1
|
Faraji N, Ebadpour N, Abavisani M, Gorji A. Unlocking Hope: Therapeutic Advances and Approaches in Modulating the Wnt Pathway for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:3630-3652. [PMID: 39313658 PMCID: PMC11790780 DOI: 10.1007/s12035-024-04462-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Neurodegenerative diseases (NDs) are conditions characterized by sensory, motor, and cognitive impairments due to alterations in the structure and function of neurons in the central nervous system (CNS). Despite their widespread occurrence, the exact causes of NDs remain largely elusive, and existing treatments fall short in efficacy. The Wnt signaling pathway is an emerging molecular pathway that has been linked to the development and progression of various NDs. Wnt signaling governs numerous cellular processes, such as survival, polarity, proliferation, differentiation, migration, and fate specification, via a complex network of proteins. In the adult CNS, Wnt signaling regulates synaptic transmission, plasticity, memory formation, neurogenesis, neuroprotection, and neuroinflammation, all essential for maintaining neuronal function and integrity. Dysregulation of both canonical and non-canonical Wnt signaling pathways contributes to neurodegeneration through various mechanisms, such as amyloid-β accumulation, tau protein hyperphosphorylation, dopaminergic neuron degeneration, and synaptic dysfunction, prompting investigations into Wnt modulation as a therapeutic target to restore neuronal function and prevent or delay neurodegenerative processes. Modulating Wnt signaling has the potential to restore neuronal function and impede or postpone neurodegenerative processes, offering a therapeutic approach for targeting NDs. In this article, the current knowledge about how Wnt signaling works in Alzheimer's disease and Parkinson's disease is discussed. Our study aims to explore the molecular mechanisms, recent discoveries, and challenges involved in developing Wnt-based therapies.
Collapse
Affiliation(s)
- Navid Faraji
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Epilepsy Research Center, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neurosurgery Department, Münster University, Münster, Germany.
| |
Collapse
|
2
|
Mei J, Li Y, Niu L, Liang R, Tang M, Cai Q, Xu J, Zhang D, Yin X, Liu X, Shen Y, Liu J, Xu M, Xia P, Ling J, Wu Y, Liang J, Zhang J, Yu P. SGLT2 inhibitors: a novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl Neurodegener 2024; 13:41. [PMID: 39123214 PMCID: PMC11312905 DOI: 10.1186/s40035-024-00431-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.
Collapse
Affiliation(s)
- Jiaqi Mei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Yi Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Ruikai Liang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyue Tang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingdong Xu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
3
|
Abela L, Gianfrancesco L, Tagliatti E, Rossignoli G, Barwick K, Zourray C, Reid KM, Budinger D, Ng J, Counsell J, Simpson A, Pearson TS, Edvardson S, Elpeleg O, Brodsky FM, Lignani G, Barral S, Kurian MA. Neurodevelopmental and synaptic defects in DNAJC6 parkinsonism, amenable to gene therapy. Brain 2024; 147:2023-2037. [PMID: 38242634 PMCID: PMC11146427 DOI: 10.1093/brain/awae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/10/2023] [Accepted: 12/16/2023] [Indexed: 01/21/2024] Open
Abstract
DNAJC6 encodes auxilin, a co-chaperone protein involved in clathrin-mediated endocytosis (CME) at the presynaptic terminal. Biallelic mutations in DNAJC6 cause a complex, early-onset neurodegenerative disorder characterized by rapidly progressive parkinsonism-dystonia in childhood. The disease is commonly associated with additional neurodevelopmental, neurological and neuropsychiatric features. Currently, there are no disease-modifying treatments for this condition, resulting in significant morbidity and risk of premature mortality. To investigate the underlying disease mechanisms in childhood-onset DNAJC6 parkinsonism, we generated induced pluripotent stem cells (iPSC) from three patients harbouring pathogenic loss-of-function DNAJC6 mutations and subsequently developed a midbrain dopaminergic neuronal model of disease. When compared to age-matched and CRISPR-corrected isogenic controls, the neuronal cell model revealed disease-specific auxilin deficiency as well as disturbance of synaptic vesicle recycling and homeostasis. We also observed neurodevelopmental dysregulation affecting ventral midbrain patterning and neuronal maturation. To explore the feasibility of a viral vector-mediated gene therapy approach, iPSC-derived neuronal cultures were treated with lentiviral DNAJC6 gene transfer, which restored auxilin expression and rescued CME. Our patient-derived neuronal model provides deeper insights into the molecular mechanisms of auxilin deficiency as well as a robust platform for the development of targeted precision therapy approaches.
Collapse
Affiliation(s)
- Lucia Abela
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Lorita Gianfrancesco
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Erica Tagliatti
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, 20089 Milano, Italy
| | - Giada Rossignoli
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Katy Barwick
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Clara Zourray
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kimberley M Reid
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Joanne Ng
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Genetic Therapy Accelerator Centre, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - John Counsell
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Arlo Simpson
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Toni S Pearson
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032-3784, USA
- Department of Pediatrics, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
- Department of Neurology, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
| | - Simon Edvardson
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Orly Elpeleg
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Frances M Brodsky
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Gabriele Lignani
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Neurology, Great Ormond Street Hospital, London, WC1N 3JH, UK
| |
Collapse
|
4
|
Anand AA, Khan M, V M, Kar D. The Molecular Basis of Wnt/ β-Catenin Signaling Pathways in Neurodegenerative Diseases. Int J Cell Biol 2023; 2023:9296092. [PMID: 37780577 PMCID: PMC10539095 DOI: 10.1155/2023/9296092] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 10/03/2023] Open
Abstract
Defective Wnt signaling is found to be associated with various neurodegenerative diseases. In the canonical pathway, the Frizzled receptor (Fzd) and the lipoprotein receptor-related proteins 5/6 (LRP5/LRP6) create a seven-pass transmembrane receptor complex to which the Wnt ligands bind. This interaction causes the tumor suppressor adenomatous polyposis coli gene product (APC), casein kinase 1 (CK1), and GSK-3β (glycogen synthase kinase-3 beta) to be recruited by the scaffold protein Dishevelled (Dvl), which in turn deactivates the β-catenin destruction complex. This inactivation stops the destruction complex from phosphorylating β-catenin. As a result, β-catenin first builds up in the cytoplasm and then migrates into the nucleus, where it binds to the Lef/Tcf transcription factor to activate the transcription of more than 50 Wnt target genes, including those involved in cell growth, survival, differentiation, neurogenesis, and inflammation. The treatments that are currently available for neurodegenerative illnesses are most commonly not curative in nature but are only symptomatic. According to all available research, restoring Wnt/β-catenin signaling in the brains of patients with neurodegenerative disorders, particularly Alzheimer's and Parkinson's disease, would improve the condition of several patients with neurological disorders. The importance of Wnt activators and modulators in patients with such illnesses is to mainly restore rather than overstimulate the Wnt/β-catenin signaling, thereby reestablishing the equilibrium between Wnt-OFF and Wnt-ON states. In this review, we have tried to summarize the significance of the Wnt canonical pathway in the pathophysiology of certain neurodegenerative diseases, such as Alzheimer's disease, cerebral ischemia, Parkinson's disease, Huntington's disease, multiple sclerosis, and other similar diseases, and as to how can it be restored in these patients.
Collapse
Affiliation(s)
- Ananya Anurag Anand
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad 211012, India
| | - Misbah Khan
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| | - Monica V
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| | - Debasish Kar
- Department of Biotechnology, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| |
Collapse
|
5
|
Jeensuk S, Ortega MS, Saleem M, Hawryluk B, Scheffler TL, Hansen PJ. Actions of WNT family member 5A to regulate characteristics of development of the bovine preimplantation embryo†. Biol Reprod 2022; 107:928-944. [PMID: 35765196 PMCID: PMC9562107 DOI: 10.1093/biolre/ioac127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
WNT signaling is important for regulation of embryonic development. The most abundant WNT gene expressed in the bovine endometrium during the preimplantation period is WNT5A. One objective was to determine whether WNT5A regulates competence of the bovine preimplantation embryo to become a blastocyst and alters the number of cells in the inner cell mass and trophectoderm. A second objective was to delineate features of the cell-signaling mechanisms involved in WNT5A actions. WNT5A caused a concentration-dependent increase in the proportion of embryos developing to the blastocyst stage and in the number of inner cell mass cells in the resultant blastocysts. A concentration of 200 ng/mL was most effective, and a higher concentration of 400 ng/mL was not stimulatory. Bovine serum albumin in culture reduced the magnitude of effects of WNT5A on development to the blastocyst stage. WNT5A affected expression of 173 genes at the morula stage; all were upregulated by WNT5A. Many of the upregulated genes were associated with cell signaling. Actions of WNT5A on development to the blastocyst stage were suppressed by a Rho-associated coiled-coil kinase (ROCK) signaling inhibitor, suggesting that WNT5A acts through Ras homology gene family member A (RhoA)/ROCK signaling. Other experiments indicated that actions of WNT5A are independent of the canonical β-catenin signaling pathway and RAC1/c-Jun N-terminal kinase (JNK) signaling. This is the first report outlining the actions of WNT5A to alter the development of the mammalian embryo. These findings provide insights into how embryokines regulate maternal-embryonic communication.
