropharm.2014.11.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/13/2014] [Accepted: 11/19/2014] [Indexed: 11/18/2022]
Abstract
The canonical Wnt/β-catenin ("Wnt") pathway is an essential signaling cascade in the embryonic central nervous system (CNS) that regulates neuronal differentiation and survival. Loss of Wnt signaling in developing and adult tissue has been implicated in numerous CNS diseases, but the precise role of Wnt in regulating neuronal survival, and how its absence could lead to disease, is not understood. In this study, we investigated the effect of Wnt activation on neuronal survival in the adult retina, and identified cellular and molecular mediators. Pan-retinal Wnt signaling activation using Wnt3a induced functional and morphological rescue of photoreceptor neurons in the rd10 mouse model of retinal degeneration. Furthermore, Wnt activation using constitutively active β-catenin specifically targeted to Muller glia increased photoreceptor survival and reduced markers of glial and neuronal remodeling. Wnt-induced photoreceptor protection was associated with elevated levels of the prosurvival protein Stat3, and was reduced by shRNA-mediated knock-down of Stat3, indicating cross-talk between survival pathways. Therefore, these data increase our understanding of the role of Wnt signaling in the retina, and identify radial Muller glia as important cellular mediators of Wnt activity.
Collapse
Affiliation(s)
- Amit K Patel
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Krishna Surapaneni
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Hyun Yi
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Rei E I Nakamura
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Sapir Z Karli
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Sarah Syeda
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Tinthu Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, USA.
| |
Collapse
|
196
|
Tu S, Okamoto SI, Lipton SA, Xu H. Oligomeric Aβ-induced synaptic dysfunction in Alzheimer's disease. Mol Neurodegener 2014; 9:48. [PMID: 25394486 PMCID: PMC4237769 DOI: 10.1186/1750-1326-9-48] [Citation(s) in RCA: 412] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/08/2014] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating disease characterized by synaptic and neuronal loss in the elderly. Compelling evidence suggests that soluble amyloid-β peptide (Aβ) oligomers induce synaptic loss in AD. Aβ-induced synaptic dysfunction is dependent on overstimulation of N-methyl-D-aspartate receptors (NMDARs) resulting in aberrant activation of redox-mediated events as well as elevation of cytoplasmic Ca2+, which in turn triggers downstream pathways involving phospho-tau (p-tau), caspases, Cdk5/dynamin-related protein 1 (Drp1), calcineurin/PP2B, PP2A, Gsk-3β, Fyn, cofilin, and CaMKII and causes endocytosis of AMPA receptors (AMPARs) as well as NMDARs. Dysfunction in these pathways leads to mitochondrial dysfunction, bioenergetic compromise and consequent synaptic dysfunction and loss, impaired long-term potentiation (LTP), and cognitive decline. Evidence also suggests that Aβ may, at least in part, mediate these events by causing an aberrant rise in extrasynaptic glutamate levels by inhibiting glutamate uptake or triggering glutamate release from glial cells. Consequent extrasynaptic NMDAR (eNMDAR) overstimulation then results in synaptic dysfunction via the aforementioned pathways. Consistent with this model of Aβ-induced synaptic loss, Aβ synaptic toxicity can be partially ameliorated by the NMDAR antagonists (such as memantine and NitroMemantine). PSD-95, an important scaffolding protein that regulates synaptic distribution and activity of both NMDA and AMPA receptors, is also functionally disrupted by Aβ. PSD-95 dysregulation is likely an important intermediate step in the pathological cascade of events caused by Aβ. In summary, Aβ-induced synaptic dysfunction is a complicated process involving multiple pathways, components and biological events, and their underlying mechanisms, albeit as yet incompletely understood, may offer hope for new therapeutic avenues.
Collapse
Affiliation(s)
- Shichun Tu
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
197
|
Parr C, Mirzaei N, Christian M, Sastre M. Activation of the Wnt/β‐catenin pathway represses the transcription of the β‐amyloid precursor protein cleaving enzyme (BACE1) via binding of T‐cell factor‐4 to BACE1 promoter. FASEB J 2014; 29:623-35. [DOI: 10.1096/fj.14-253211] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Callum Parr
- Division of Brain SciencesImperial College LondonLondonUnited Kingdom
| | - Nazanin Mirzaei
- Division of Brain SciencesImperial College LondonLondonUnited Kingdom
| | - Mark Christian
- Division of Metabolic and Vascular HealthWarwick Medical SchoolUniversity of WarwickCoventryUnited Kingdom
| | - Magdalena Sastre
- Division of Brain SciencesImperial College LondonLondonUnited Kingdom
| |
Collapse
|
198
|
Galli S, Lopes DM, Ammari R, Kopra J, Millar SE, Gibb A, Salinas PC. Deficient Wnt signalling triggers striatal synaptic degeneration and impaired motor behaviour in adult mice. Nat Commun 2014; 5:4992. [PMID: 25318560 PMCID: PMC4218967 DOI: 10.1038/ncomms5992] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 08/15/2014] [Indexed: 12/31/2022] Open
Abstract
Synapse degeneration is an early and invariant feature of neurodegenerative diseases. Indeed, synapse loss occurs prior to neuronal degeneration and correlates with the symptom severity of these diseases. However, the molecular mechanisms that trigger synaptic loss remain poorly understood. Here we demonstrate that deficient Wnt signalling elicits synaptic degeneration in the adult striatum. Inducible expression of the secreted Wnt antagonist Dickkopf1 (Dkk1) in adult mice (iDkk1) decreases the number of cortico-striatal glutamatergic synapses and of D1 and D2 dopamine receptor clusters. Synapse loss occurs in the absence of axon retraction or cell death. The remaining excitatory terminals contain fewer synaptic vesicles and have a reduced probability of evoked transmitter release. IDkk1 mice show impaired motor coordination and are irresponsive to amphetamine. These studies identify Wnts as key endogenous regulators of synaptic maintenance and suggest that dysfunction in Wnt signalling contributes to synaptic degeneration at early stages in neurodegenerative diseases.