Collapse
Affiliation(s)
- Surawich Jeensuk
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
- Department of Livestock Development, Bureau of Biotechnology in Livestock Production, Pathum Thani, Thailand
| | - M Sofia Ortega
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Muhammad Saleem
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
- Department of Theriogenology, Faculty of Veterinary Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Briana Hawryluk
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Tracy L Scheffler
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Peter J Hansen
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
6
|
Ji XF, Fan YC, Sun F, Wang JW, Wang K. Noncanonical Wnt5a/JNK Signaling Contributes to the Development of D-Gal/LPS-Induced Acute Liver Failure. Inflammation 2022; 45:1362-1373. [PMID: 35098406 DOI: 10.1007/s10753-022-01627-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022]
Abstract
Acute liver failure (ALF) is a deadly clinical disorder with few effective treatments and unclear pathogenesis. In our previous study, we demonstrated that aberrant Wnt5a expression was involved in acute-on-chronic liver failure. However, the role of Wnt5a in ALF is unknown. We investigated the expression of Wnt5a and its downstream c-Jun N-terminal kinase (JNK) signaling in a mouse model of ALF established by coinjection of D-galactosamine (D-Gal) and lipopolysaccharide (LPS) in C57BL/6 mice. We also investigated the role of Box5, a Wnt5a antagonist, in vivo. Moreover, the effect of Wnt5a/JNK signaling on downstream inflammatory cytokine expression, phagocytosis, and migration in THP-1 macrophages was studied in vitro. Aberrant Wnt5a expression and JNK activation were detected in D-Gal/LPS-induced ALF mice. Box5 pretreatment reversed JNK activation and eventually decreased the mortality rate of D-Gal/LPS-treated mice, with reduced hepatic necrosis and apoptosis, serum ALT and AST levels, and liver inflammatory cytokine expression, although the latter was not significant. We further demonstrated that recombinant Wnt5a (rWnt5a)-induced tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) mRNA expression and increased THP-1 macrophage phagocytosis in a JNK-dependent manner, which could be restored by Box5. In addition, rWnt5a-induced migration of THP-1 macrophages was also reversed by Box5. Our findings suggested that Wnt5a/JNK signaling plays an important role in the development of ALF and that Box5 could have particular hepatoprotective effects in ALF.
Collapse
Affiliation(s)
- Xiang-Fen Ji
- Department of Hepatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Institute of Hepatology, Shandong University, Jinan, 250012, China
| | - Fei Sun
- Department of Hepatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Jing-Wei Wang
- Department of Hepatology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Institute of Hepatology, Shandong University, Jinan, 250012, China.
- Hepatology Institute of Shandong University, Wenhuaxi Road 107#, 250012, Jinan, Shandong, China.
| |
Collapse
|
7
|
Abstract
The sympathetic nervous system prepares the body for 'fight or flight' responses and maintains homeostasis during daily activities such as exercise, eating a meal or regulation of body temperature. Sympathetic regulation of bodily functions requires the establishment and refinement of anatomically and functionally precise connections between postganglionic sympathetic neurons and peripheral organs distributed widely throughout the body. Mechanistic studies of key events in the formation of postganglionic sympathetic neurons during embryonic and early postnatal life, including axon growth, target innervation, neuron survival, and dendrite growth and synapse formation, have advanced the understanding of how neuronal development is shaped by interactions with peripheral tissues and organs. Recent progress has also been made in identifying how the cellular and molecular diversity of sympathetic neurons is established to meet the functional demands of peripheral organs. In this Review, we summarize current knowledge of signalling pathways underlying the development of the sympathetic nervous system. These findings have implications for unravelling the contribution of sympathetic dysfunction stemming, in part, from developmental perturbations to the pathophysiology of peripheral neuropathies and cardiovascular and metabolic disorders.
Collapse
|
8
|
Godin SK, Wagner J, Huang P, Bree D. The role of peripheral nerve signaling in endometriosis. FASEB Bioadv 2021; 3:802-813. [PMID: 34632315 PMCID: PMC8493968 DOI: 10.1096/fba.2021-00063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
A hallmark of endometriosis - a chronic debilitating condition whose causes are poorly understood - is neuronal innervation of lesions. Recent evidence demonstrates that the peripheral nervous system plays an important role in the pathophysiology of this disease. Sensory nerves, which surround and innervate endometriotic lesions, not only drive the chronic and debilitating pain associated with endometriosis but also contribute to a pro-growth phenotype by secreting neurotrophic factors and interacting with surrounding immune cells. The diverse array of contributions that neurons play in endometriosis indicate that it should be considered as a nerve-centric disease. This review is focused on the emerging field of exoneural biology and how it applies to the field of endometriosis, in particular the role that peripheral nerves play in driving and maintaining endometriotic lesions. A better understanding of the mechanisms of neuronal contribution to endometriosis, as well as their interactions with accompanying stromal and immune cells, will unearth novel disease-relevant pathways and targets, providing additional, more selective therapeutic horizons.
Collapse
|
9
|
Freeman AK, Glendining KA, Jasoni CL. Developmental genes controlling neural circuit formation are expressed in the early postnatal hypothalamus and cellular lining of the third ventricle. J Neuroendocrinol 2021; 33:e13020. [PMID: 34423876 DOI: 10.1111/jne.13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
The arcuate nucleus of the hypothalamus is central in the regulation of body weight homeostasis through its ability to sense peripheral metabolic signals and relay them, through neural circuits, to other brain areas, ultimately affecting physiological and behavioural changes. The early postnatal development of these neural circuits is critical for normal body weight homeostasis, such that perturbations during this critical period can lead to obesity. The role for peripheral regulators of body weight homeostasis, including leptin, insulin and ghrelin, in this postnatal development is well described, yet some of the fundamental processes underpinning axonal and dendritic growth remain unclear. Here, we hypothesised that molecules known to regulate axonal and dendritic growth processes in other areas of the developing brain would be expressed in the postnatal arcuate nucleus and/or target nuclei where they would function to mediate the development of this circuitry. Using state-of-the-art RNAscope® technology, we have revealed the expression patterns of genes encoding Dcc/Netrin-1, Robo1/Slit1 and Fzd5/Wnt5a receptor/ligand pairs in the early postnatal mouse hypothalamus. We found that individual genes had unique expression patterns across developmental time in the arcuate nucleus, paraventricular nucleus of the hypothalamus, ventromedial nucleus of the hypothalamus, dorsomedial nucleus of the hypothalamus, median eminence and, somewhat unexpectedly, the third ventricle epithelium. These observations indicate a number of new molecular players in the development of neural circuits regulating body weight homeostasis, as well as novel molecular markers of tanycyte heterogeneity.
Collapse
Affiliation(s)
- Alice Katherine Freeman
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
10
|
van Vliet AC, Lee J, van der Poel M, Mason MRJ, Noordermeer JN, Fradkin LG, Tannemaat MR, Malessy MJA, Verhaagen J, De Winter F. Coordinated changes in the expression of Wnt pathway genes following human and rat peripheral nerve injury. PLoS One 2021; 16:e0249748. [PMID: 33848304 PMCID: PMC8043392 DOI: 10.1371/journal.pone.0249748] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
A human neuroma-in continuity (NIC), formed following a peripheral nerve lesion, impedes functional recovery. The molecular mechanisms that underlie the formation of a NIC are poorly understood. Here we show that the expression of multiple genes of the Wnt family, including Wnt5a, is changed in NIC tissue from patients that underwent reconstructive surgery. The role of Wnt ligands in NIC pathology and nerve regeneration is of interest because Wnt ligands are implicated in tissue regeneration, fibrosis, axon repulsion and guidance. The observations in NIC prompted us to investigate the expression of Wnt ligands in the injured rat sciatic nerve and in the dorsal root ganglia (DRG). In the injured nerve, four gene clusters were identified with temporal expression profiles corresponding to particular phases of the regeneration process. In the DRG up- and down regulation of certain Wnt receptors suggests that nerve injury has an impact on the responsiveness of injured sensory neurons to Wnt ligands in the nerve. Immunohistochemistry showed that Schwann cells in the NIC and in the injured nerve are the source of Wnt5a, whereas the Wnt5a receptor Ryk is expressed by axons traversing the NIC. Taken together, these observations suggest a central role for Wnt signalling in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Arie C. van Vliet
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jinhui Lee
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Marlijn van der Poel
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Matthew R. J. Mason
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | | | - Lee G. Fradkin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Martijn R. Tannemaat
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J. A. Malessy
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Fred De Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Abstract
Investigations of the cellular and molecular mechanisms that mediate the development of the autonomic nervous system have identified critical genes and signaling pathways that, when disrupted, cause disorders of the autonomic nervous system. This review summarizes our current understanding of how the autonomic nervous system emerges from the organized spatial and temporal patterning of precursor cell migration, proliferation, communication, and differentiation, and discusses potential clinical implications for developmental disorders of the autonomic nervous system, including familial dysautonomia, Hirschsprung disease, Rett syndrome, and congenital central hypoventilation syndrome.
Collapse
Affiliation(s)
- Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana
| |
Collapse
|
12
|
Turovskaya MV, Epifanova EA, Tarabykin VS, Babaev AA, Turovsky EA. Interleukin-10 restores glutamate receptor-mediated Ca 2+-signaling in brain circuits under loss of Sip1 transcription factor. Int J Neurosci 2020; 132:114-125. [PMID: 32727246 DOI: 10.1080/00207454.2020.1803305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE This study aimed to investigate the connection between the mutation of the Sip1 transcription factor and impaired Ca2+-signaling, which reflects changes in neurotransmission in the cerebral cortex in vitro. METHODS We used mixed neuroglial cortical cell cultures derived from Sip1 mutant mice. The cells were loaded with a fluorescent ratiometric calcium-sensitive probe Fura-2 AM and epileptiform activity was modeled by excluding magnesium ions from the external media or adding a GABA(A) receptor antagonist, bicuculline. Intracellular calcium dynamics were recorded using fluorescence microscopy. To identify the level of gene expression, the Real-Time PCR method was used. RESULTS It was found that cortical neurons isolated from homozygous (Sip1fl/fl) mice with the Sip1 mutation demonstrate suppressed Ca2+ signals in models of epileptiform activity in vitro. Wild-type cortical neurons are characterized by synchronous high-frequency and high-amplitude Ca2+ oscillations occurring in all neurons of the network in response to Mg2+-free medium and bicuculline. But cortical Sip1fl/fl neurons only single Ca2+ pulses or attenuated Ca2+ oscillations are recorded and only in single neurons, while most of the cell network does not respond to these stimuli. This signal deficiency of Sip1fl/fl neurons correlates with a suppressed expression level of the genes encoding the subunits of NMDA, AMPA, and KA receptors; protein kinases PKA, JNK, CaMKII; and also the transcription factor Hif1α. These negative effects were partially abolished when Sip1fl/fl neurons are grown in media with anti-inflammatory cytokine IL-10. IL-10 increases the expression of the above-mentioned genes but not to the level of expression in wild-type. At the same time, the amplitudes of Ca2+ signals increase in response to the selective agonists of NMDA, AMPA and KA receptors, and the proportion of neurons responding with Ca2+ oscillations to a Mg2+-free medium and bicuculline increases. CONCLUSION IL-10 restores neurotransmission in neuronal networks with the Sip1 mutation by regulating the expression of genes encoding signaling proteins.
Collapse
Affiliation(s)
- Maria V Turovskaya
- Laboratory of Intracellular Signaling, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences," Russia
| | - Ekaterina A Epifanova
- Laboratory of Genetic Engineering Technologies, Lobachevsky State University of Nizhni Novgorod, Russia
| | - Victor S Tarabykin
- Laboratory of Genetic Engineering Technologies, Lobachevsky State University of Nizhni Novgorod, Russia
| | - Alexei A Babaev
- Laboratory of Genetic Engineering Technologies, Lobachevsky State University of Nizhni Novgorod, Russia
| | - Egor A Turovsky
- Laboratory of Intracellular Signaling, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences," Russia.,Laboratory of Genetic Engineering Technologies, Lobachevsky State University of Nizhni Novgorod, Russia
| |
Collapse
|
13
|
Ferrero Restelli F, Fontanet PA, De Vincenti AP, Falzone TL, Ledda F, Paratcha G. Tetraspanin1 promotes NGF signaling by controlling TrkA receptor proteostasis. Cell Mol Life Sci 2020; 77:2217-2233. [PMID: 31440771 PMCID: PMC11104797 DOI: 10.1007/s00018-019-03282-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/06/2019] [Accepted: 08/15/2019] [Indexed: 11/27/2022]
Abstract
The molecular mechanisms that control the biosynthetic trafficking, surface delivery, and degradation of TrkA receptor are essential for proper nerve growth factor (NGF) function, and remain poorly understood. Here, we identify Tetraspanin1 (Tspan1) as a critical regulator of TrkA signaling and neuronal differentiation induced by NGF. Tspan1 is expressed by developing TrkA-positive dorsal root ganglion (DRG) neurons and its downregulation in sensory neurons inhibits NGF-mediated axonal growth. In addition, our data demonstrate that Tspan1 forms a molecular complex with the immature form of TrkA localized in the endoplasmic reticulum (ER). Finally, knockdown of Tspan1 reduces the surface levels of TrkA by promoting its preferential sorting towards the autophagy/lysosomal degradation pathway. Together, these data establish a novel homeostatic role of Tspan1, coordinating the biosynthetic trafficking and degradation of TrkA, regardless the presence of NGF.
Collapse
Affiliation(s)
- Facundo Ferrero Restelli
- División de Neurobiología Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina
| | - Paula Aldana Fontanet
- División de Neurobiología Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina
| | - Ana Paula De Vincenti
- División de Neurobiología Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina
| | - Tomás Luis Falzone
- Laboratorio de Transporte Axonal y Enfermedades Neurodegenerativas, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina
| | - Fernanda Ledda
- División de Neurobiología Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), CONICET, Buenos Aires, Argentina
| | - Gustavo Paratcha
- División de Neurobiología Molecular y Celular, Instituto de Biología Celular y Neurociencias (IBCN)-CONICET-UBA, Facultad de Medicina, University of Buenos Aires (UBA), CP1121, Buenos Aires, Argentina.
| |
Collapse
|
14
|
Sasaki K, Arimoto K, Kankawa K, Terada C, Yamamori T, Watakabe A, Yamamoto N. Rho Guanine Nucleotide Exchange Factors Regulate Horizontal Axon Branching of Cortical Upper Layer Neurons. Cereb Cortex 2020; 30:2506-2518. [PMID: 31768529 DOI: 10.1093/cercor/bhz256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/23/2019] [Indexed: 11/14/2022] Open
Abstract
Axon branching is a crucial process for cortical circuit formation. However, how the cytoskeletal changes in axon branching are regulated is not fully understood. In the present study, we investigated the role of RhoA guanine nucleotide exchange factors (RhoA-GEFs) in branch formation of horizontally elongating axons (horizontal axons) in the mammalian cortex. In situ hybridization showed that more than half of all known RhoA-GEFs were expressed in the developing rat cortex. These RhoA-GEFs were mostly expressed in the macaque cortex as well. An overexpression study using organotypic cortical slice cultures demonstrated that several RhoA-GEFs strongly promoted horizontal axon branching. Moreover, branching patterns were different between overexpressed RhoA-GEFs. In particular, ARHGEF18 markedly increased terminal arbors, whereas active breakpoint cluster region-related protein (ABR) increased short branches in both distal and proximal regions of horizontal axons. Rho kinase inhibitor treatment completely suppressed the branch-promoting effect of ARHGEF18 overexpression, but only partially affected that of ABR, suggesting that these RhoA-GEFs employ distinct downstream pathways. Furthermore, knockdown of either ARHGEF18 or ABR considerably suppressed axon branching. Taken together, the present study revealed that subsets of RhoA-GEFs differentially promote axon branching of mammalian cortical neurons.
Collapse
Affiliation(s)
- Kensuke Sasaki
- Cellular and Molecular Neurobiology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kei Arimoto
- Cellular and Molecular Neurobiology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kento Kankawa
- Cellular and Molecular Neurobiology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chikayo Terada
- Cellular and Molecular Neurobiology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuo Yamamori
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Akiya Watakabe
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Nobuhiko Yamamoto
- Cellular and Molecular Neurobiology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
15
|
Becker J, Wilting J. WNT Signaling in Neuroblastoma. Cancers (Basel) 2019; 11:cancers11071013. [PMID: 31331081 PMCID: PMC6679057 DOI: 10.3390/cancers11071013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 01/09/2023] Open
Abstract
The term WNT (wingless-type MMTV integration site family) signaling comprises a complex molecular pathway consisting of ligands, receptors, coreceptors, signal transducers and transcriptional modulators with crucial functions during embryonic development, including all aspects of proliferation, morphogenesis and differentiation. Its involvement in cancer biology is well documented. Even though WNT signaling has been divided into mainly three distinct branches in the past, increasing evidence shows that some molecular hubs can act in various branches by exchanging interaction partners. Here we discuss developmental and clinical aspects of WNT signaling in neuroblastoma (NB), an embryonic tumor with an extremely broad clinical spectrum, ranging from spontaneous differentiation to fatal outcome. We discuss implications of WNT molecules in NB onset, progression, and relapse due to chemoresistance. In the light of the still too high number of NB deaths, new pathways must be considered.
Collapse
Affiliation(s)
- Juergen Becker
- Department of Anatomy and Cell Biology, University Medical School Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany.
| | - Joerg Wilting
- Department of Anatomy and Cell Biology, University Medical School Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| |
Collapse
|
16
|
Szemes M, Greenhough A, Malik K. Wnt Signaling Is a Major Determinant of Neuroblastoma Cell Lineages. Front Mol Neurosci 2019; 12:90. [PMID: 31040767 PMCID: PMC6476918 DOI: 10.3389/fnmol.2019.00090] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 01/09/2023] Open
Abstract
The neural crest (NC), which has been referred to as the fourth germ layer, comprises a multipotent cell population which will specify diverse cells and tissues, including craniofacial cartilage and bones, melanocytes, the adrenal medulla and the peripheral nervous system. These cell fates are known to be determined by gene regulatory networks (GRNs) acting at various stages of NC development, such as induction, specification, and migration. Although transcription factor hierarchies and some of their interplay with morphogenetic signaling pathways have been characterized, the full complexity of activities required for regulated development remains uncharted. Deregulation of these pathways may contribute to tumorigenesis, as in the case of neuroblastoma, a frequently lethal embryonic cancer thought to arise from the sympathoadrenal lineage of the NC. In this “Hypothesis and Theory” article, we utilize the next generation sequencing data from neuroblastoma cells and tumors to evaluate the possible influences of Wnt signaling on NC GRNs and on neuroblastoma cell lineages. We propose that Wnt signaling is a major determinant of regulatory networks that underlie mesenchymal/neural crest cell (NCC)-like cell identities through PRRX1 and YAP/TAZ transcription factors. Furthermore, Wnt may also co-operate with Hedgehog signaling in driving proneural differentiation programmes along the adrenergic (ADRN) lineage. Elucidation of Signaling Regulatory Networks can augment and complement GRNs in characterizing cell identities, which may in turn contribute to the design of improved therapeutics tailored to primary and relapsing neuroblastoma.
Collapse
Affiliation(s)
- Marianna Szemes
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Alexander Greenhough
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
17
|
Yu F, Weng J, Yuan YS, Kou YH, Han N, Jiang BG, Zhang PX. Wnt5a Affects Schwann Cell Proliferation and Regeneration via Wnt/c-Jun and PTEN Signaling Pathway. Chin Med J (Engl) 2018; 131:2623-2625. [PMID: 30381602 PMCID: PMC6213825 DOI: 10.4103/0366-6999.244116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Fei Yu
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Jian Weng
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Yu-Song Yuan
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Yu-Hui Kou
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Na Han
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Bao-Guo Jiang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| | - Pei-Xun Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
18
|
He CW, Liao CP, Pan CL. Wnt signalling in the development of axon, dendrites and synapses. Open Biol 2018; 8:rsob.180116. [PMID: 30282660 PMCID: PMC6223216 DOI: 10.1098/rsob.180116] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022] Open
Abstract
Wnts are a highly conserved family of secreted glycoproteins that play essential roles in the morphogenesis and body patterning during the development of metazoan species. In recent years, mounting evidence has revealed important functions of Wnt signalling in diverse aspects of neural development, including neuronal polarization, guidance and branching of the axon and dendrites, as well as synapse formation and its structural remodelling. In contrast to Wnt signalling in cell proliferation and differentiation, which mostly acts through β-catenin-dependent pathways, Wnts engage a diverse array of non-transcriptional cascades in neuronal development, such as the planar cell polarity, cytoskeletal or calcium signalling pathways. In this review, we summarize recent advances in the mechanisms of Wnt signalling in the development of axon, dendrite and synapse formation.