Collapse
Affiliation(s)
- Soledad Galli
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Douglas M. Lopes
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Rachida Ammari
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Sarah E. Millar
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Alasdair Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
199
|
Bayod S, Felice P, Andrés P, Rosa P, Camins A, Pallàs M, Canudas AM. Downregulation of canonical Wnt signaling in hippocampus of SAMP8 mice. Neurobiol Aging 2014; 36:720-9. [PMID: 25443287 DOI: 10.1016/j.neurobiolaging.2014.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 09/14/2014] [Accepted: 09/17/2014] [Indexed: 01/09/2023]
Abstract
In the adult brain, canonical Wnt (Wnt/β-catenin) signaling modulates neuronal function, hippocampal neurogenesis, and synaptic plasticity. Indeed, growing evidence suggests that downregulation of Wnt signaling could be involved in the cognitive decline associated with aging and also with the physiopathology of Alzheimer's disease (AD). However, the molecular basis remains unknown. At present, SAMP8 is an experimental model that has been proposed for studying age-related neurodegenerative changes associated with aging and the pathogenesis of AD. Here, we examined Wnt signaling in the hippocampus of SAMP8 mice at 9 and 12 months of age, as well as in its control-strain SAMR1 mice. Our results showed increased Dickkopf-1 protein levels in SAMP8 with age, in addition to GSK-3 α/β activation and hyperphosphorylated tau. Consequently, higher β-catenin phosphorylation at Ser(33,37) and Thr(41), which promotes its degradation, along with a decrease in active β-catenin (ABC) in the nucleus, were observed in SAMP8, mainly at the age of 12 months. Moreover, nuclear levels of Dvl3 were lower in 9- and 12-month-old SAMP8 mice. Related to these findings, SAMP8 showed an increase in neuronal loss in the hippocampus that was associated with lower protein levels of the antiapoptotic protein and the Wnt target gene, Bcl-2, in addition to an increase in the proapototic protein Bax. Our results suggest a relationship between age-related downregulation of canonical Wnt signaling and neuronal loss observed in the hippocampus of SAMP8 mice. Thus, enhancing Wnt signaling may represent a novel neuroprotective strategy aimed at counteracting the cognitive decline that is associated not only with aging but also with AD.
Collapse
Affiliation(s)
- Sergi Bayod
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Paolo Felice
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pol Andrés
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Paolo Rosa
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mercè Pallàs
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna-Maria Canudas
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Nucli Universitari de Pedralbes, Barcelona, Spain; Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
200
|
Hussaini SMQ, Choi CI, Cho CH, Kim HJ, Jun H, Jang MH. Wnt signaling in neuropsychiatric disorders: ties with adult hippocampal neurogenesis and behavior. Neurosci Biobehav Rev 2014; 47:369-83. [PMID: 25263701 DOI: 10.1016/j.neubiorev.2014.09.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 07/20/2014] [Accepted: 09/05/2014] [Indexed: 12/27/2022]
Abstract
In an effort to better understand and treat mental disorders, the Wnt pathway and adult hippocampal neurogenesis have received increased attention in recent years. One is a signaling pathway regulating key aspects of embryonic patterning, cell specification and adult tissue homeostasis. The other is the generation of newborn neurons in adulthood that integrate into the neural circuit and function in learning and memory, and mood behavior. In this review, we discuss the growing relationship between Wnt signaling-mediated regulation of adult hippocampal neurogenesis as it applies to neuropsychiatric disorders. Evidence suggests dysfunctional Wnt signaling may aberrantly regulate new neuron development and cognitive function. Indeed, altered expression of key Wnt pathway components are observed in the hippocampus of patients suffering from neuropsychiatric disorders. Clinically-utilized mood stabilizers also proceed through modulation of Wnt signaling in the hippocampus, while Wnt pathway antagonists can regulate the antidepressant response. Here, we review the role of Wnt signaling in disease etiology and pathogenesis, regulation of adult neurogenesis and behavior, and the therapeutic targeting of disease symptoms.
Collapse
Affiliation(s)
| | - Chan-Il Choi
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Chang Hoon Cho
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Hyo Jin Kim
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Heechul Jun
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Mi-Hyeon Jang
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|