Collapse
Affiliation(s)
- Chun-Wei He
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan, Republic of China
| | - Chien-Po Liao
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan, Republic of China
| | - Chun-Liang Pan
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 10002, Taiwan, Republic of China
| |
Collapse
|
19
|
Liao CP, Li H, Lee HH, Chien CT, Pan CL. Cell-Autonomous Regulation of Dendrite Self-Avoidance by the Wnt Secretory Factor MIG-14/Wntless. Neuron 2018; 98:320-334.e6. [DOI: 10.1016/j.neuron.2018.03.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/06/2018] [Accepted: 03/16/2018] [Indexed: 11/26/2022]
|
20
|
Scott-Solomon E, Kuruvilla R. Mechanisms of neurotrophin trafficking via Trk receptors. Mol Cell Neurosci 2018; 91:25-33. [PMID: 29596897 DOI: 10.1016/j.mcn.2018.03.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/19/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022] Open
Abstract
In neurons, long-distance communication between axon terminals and cell bodies is a critical determinant in establishing and maintaining neural circuits. Neurotrophins are soluble factors secreted by post-synaptic target tissues that retrogradely control axon and dendrite growth, survival, and synaptogenesis of innervating neurons. Neurotrophins bind Trk receptor tyrosine kinases in axon terminals to promote endocytosis of ligand-bound phosphorylated receptors into signaling endosomes. Trk-harboring endosomes function locally in axons to acutely promote growth events, and can also be retrogradely transported long-distances to remote cell bodies and dendrites to stimulate cytoplasmic and transcriptional signaling necessary for neuron survival, morphogenesis, and maturation. Neuronal responsiveness to target-derived neurotrophins also requires the precise axonal targeting of newly synthesized Trk receptors. Recent studies suggest that anterograde delivery of Trk receptors is regulated by retrograde neurotrophin signaling. In this review, we summarize current knowledge on the functions and mechanisms of retrograde trafficking of Trk signaling endosomes, and highlight recent discoveries on the forward trafficking of nascent receptors.
Collapse
Affiliation(s)
- Emily Scott-Solomon
- Department of Biology, Johns Hopkins University, 3400 N. Charles St, 227 Mudd Hall, Baltimore, MD 21218, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, 3400 N. Charles St, 227 Mudd Hall, Baltimore, MD 21218, USA.
| |
Collapse
|
21
|
Herrmann KA, Broihier HT. What neurons tell themselves: autocrine signals play essential roles in neuronal development and function. Curr Opin Neurobiol 2018; 51:70-79. [PMID: 29547843 DOI: 10.1016/j.conb.2018.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 12/12/2022]
Abstract
Although retrograde neurotrophin signaling has provided an immensely influential paradigm for understanding growth factor signaling in the nervous system, recent studies indicate that growth factors also signal via cell-autonomous, or autocrine, mechanisms. Autocrine signals have been discovered in many neuronal contexts, providing insights into their regulation and function. The growing realization of the importance of cell-autonomous signaling stems from advances in both conditional genetic approaches and in sophisticated analyses of growth factor dynamics, which combine to enable rigorous in vivo dissection of signaling pathways. Here we review recent studies defining autocrine roles for growth factors such as BDNF, and classical morphogens, including Wnts and BMPs, in regulating neuronal development and plasticity. Collectively, these studies highlight an intimate relationship between activity-dependent autocrine signaling and synaptic plasticity, and further suggest a common principle for coordinating paracrine and autocrine signaling in the nervous system.
Collapse
Affiliation(s)
- Kelsey A Herrmann
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44016, United States
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44016, United States.
| |
Collapse
|
22
|
Becker J, Wilting J. WNT signaling, the development of the sympathoadrenal-paraganglionic system and neuroblastoma. Cell Mol Life Sci 2018; 75:1057-1070. [PMID: 29058015 PMCID: PMC5814469 DOI: 10.1007/s00018-017-2685-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/22/2017] [Accepted: 10/11/2017] [Indexed: 12/04/2022]
Abstract
Neuroblastoma (NB) is a tumor of the sympathoadrenal system arising in children under 15 years of age. In Germany, NB accounts for 7% of childhood cancer cases, but 11% of cancer deaths. It originates from highly migratory progenitor cells that leave the dorsal neural tube and contribute neurons and glial cells to sympathetic ganglia, and chromaffin and supportive cells to the adrenal medulla and paraganglia. Clinically, histologically and molecularly, NBs present as extremely heterogeneous, ranging from very good to very poor prognosis. The etiology of NB still remains unclear and needs to be elucidated, however, aberrant auto- and paracrine embryonic cell communications seem to be likely candidates to initiate or facilitate the emergence, progression and regression of NB. The wingless-type MMTV integration site (WNT) family of proteins represents an evolutionary highly conserved signaling system that orchestrates embryogenesis. At least 19 ligands in the human, numerous receptors and co-receptors are known, which control not only proliferation, but also cell polarity, migration and differentiation. Here we seek to interconnect aspects of WNT signaling with sympathoadrenal and paraganglionic development to define new WNT signaling cues in the etiology and progression of NB.
Collapse
Affiliation(s)
- Jürgen Becker
- Institute of Anatomy and Cell Biology, University Medical School Göttingen, 37075, Göttingen, Germany.
| | - Jörg Wilting
- Institute of Anatomy and Cell Biology, University Medical School Göttingen, 37075, Göttingen, Germany
| |
Collapse
|
23
|
Jang S, Cho HH, Park JS, Jeong HS. Non-canonical Wnt mediated neurogenic differentiation of human bone marrow-derived mesenchymal stem cells. Neurosci Lett 2017; 660:68-73. [PMID: 28916299 DOI: 10.1016/j.neulet.2017.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs), which are characterized by multipotency and self-renewal, are responsible for tissue regeneration and repair. We have previously reported in adipose tissue-derived MSCs that only Wnt5a is enhanced at neurogenic differentiation, and the mechanism of differentiation is dependent on the Wnt5a/JNK pathway; however, the role of Wnt/MAPK pathway is yet to be investigated in neurogenic differentiation in BM-MSCs. We compared the transcriptional expression of Wnt in neurogenic induced-hBM-MSCs (NI-hBM-MSCs) with that in primary hBM-MSCs, using RT-PCR, qPCR, and western blotting. Although the expression of Wnt1 and Wnt2 was unchanged, the expression of Wnt4, Wnt5a, and Wnt11 increased after neurogenic differentiation. In addition, only the expression of frizzled class receptor (Fzd) 3 gene was increased, but not of most of the Fzds and Wnt ligands in NI-hBM-MSCs. Interestingly, Wnt4, Wnt5a, and Wnt11 gene expressions significantly increased in NI-hBM-MSCs by qPCR. In addition, the protein expression level of Wnt4 and Wnt5a, but not Wnt3, increased after neurogenic induction. Furthermore, the expressions of phosphorylated-GSK-3β, ERK1/2, and PKC decreased; however, JNK was activated after neurogenic differentiation. Thus, non-canonical Wnts, i.e., Wnt4, Wnt5a, and Wnt11, regulate neurogenic differentiation through Fzd3 activation and the increase in downstream targets of JNK, which is one of the non-canonical pathways, in hBM-MSCs.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jong-Seong Park
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
24
|
Chan WH, Anderson CR, Gonsalvez DG. From proliferation to target innervation: signaling molecules that direct sympathetic nervous system development. Cell Tissue Res 2017; 372:171-193. [PMID: 28971249 DOI: 10.1007/s00441-017-2693-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
The sympathetic division of the autonomic nervous system includes a variety of cells including neurons, endocrine cells and glial cells. A recent study (Furlan et al. 2017) has revised thinking about the developmental origin of these cells. It now appears that sympathetic neurons and chromaffin cells of the adrenal medulla do not have an immediate common ancestor in the form a "sympathoadrenal cell", as has been long believed. Instead, chromaffin cells arise from Schwann cell precursors. This review integrates the new findings with the expanding body of knowledge on the signalling pathways and transcription factors that regulate the origin of cells of the sympathetic division of the autonomic nervous system.
Collapse
Affiliation(s)
- W H Chan
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - C R Anderson
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
25
|
Susman MW, Karuna EP, Kunz RC, Gujral TS, Cantú AV, Choi SS, Jong BY, Okada K, Scales MK, Hum J, Hu LS, Kirschner MW, Nishinakamura R, Yamada S, Laird DJ, Jao LE, Gygi SP, Greenberg ME, Ho HYH. Kinesin superfamily protein Kif26b links Wnt5a-Ror signaling to the control of cell and tissue behaviors in vertebrates. eLife 2017; 6:e26509. [PMID: 28885975 PMCID: PMC5590807 DOI: 10.7554/elife.26509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022] Open
Abstract
Wnt5a-Ror signaling constitutes a developmental pathway crucial for embryonic tissue morphogenesis, reproduction and adult tissue regeneration, yet the molecular mechanisms by which the Wnt5a-Ror pathway mediates these processes are largely unknown. Using a proteomic screen, we identify the kinesin superfamily protein Kif26b as a downstream target of the Wnt5a-Ror pathway. Wnt5a-Ror, through a process independent of the canonical Wnt/β-catenin-dependent pathway, regulates the cellular stability of Kif26b by inducing its degradation via the ubiquitin-proteasome system. Through this mechanism, Kif26b modulates the migratory behavior of cultured mesenchymal cells in a Wnt5a-dependent manner. Genetic perturbation of Kif26b function in vivo caused embryonic axis malformations and depletion of primordial germ cells in the developing gonad, two phenotypes characteristic of disrupted Wnt5a-Ror signaling. These findings indicate that Kif26b links Wnt5a-Ror signaling to the control of morphogenetic cell and tissue behaviors in vertebrates and reveal a new role for regulated proteolysis in noncanonical Wnt5a-Ror signal transduction.
Collapse
Affiliation(s)
- Michael W Susman
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Edith P Karuna
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Ryan C Kunz
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | - Taranjit S Gujral
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleUnited States
| | - Andrea V Cantú
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Shannon S Choi
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Brigette Y Jong
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Kyoko Okada
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Michael K Scales
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Jennie Hum
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Linda S Hu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Marc W Kirschner
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Ryuichi Nishinakamura
- Department of Kidney DevelopmentInstitute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Soichiro Yamada
- Department of Biomedical EngineeringUniversity of CaliforniaDavisUnited States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Li-En Jao
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Steven P Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | | | - Hsin-Yi Henry Ho
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| |
Collapse
|
26
|
|
27
|
Salehi H, Amirpour N, Niapour A, Razavi S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev Rep 2016; 12:26-41. [PMID: 26490462 DOI: 10.1007/s12015-015-9631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide interest in application of adult stem cells due to easy to obtain with a minimal patient discomfort, capable of producing cell numbers in large quantities and their immunocompatible properties without restriction by ethical concerns. Among these stem cells, multipotent mesenchymal stem cells (MSCs) from human adipose tissue are considered as an ideal source for various regenerative medicine. In spite of mesodermal origin of human adipose-derived stem cells (hADSCs), these cells have differentiation potential toward mesodermal and non-mesodermal lineages. Up to now, several studies have shown that hADSCs can undergo transdifferentiation and produce cells outside of their lineage, especially into neural cells when they are transferred to a specific cell environment. The purpose of this literature review is to provide an overview of the existing state of knowledge of the differentiation potential of hADSCs, specifically their ability to give rise to neuronal cells. The following review discusses different protocols considered for differentiation of hADSCs to neural cells, the neural markers that are used in each procedure and possible mechanisms that are involved in this differentiation.
Collapse
|
28
|
Lin JYS, Wu CL, Liao CN, Higuchi A, Ling QD. Chemogenomic analysis of neuronal differentiation with pathway changes in PC12 cells. MOLECULAR BIOSYSTEMS 2016; 12:283-94. [PMID: 26595144 DOI: 10.1039/c5mb00338e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database creates networks from interrelations between molecular biology and underlying chemical elements. This allows for analysis of biologic networks, genomic information, and higher-order functional information at a system level. Through high throughput experiments and system biology analysis, we investigated the genes and pathways associated with NGF induced neuronal differentiation. We performed microarray experiments and used the KEGG database, system biology analysis, and annotation of pathway functions to study NGF-induced differentiation in PC12 cells. We identified 2020 NGF-induced genes with altered expressions over time. Cross-matching with the KEGG database revealed 830 genes; among which, 395 altered genes were found to have a 2-fold increase in gene expression over a two-hour period. We then identified 191 associated biologic pathways in the KEGG database; the top 15 pathways showed correlation with neural differentiation. These included the neurotrophin pathways, mitogen-activated protein kinase (MAPK) pathways, genes associated with axonal guidance and the Wnt pathways. The activation of these pathways synchronized with nerve growth factor (NGF)-induced differentiation in PC12 cells. In summary, we have established a model system that allows one to systematically characterize the functional pathway changes in a group of neuronal population after an external stimulus.
Collapse
Affiliation(s)
- Jack Yu-Shih Lin
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Chungli, Taiwan, Republic of China. and Taipei Medical University Municipal Wan-Fang Hospital, Taipei, Taiwan, Republic of China
| | - Chien Liang Wu
- Taipei Medical University Municipal Wan-Fang Hospital, Taipei, Taiwan, Republic of China
| | - Chia Nan Liao
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Chungli, Taiwan, Republic of China.
| | - Akon Higuchi
- Department of Chemical & Materials Engineering, National Central University, Chungli, Taiwan, Republic of China and Department of Botany and Microbiology, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Qing-Dong Ling
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Chungli, Taiwan, Republic of China. and Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Shi-Zhi, Taipei, Taiwan, Republic of China.
| |
Collapse
|
29
|
Yu CY, Kuo HC. The Trans-Spliced Long Noncoding RNA ts RMST Impedes Human Embryonic Stem Cell Differentiation Through WNT5A-Mediated Inhibition of the Epithelial-to-Mesenchymal Transition. Stem Cells 2016; 34:2052-2062. [DOI: 10.1002/stem.2386] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The trans-spliced noncoding RNA RMST (tsRMST) is an emerging regulatory lncRNA in the human pluripotency circuit. Previously, we found that tsRMST represses lineage-specific transcription factors through the PRC2 complex and NANOG in human pluripotent stem cells (hESCs). Here, we demonstrate that tsRMST also modulates noncanonical Wnt signaling to suppress the epithelial-to-mesenchymal transition (EMT) and in vitro differentiation of embryonic stem cells (ESCs). Our results demonstrate that disruption of tsRMST expression in hESCs results in the upregulation of WNT5A, EMT, and lineage-specific genes/markers. Furthermore, we found that the PKC inhibitors Go6983 and Go6976 inhibited the effects of WNT5A, indicating that WNT5A promotes the EMT and in vitro differentiation although conventional and novel PKC activation in hESCs. Finally, we showed that either antiserum neutralization of WNT5A or Go6983 treatment in tsRMST knockdown cells decreased the expression of mesenchymal and lineage-specific markers. Together, these findings indicate that tsRMST regulates Wnt and EMT signaling pathways in hESCs by repressing WNT5A, which is a potential EMT inducer for promoting in vitro differentiation of hESCs through PKC activation. Our findings provide further insights into the role of trans-spliced RNA and WNT5A in hESC differentiation, in which EMT plays an important role.
Collapse
Affiliation(s)
- Chun-Ying Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Kumawat K, Gosens R. WNT-5A: signaling and functions in health and disease. Cell Mol Life Sci 2016; 73:567-87. [PMID: 26514730 PMCID: PMC4713724 DOI: 10.1007/s00018-015-2076-y] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 12/14/2022]
Abstract
WNT-5A plays critical roles in a myriad of processes from embryonic morphogenesis to the maintenance of post-natal homeostasis. WNT-5A knock-out mice fail to survive and present extensive structural malformations. WNT-5A predominantly activates β-catenin-independent WNT signaling cascade but can also activate β-catenin signaling to relay its diverse cellular effects such as cell polarity, migration, proliferation, cell survival, and immunomodulation. Moreover, aberrant WNT-5A signaling is associated with several human pathologies such as cancer, fibrosis, and inflammation. Thus, owing to its diverse functions, WNT-5A is a crucial signaling molecule currently under intense investigation with efforts to not only delineate its signaling mechanisms and functions in physiological and pathological conditions, but also to develop strategies for its therapeutic targeting.
Collapse
Affiliation(s)
- Kuldeep Kumawat
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Horigane SI, Ageta-Ishihara N, Kamijo S, Fujii H, Okamura M, Kinoshita M, Takemoto-Kimura S, Bito H. Facilitation of axon outgrowth via a Wnt5a-CaMKK-CaMKIα pathway during neuronal polarization. Mol Brain 2016; 9:8. [PMID: 26772170 PMCID: PMC4715351 DOI: 10.1186/s13041-016-0189-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/10/2016] [Indexed: 11/10/2022] Open
Abstract
Background Wnt5a, originally identified as a guidance cue for commissural axons, activates a non-canonical pathway critical for cortical axonal morphogenesis. The molecular signaling cascade underlying this event remains obscure. Results Through Ca2+ imaging in acute embryonic cortical slices, we tested if radially migrating cortical excitatory neurons that already bore primitive axons were sensitive to Wnt5a. While Wnt5a only evoked brief Ca2+ transients in immature neurons present in the intermediate zone (IZ), Wnt5a-induced Ca2+ oscillations were sustained in neurons that migrated out to the cortical plate (CP). We wondered whether this early Wnt5a-Ca2+ signaling during neuronal polarization has a morphogenetic consequence. During transition from round to polarized shape, Wnt5a administration to immature cultured cortical neurons specifically promoted axonal, but not dendritic, outgrowth. Pharmacological and genetic inhibition of the CaMKK-CaMKIα pathway abolished Wnt5a-induced axonal elongation, and rescue of CaMKIα in CaMKIα-knockdown neurons restored Wnt5a-mediated axon outgrowth. Conclusions This study suggests that Wnt5a activates Ca2+ signaling during a neuronal morphogenetic time window when axon outgrowth is critically facilitated. Furthermore, the CaMKK-CaMKIα cascade is required for the axonal growth effect of Wnt5a during neuronal polarization. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0189-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shin-ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.
| | - Natsumi Ageta-Ishihara
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Satoshi Kamijo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Michiko Okamura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Furo-cho, Chikusa, Nagoya, 464-8602, Japan.
| | - Sayaka Takemoto-Kimura
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan. .,PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, 102-0076, Japan.
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Igakubu-3-gokan, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
32
|
Coullery RP, Ferrari ME, Rosso SB. Neuronal development and axon growth are altered by glyphosate through a WNT non-canonical signaling pathway. Neurotoxicology 2016; 52:150-61. [PMID: 26688330 DOI: 10.1016/j.neuro.2015.12.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/25/2023]
Abstract
The growth and morphological differentiation of neurons are critical events in the establishment of proper neuronal connectivity and functioning. The developing nervous system is highly susceptible to damage caused by exposure to environmental contaminants. Glyphosate-containing herbicides are the most used agrochemicals in the world, particularly on genetically modified plants. Previous studies have demonstrated that glyphosate induces neurotoxicity in mammals. Therefore, its action mechanism on the nervous system needs to be determined. In this study, we report about impaired neuronal development caused by glyphosate exposure. Particularly, we observed that the initial axonal differentiation and growth of cultured neurons is affected by glyphosate since most treated cells remained undifferentiated after 1 day in culture. Although they polarized at 2 days in vitro, they elicited shorter and unbranched axons and they also developed less complex dendritic arbors compared to controls. To go further, we attempted to identify the cellular mechanism by which glyphosate affected neuronal morphology. Biochemical approaches revealed that glyphosate led to a decrease in Wnt5a level, a key factor for the initial neurite development and maturation, as well as inducing a down-regulation of CaMKII activity. This data suggests that the morphological defects would likely be a consequence of the decrease in both Wnt5a expression and CaMKII activity induced by glyphosate. Additionally, these changes might be reflected in a subsequent neuronal dysfunction. Therefore, our findings highlight the importance of establishing rigorous control on the use of glyphosate-based herbicides in order to protect mammals' health.
Collapse
Affiliation(s)
- Romina P Coullery
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - María E Ferrari
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina
| | - Silvana B Rosso
- Experimental Toxicology Laboratory, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, S2002LRK Rosario, Argentina.
| |
Collapse
|
33
|
Patel A, Yamashita N, Ascaño M, Bodmer D, Boehm E, Bodkin-Clarke C, Ryu YK, Kuruvilla R. RCAN1 links impaired neurotrophin trafficking to aberrant development of the sympathetic nervous system in Down syndrome. Nat Commun 2015; 6:10119. [PMID: 26658127 PMCID: PMC4682116 DOI: 10.1038/ncomms10119] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/05/2015] [Indexed: 02/08/2023] Open
Abstract
Down syndrome is the most common chromosomal disorder affecting the nervous system in humans. To date, investigations of neural anomalies in Down syndrome have focused on the central nervous system, although dysfunction of the peripheral nervous system is a common manifestation. The molecular and cellular bases underlying peripheral abnormalities have remained undefined. Here, we report the developmental loss of sympathetic innervation in human Down syndrome organs and in a mouse model. We show that excess regulator of calcineurin 1 (RCAN1), an endogenous inhibitor of the calcineurin phosphatase that is triplicated in Down syndrome, impairs neurotrophic support of sympathetic neurons by inhibiting endocytosis of the nerve growth factor (NGF) receptor, TrkA. Genetically correcting RCAN1 levels in Down syndrome mice markedly improves NGF-dependent receptor trafficking, neuronal survival and innervation. These results uncover a critical link between calcineurin signalling, impaired neurotrophin trafficking and neurodevelopmental deficits in the peripheral nervous system in Down syndrome.
Collapse
Affiliation(s)
- Ami Patel
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Naoya Yamashita
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Maria Ascaño
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Daniel Bodmer
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Erica Boehm
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Chantal Bodkin-Clarke
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Yun Kyoung Ryu
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, 3400N. Charles Street, 224 Mudd Hall, Baltimore, Maryland 21218, USA
| |
Collapse
|
34
|
Snow-Lisy DC, Diaz EC, Bury MI, Fuller NJ, Hannick JH, Ahmad N, Sharma AK. The Role of Genetically Modified Mesenchymal Stem Cells in Urinary Bladder Regeneration. PLoS One 2015; 10:e0138643. [PMID: 26398705 PMCID: PMC4580420 DOI: 10.1371/journal.pone.0138643] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/03/2015] [Indexed: 01/01/2023] Open
Abstract
Recent studies have demonstrated that mesenchymal stem cells (MSCs) combined with CD34+ hematopoietic/stem progenitor cells (HSPCs) can function as surrogate urinary bladder cells to synergistically promote multi-faceted bladder tissue regeneration. However, the molecular pathways governing these events are unknown. The pleiotropic effects of Wnt5a and Cyr61 are known to affect aspects of hematopoiesis, angiogenesis, and muscle and nerve regeneration. Within this study, the effects of Cyr61 and Wnt5a on bladder tissue regeneration were evaluated by grafting scaffolds containing modified human bone marrow derived MSCs. These cell lines were engineered to independently over-express Wnt5a or Cyr61, or to exhibit reduced expression of Cyr61 within the context of a nude rat bladder augmentation model. At 4 weeks post-surgery, data demonstrated increased vessel number (~250 vs ~109 vessels/mm2) and bladder smooth muscle content (~42% vs ~36%) in Cyr61OX (over-expressing) vs Cyr61KD (knock-down) groups. Muscle content decreased to ~25% at 10 weeks in Cyr61KD groups. Wnt5aOX resulted in high numbers of vessels and muscle content (~206 vessels/mm2 and ~51%, respectively) at 4 weeks. Over-expressing cell constructs resulted in peripheral nerve regeneration while Cyr61KD animals were devoid of peripheral nerve regeneration at 4 weeks. At 10 weeks post-grafting, peripheral nerve regeneration was at a minimal level for both Cyr61OX and Wnt5aOX cell lines. Blood vessel and bladder functionality were evident at both time-points in all animals. Results from this study indicate that MSC-based Cyr61OX and Wnt5aOX cell lines play pivotal roles with regards to increasing the levels of functional vasculature, influencing muscle regeneration, and the regeneration of peripheral nerves in a model of bladder augmentation. Wnt5aOX constructs closely approximated the outcomes previously observed with the co-transplantation of MSCs with CD34+ HSPCs and may be specifically targeted as an alternate means to achieve functional bladder regeneration.
Collapse
Affiliation(s)
- Devon C. Snow-Lisy
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
| | - Edward C. Diaz
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
| | - Matthew I. Bury
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
| | - Natalie J. Fuller
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
| | - Jessica H. Hannick
- Department of Urology, Loyola University Health System, Maywood, IL, United States of America
| | - Nida Ahmad
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
| | - Arun K. Sharma
- Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Pediatric Urology, Chicago, IL, United States of America
- Northwestern University Feinberg School of Medicine, Department of Urology, Chicago, IL, United States of America
- Northwestern University, Simpson Querrey Institute for BioNanotechnology, Chicago, IL, United States of America
- Northwestern University, Department of Biomedical Engineering, Evanston, IL, United States of America
- * E-mail:
| |
Collapse
|
35
|
Jang S, Park JS, Jeong HS. Neural Differentiation of Human Adipose Tissue-Derived Stem Cells Involves Activation of the Wnt5a/JNK Signalling. Stem Cells Int 2015; 2015:178618. [PMID: 26106419 PMCID: PMC4461786 DOI: 10.1155/2015/178618] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/13/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022] Open
Abstract
Stem cells are a powerful resource for cell-based transplantation therapies, but understanding of stem cell differentiation at the molecular level is not clear yet. We hypothesized that the Wnt pathway controls stem cell maintenance and neural differentiation. We have characterized the transcriptional expression of Wnt during the neural differentiation of hADSCs. After neural induction, the expressions of Wnt2, Wnt4, and Wnt11 were decreased, but the expression of Wnt5a was increased compared with primary hADSCs in RT-PCR analysis. In addition, the expression levels of most Fzds and LRP5/6 ligand were decreased, but not Fzd3 and Fzd5. Furthermore, Dvl1 and RYK expression levels were downregulated in NI-hADSCs. There were no changes in the expression of ß-catenin and GSK3ß. Interestingly, Wnt5a expression was highly increased in NI-hADSCs by real time RT-PCR analysis and western blot. Wnt5a level was upregulated after neural differentiation and Wnt3, Dvl2, and Naked1 levels were downregulated. Finally, we found that the JNK expression was increased after neural induction and ERK level was decreased. Thus, this study shows for the first time how a single Wnt5a ligand can activate the neural differentiation pathway through the activation of Wnt5a/JNK pathway by binding Fzd3 and Fzd5 and directing Axin/GSK-3ß in hADSCs.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju 501746, Republic of Korea
| | - Jong-Seong Park
- Department of Physiology, Chonnam National University Medical School, Gwangju 501746, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju 501746, Republic of Korea
| |
Collapse
|
36
|
Newbern JM. Molecular control of the neural crest and peripheral nervous system development. Curr Top Dev Biol 2015; 111:201-31. [PMID: 25662262 DOI: 10.1016/bs.ctdb.2014.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A transient and unique population of multipotent stem cells, known as neural crest cells (NCCs), generate a bewildering array of cell types during vertebrate development. An attractive model among developmental biologists, the study of NCC biology has provided a wealth of knowledge regarding the cellular and molecular mechanisms important for embryogenesis. Studies in numerous species have defined how distinct phases of NCC specification, proliferation, migration, and survival contribute to the formation of multiple functionally distinct organ systems. NCC contributions to the peripheral nervous system (PNS) are well known. Critical developmental processes have been defined that provide outstanding models for understanding how extracellular stimuli, cell-cell interactions, and transcriptional networks cooperate to direct cellular diversification and PNS morphogenesis. Dissecting the complex extracellular and intracellular mechanisms that mediate the formation of the PNS from NCCs may have important therapeutic implications for neurocristopathies, neuropathies, and certain forms of cancer.
Collapse
Affiliation(s)
- Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
37
|
Sinha T, Li D, Théveniau-Ruissy M, Hutson MR, Kelly RG, Wang J. Loss of Wnt5a disrupts second heart field cell deployment and may contribute to OFT malformations in DiGeorge syndrome. Hum Mol Genet 2014; 24:1704-16. [PMID: 25410658 DOI: 10.1093/hmg/ddu584] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Outflow tract (OFT) malformation accounts for ∼30% of human congenital heart defects and manifests frequently in TBX1 haplo-insufficiency associated DiGeorge (22q11.2 deletion) syndrome. OFT myocardium originates from second heart field (SHF) progenitors in the pharyngeal and splanchnic mesoderm (SpM), but how these progenitors are deployed to the OFT is unclear. We find that SHF progenitors in the SpM gradually gain epithelial character and are deployed to the OFT as a cohesive sheet. Wnt5a, a non-canonical Wnt, is expressed specifically in the caudal SpM and may regulate oriented cell intercalation to incorporate SHF progenitors into an epithelial-like sheet, thereby generating the pushing force to deploy SHF cells rostrally into the OFT. Using enhancer trap and Cre transgenes, our lineage tracing experiments show that in Wnt5a null mice, SHF progenitors are trapped in the SpM and fail to be deployed to the OFT efficiently, resulting in a reduction in the inferior OFT myocardial wall and its derivative, subpulmonary myocardium. Concomitantly, the superior OFT and subaortic myocardium are expanded. Finally, in chick embryos, blocking the Wnt5a function in the caudal SpM perturbs polarized elongation of SHF progenitors, and compromises their deployment to the OFT. Collectively, our results highlight a critical role for Wnt5a in deploying SHF progenitors from the SpM to the OFT. Given that Wnt5a is a putative transcriptional target of Tbx1, and the similar reduction of subpulmonary myocardium in Tbx1 mutant mice, our results suggest that perturbing Wnt5a-mediated SHF deployment may be an important pathogenic mechanism contributing to OFT malformations in DiGeorge syndrome.
Collapse
Affiliation(s)
- Tanvi Sinha
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | - Ding Li
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | | | - Mary R Hutson
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Robert G Kelly
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Jianbo Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA,
| |
Collapse
|
38
|
Wnt signalling in neuronal differentiation and development. Cell Tissue Res 2014; 359:215-23. [PMID: 25234280 DOI: 10.1007/s00441-014-1996-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/25/2014] [Indexed: 12/15/2022]
Abstract
Wnts are secreted glycoproteins that play multiple roles in early development, including the differentiation of precursor cells. During this period, gradients of Wnts and other morphogens are formed and regulate the differentiation and migration of neural progenitor cells. Afterwards, Wnt signalling cascades participate in the formation of neuronal circuits, playing roles in dendrite and axon development, dendritic spine formation and synaptogenesis. Finally, in the adult brain, Wnts control hippocampal plasticity, regulating synaptic transmission and neurogenesis. In this review, we summarize the reported roles of Wnt signalling cascades in these processes with a particular emphasis on the role of Wnts in neuronal differentiation and development.
Collapse
|
39
|
McLoon LK, Harandi VM, Brännström T, Andersen PM, Liu JX. Wnt and extraocular muscle sparing in amyotrophic lateral sclerosis. Invest Ophthalmol Vis Sci 2014; 55:5482-96. [PMID: 25125606 DOI: 10.1167/iovs.14-14886] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The extraocular muscles (EOM) and their motor neurons are spared in amyotrophic lateral sclerosis (ALS). In limb muscle, axon retraction from the neuromuscular junctions occurs early in the disease. Wnts, a conserved family of secreted signaling molecules, play a critical role in neuromuscular junction formation. This is the first study to examine Wnt signaling for its potential involvement in maintenance of normal morphology in EOM in ALS. METHODS Extraocular muscle and limb muscle axons, neuromuscular junctions, and myofibers from control, aging, and ALS subjects and the SOD1(G93A) mouse model of ALS were quantified for their expression of Wnt1, Wnt3a, Wnt5a, Wnt7a, and β-catenin. RESULTS All four Wnt isoforms were expressed in most axon profiles in all human EOM. Significantly fewer were positive for Wnt1, Wnt3a, and Wnt7a in the human limb muscles. Similar differential patterns in Wnt myofiber expression were also seen except in the case of Wnt7a, where expression was elevated. In the SOD1(G93A) mouse, all four Wnt isoforms were significantly decreased in the neuromuscular junctions at the terminal stage compared to values in age-matched controls. β-Catenin was activated in a subset of myofibers in EOM and limb muscle in all subjects. CONCLUSIONS The differences in expression of Wnts in EOM and limb muscle, particularly at the neuromuscular junction level, suggest that they play a role in the pathophysiology of ALS. Collectively, the data support a role for signaling of Wnts in the preservation of the EOM in ALS and their dysregulation and the subsequent development of pathology in the ALS limb muscles.
Collapse
Affiliation(s)
- Linda K McLoon
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | - Vahid M Harandi
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | - Thomas Brännström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Jing-Xia Liu
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| |
Collapse
|
40
|
Wan W, Xia S, Kalionis B, Liu L, Li Y. The role of Wnt signaling in the development of Alzheimer's disease: a potential therapeutic target? BIOMED RESEARCH INTERNATIONAL 2014; 2014:301575. [PMID: 24883305 PMCID: PMC4026919 DOI: 10.1155/2014/301575] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 04/10/2014] [Indexed: 12/31/2022]
Abstract
Accumulating evidence supports a key role for Wnt signaling in the development of the central nervous system (CNS) during embryonic development and in the regulation of the structure and function of the adult brain. Alzheimer's disease (AD) is the most common form of senile dementia, which is characterized by β -amyloid (A β ) deposition in specific brain regions. However, the molecular mechanism underlying AD pathology remains elusive. Dysfunctional Wnt signaling is associated with several diseases such as epilepsy, cancer, metabolic disease, and AD. Increasing evidence suggests that downregulation of Wnt signaling, induced by A β , is associated with disease progression of AD. More importantly, persistent activation of Wnt signaling through Wnt ligands, or inhibition of negative regulators of Wnt signaling, such as Dickkopf-1 (DKK-1) and glycogen synthase kinase-3 β (GSK-3 β ) that are hyperactive in the disease state, is able to protect against A β toxicity and ameliorate cognitive performance in AD. Together, these data suggest that Wnt signaling might be a potential therapeutic target of AD. Here, we review recent studies related to the progression of AD where Wnt signaling might be relevant and participate in the development of the disease. Then, we focus on the potential relevance of manipulating the Wnt signaling pathway for the treatment of AD.
Collapse
Affiliation(s)
- Wenbin Wan
- Geriatrics Department of Traditional Chinese Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Bill Kalionis
- Department of Perinatal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Lumei Liu
- Geriatrics Department of Traditional Chinese Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Yaming Li
- Geriatrics Department of Traditional Chinese Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
41
|
Branch management: mechanisms of axon branching in the developing vertebrate CNS. Nat Rev Neurosci 2014; 15:7-18. [PMID: 24356070 DOI: 10.1038/nrn3650] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The remarkable ability of a single axon to extend multiple branches and form terminal arbors enables vertebrate neurons to integrate information from divergent regions of the nervous system. Axons select appropriate pathways during development, but it is the branches that extend interstitially from the axon shaft and arborize at specific targets that are responsible for virtually all of the synaptic connectivity in the vertebrate CNS. How do axons form branches at specific target regions? Recent studies have identified molecular cues that activate intracellular signalling pathways in axons and mediate dynamic reorganization of the cytoskeleton to promote the formation of axon branches.
Collapse
|
42
|
Inhibiting geranylgeranylation increases neurite branching and differentially activates cofilin in cell bodies and growth cones. Mol Neurobiol 2014; 50:49-59. [PMID: 24515839 DOI: 10.1007/s12035-014-8653-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/23/2014] [Indexed: 10/25/2022]
Abstract
Inhibitors of the mevalonate pathway, including the highly prescribed statins, reduce the production of cholesterol and isoprenoids such as geranylgeranyl pyrophosphates. The Rho family of small guanine triphosphatases (GTPases) requires isoprenylation, specifically geranylgeranylation, for activation. Because Rho GTPases are primary regulators of actin filament rearrangements required for process extension, neurite arborization, and synaptic plasticity, statins may affect cognition or recovery from nervous system injury. Here, we assessed how manipulating geranylgeranylation affects neurite initiation, elongation, and branching in neuroblastoma growth cones. Treatment with the statin, lovastatin (20 μM), decreased measures of neurite initiation by 17.0 to 19.0 % when a source of cholesterol was present and increased neurite branching by 4.03- to 9.54-fold (regardless of exogenous cholesterol). Neurite elongation was increased by treatment with lovastatin only in cholesterol-free culture conditions. Treatment with lovastatin decreased growth cone actin filament content by up to 24.3 %. In all cases, co-treatment with the prenylation precursor, geranylgeraniol (10 μM), reversed the effect of lovastatin. In a prior work, statin effects on outgrowth were linked to modulating the actin depolymerizing factor, cofilin. In our assays, treatment with lovastatin or geranylgeraniol decreased cofilin phosphorylation in whole cell lysates. However, lovastatin increased cofilin phosphorylation in cell bodies and decreased it in growth cones, indicating differential regulation in specific cell regions. Together, we interpret these data to suggest that protein geranylgeranylation likely regulates growth cone actin filament content and subsequent neurite outgrowth through mechanisms that also affect actin nucleation and polymerization.
Collapse
|
43
|
Clark CEJ, Richards LJ, Stacker SA, Cooper HM. Wnt5a induces Ryk-dependent and -independent effects on callosal axon and dendrite growth. Growth Factors 2014; 32:11-7. [PMID: 24471468 DOI: 10.3109/08977194.2013.875544] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The non-canonical Wnt receptor, Ryk, promotes chemorepulsive axon guidance in the developing mouse brain and spinal cord in response to Wnt5a. Ryk has also been identified as a major suppressor of axonal regrowth after spinal cord injury. Thus, a comprehensive understanding of how growing axons and dendrites respond to Wnt5a-mediated Ryk activation is required if we are to overcome this detrimental activity. Here we undertook a detailed analysis of the effect of Wnt5a/Ryk interactions on axonal and dendritic growth in dissociated embryonic mouse cortical neuron cultures, focusing on callosal neurons known to be responsive to Ryk-induced chemorepulsion. We show that Ryk inhibits axonal growth in response to Wnt5a. We also show that Wnt5a inhibits dendrite growth independently of Ryk. However, this inhibition is relieved when Ryk is present. Therefore, Wnt5a-mediated Ryk activation triggers divergent responses in callosal axons and dendrites in the in vitro context.
Collapse
Affiliation(s)
- Charlotte E J Clark
- Queensland Brain Institute, The University of Queensland, St Lucia , Queensland , Australia
| | | | | | | |
Collapse
|
44
|
Slater PG, Ramirez VT, Gonzalez-Billault C, Varela-Nallar L, Inestrosa NC. Frizzled-5 receptor is involved in neuronal polarity and morphogenesis of hippocampal neurons. PLoS One 2013; 8:e78892. [PMID: 24205342 PMCID: PMC3800132 DOI: 10.1371/journal.pone.0078892] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 09/17/2013] [Indexed: 01/09/2023] Open
Abstract
The Wnt signaling pathway plays important roles during different stages of neuronal development, including neuronal polarization and dendritic and axonal outgrowth. However, little is known about the identity of the Frizzled receptors mediating these processes. In the present study, we investigated the role of Frizzled-5 (Fzd5) on neuronal development in cultured Sprague-Dawley rat hippocampal neurons. We found that Fzd5 is expressed early in cultured neurons on actin-rich structures localized at minor neurites and axonal growth cones. At 4 DIV, Fzd5 polarizes towards the axon, where its expression is detected mainly at the peripheral zone of axonal growth cones, with no obvious staining at dendrites; suggesting a role of Fzd5 in neuronal polarization. Overexpression of Fzd5 during the acquisition of neuronal polarity induces mislocalization of the receptor and a loss of polarized axonal markers. Fzd5 knock-down leads to loss of axonal proteins, suggesting an impaired neuronal polarity. In contrast, overexpression of Fzd5 in neurons that are already polarized did not alter polarity, but decreased the total length of axons and increased total dendrite length and arborization. Fzd5 activated JNK in HEK293 cells and the effects triggered by Fzd5 overexpression in neurons were partially prevented by inhibition of JNK, suggesting that a non-canonical Wnt signaling mechanism might be involved. Our results suggest that, Fzd5 has a role in the establishment of neuronal polarity, and in the morphogenesis of neuronal processes, in part through the activation of the non-canonical Wnt mechanism involving JNK.
Collapse
Affiliation(s)
- Paula G. Slater
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Valerie T. Ramirez
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | | | - Lorena Varela-Nallar
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
- * . E-mail:
| |
Collapse
|
45
|
Rosso SB, Inestrosa NC. WNT signaling in neuronal maturation and synaptogenesis. Front Cell Neurosci 2013; 7:103. [PMID: 23847469 PMCID: PMC3701138 DOI: 10.3389/fncel.2013.00103] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/12/2013] [Indexed: 01/01/2023] Open
Abstract
The Wnt signaling pathway plays a role in the development of the central nervous system and growing evidence indicates that Wnts also regulates the structure and function of the adult nervous system. Wnt components are key regulators of a variety of developmental processes, including embryonic patterning, cell specification, and cell polarity. In the nervous system, Wnt signaling also regulates the formation and function of neuronal circuits by controlling neuronal differentiation, axon outgrowth and guidance, dendrite development, synaptic function, and neuronal plasticity. Wnt factors can signal through three very well characterized cascades: canonical or β-catenin pathway, planar cell polarity pathway and calcium pathway that control different processes. However, divergent downstream cascades have been identified to control neuronal morphogenesis. In the nervous system, the expression of Wnt proteins is a highly controlled process. In addition, deregulation of Wnt signaling has been associated with neurodegenerative diseases. Here, we will review different aspects of neuronal and dendrite maturation, including spinogenesis and synaptogenesis. Finally, the role of Wnt pathway components on Alzheimer’s disease will be revised.
Collapse
Affiliation(s)
- Silvana B Rosso
- Laboratorio de Toxicología Experimental, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario Rosario, Santa Fe, Argentina
| | | |
Collapse
|
46
|
Hiester BG, Galati DF, Salinas PC, Jones KR. Neurotrophin and Wnt signaling cooperatively regulate dendritic spine formation. Mol Cell Neurosci 2013; 56:115-27. [PMID: 23639831 PMCID: PMC3793870 DOI: 10.1016/j.mcn.2013.04.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 04/20/2013] [Accepted: 04/23/2013] [Indexed: 01/13/2023] Open
Abstract
Dendritic spines are major sites of excitatory synaptic transmission and changes in their numbers and morphology have been associated with neurodevelopmental and neurodegenerative disorders. Brain-derived Neurotrophic Factor (BDNF) is a secreted growth factor that influences hippocampal, striatal and neocortical pyramidal neuron dendritic spine density. However, the mechanisms by which BDNF regulates dendritic spines and how BDNF interacts with other regulators of spines remain unclear. We propose that one mechanism by which BDNF promotes dendritic spine formation is through an interaction with Wnt signaling. Here, we show that Wnt signaling inhibition in cultured cortical neurons disrupts dendritic spine development, reduces dendritic arbor size and complexity, and blocks BDNF-induced dendritic spine formation and maturation. Additionally, we show that BDNF regulates expression of Wnt2, and that Wnt2 is sufficient to promote cortical dendrite growth and dendritic spine formation. Together, these data suggest that BDNF and Wnt signaling cooperatively regulate dendritic spine formation.
Collapse
Affiliation(s)
- Brian G Hiester
- Department of Molecular, Cellular and Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309, United States
| | | | | | | |
Collapse
|
47
|
A sympathetic neuron autonomous role for Egr3-mediated gene regulation in dendrite morphogenesis and target tissue innervation. J Neurosci 2013; 33:4570-83. [PMID: 23467373 DOI: 10.1523/jneurosci.5481-12.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Egr3 is a nerve growth factor (NGF)-induced transcriptional regulator that is essential for normal sympathetic nervous system development. Mice lacking Egr3 in the germline have sympathetic target tissue innervation abnormalities and physiologic sympathetic dysfunction similar to humans with dysautonomia. However, since Egr3 is widely expressed and has pleiotropic function, it has not been clear whether it has a role within sympathetic neurons and if so, what target genes it regulates to facilitate target tissue innervation. Here, we show that Egr3 expression within sympathetic neurons is required for their normal innervation since isolated sympathetic neurons lacking Egr3 have neurite outgrowth abnormalities when treated with NGF and mice with sympathetic neuron-restricted Egr3 ablation have target tissue innervation abnormalities similar to mice lacking Egr3 in all tissues. Microarray analysis performed on sympathetic neurons identified many target genes deregulated in the absence of Egr3, with some of the most significantly deregulated genes having roles in axonogenesis, dendritogenesis, and axon guidance. Using a novel genetic technique to visualize axons and dendrites in a subpopulation of randomly labeled sympathetic neurons, we found that Egr3 has an essential role in regulating sympathetic neuron dendrite morphology and terminal axon branching, but not in regulating sympathetic axon guidance to their targets. Together, these results indicate that Egr3 has a sympathetic neuron autonomous role in sympathetic nervous system development that involves modulating downstream target genes affecting the outgrowth and branching of sympathetic neuron dendrites and axons.
Collapse
|
48
|
Matusica D, Skeldal S, Sykes AM, Palstra N, Sharma A, Coulson EJ. An intracellular domain fragment of the p75 neurotrophin receptor (p75(NTR)) enhances tropomyosin receptor kinase A (TrkA) receptor function. J Biol Chem 2013; 288:11144-54. [PMID: 23471969 PMCID: PMC3630867 DOI: 10.1074/jbc.m112.436469] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/17/2013] [Indexed: 11/06/2022] Open
Abstract
Facilitation of nerve growth factor (NGF) signaling by the p75 neurotrophin receptor (p75(NTR)) is critical for neuronal survival and differentiation. However, the interaction between p75(NTR) and TrkA receptors required for this activity is not understood. Here, we report that a specific 29-amino acid peptide derived from the intracellular domain fragment of p75(NTR) interacts with and potentiates binding of NGF to TrkA-expressing cells, leading to increased neurite outgrowth in sympathetic neurons as a result of enhanced Erk1/2 and Akt signaling. An endogenous intracellular domain fragment of p75(NTR) (p75(ICD)) containing these 29 amino acids is produced by regulated proteolysis of the full-length receptor. We demonstrate that generation of this fragment is a requirement for p75(NTR) to facilitate TrkA signaling in neurons and propose that the juxtamembrane region of p75(ICD) acts to cause a conformational change within the extracellular domain of TrkA. This finding provides new insight into the mechanism by which p75(NTR) and TrkA interact to enhance neurotrophic signaling.
Collapse
Affiliation(s)
- Dusan Matusica
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sune Skeldal
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alex M. Sykes
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nickless Palstra
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Aanchal Sharma
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elizabeth J. Coulson
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
49
|
An autocrine Wnt5a-Ror signaling loop mediates sympathetic target innervation. Dev Biol 2013; 377:79-89. [PMID: 23454479 DOI: 10.1016/j.ydbio.2013.02.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/15/2013] [Accepted: 02/15/2013] [Indexed: 11/24/2022]
Abstract
During nervous system development, axon branching at nerve terminals is an essential step in the formation of functional connections between neurons and target cells. It is known that target tissues exert control of terminal arborization through secretion of trophic factors. However, whether the in-growing axons themselves produce diffusible cues to instruct target innervation remains unclear. Here, we use conditional mutant mice to show that Wnt5a derived from sympathetic neurons is required for their target innervation in vivo. Conditional deletion of Wnt5a resulted in specific deficits in the extension and arborization of sympathetic fibers in their final target fields, while no defects were observed in the overall tissue patterning, proliferation, migration or differentiation of neuronal progenitors. Using compartmentalized neuronal cultures, we further demonstrate that the Ror receptor tyrosine kinases are required locally in sympathetic axons to mediate Wnt5a-dependent branching. Thus, our study suggests an autocrine Wnt5a-Ror signaling pathway that directs sympathetic axon branching during target innervation.
Collapse
|
50
|
Ter-Avetisyan G, Tröster P, Schmidt H, Rathjen FG. cGMP signaling and branching of sensory axons in the spinal cord. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Axonal branching is essential for neurons to establish contacts to different targets. It therefore provides the physical basis for the integration and distribution of information within the nervous system. During embryonic and early postnatal development, several axonal branching modes may be distinguished that might be regulated by activities of the growth cone or by the axon shaft. The various forms of axonal branching are dependent on intrinsic components and are regulated by extrinsic factors that activate specific signaling systems. This article focuses on components implicated in cyclic guanosine monophosphate signaling that regulate axon bifurcation – a specific form of branching – within the spinal cord in animal models. This cascade is composed of the ligand CNP, the guanylyl cyclase Npr2 and the cyclic guanosine monophosphate-dependent kinase I. In the absence of one of these components, axons of dorsal root ganglion neurons do not form T-shaped branches when entering the spinal cord, while collateral (interstitial) branching, another branching mode of the same type of the neuron, is not affected. It will be important to analyze human patients with mutations in the corresponding genes to get insights into the pathophysiological effects of impaired sensory axon branching in the spinal cord.
Collapse
Affiliation(s)
- Gohar Ter-Avetisyan
- MaxDelbrück Center of Molecular Medicine, Robert-Rössle-Str.10, 13092 Berlin, Germany
| | - Philip Tröster
- MaxDelbrück Center of Molecular Medicine, Robert-Rössle-Str.10, 13092 Berlin, Germany
| | - Hannes Schmidt
- MaxDelbrück Center of Molecular Medicine, Robert-Rössle-Str.10, 13092 Berlin, Germany
| | - Fritz G Rathjen
- MaxDelbrück Center of Molecular Medicine, Robert-Rössle-Str.10, 13092 Berlin, Germany
| |
Collapse
|