1
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
Miyazawa K, Itoh Y, Fu H, Miyazono K. Receptor-activated transcription factors and beyond: multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem 2024; 300:107256. [PMID: 38569937 PMCID: PMC11063908 DOI: 10.1016/j.jbc.2024.107256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine that is widely distributed throughout the body. Its receptor proteins, TGF-β type I and type II receptors, are also ubiquitously expressed. Therefore, the regulation of various signaling outputs in a context-dependent manner is a critical issue in this field. Smad proteins were originally identified as signal-activated transcription factors similar to signal transducer and activator of transcription proteins. Smads are activated by serine phosphorylation mediated by intrinsic receptor dual specificity kinases of the TGF-β family, indicating that Smads are receptor-restricted effector molecules downstream of ligands of the TGF-β family. Smad proteins have other functions in addition to transcriptional regulation, including post-transcriptional regulation of micro-RNA processing, pre-mRNA splicing, and m6A methylation. Recent technical advances have identified a novel landscape of Smad-dependent signal transduction, including regulation of mitochondrial function without involving regulation of gene expression. Therefore, Smad proteins are receptor-activated transcription factors and also act as intracellular signaling modulators with multiple modes of function. In this review, we discuss the role of Smad proteins as receptor-activated transcription factors and beyond. We also describe the functional differences between Smad2 and Smad3, two receptor-activated Smad proteins downstream of TGF-β, activin, myostatin, growth and differentiation factor (GDF) 11, and Nodal.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hao Fu
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory for Cancer Invasion and Metastasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
El Hayek L, DeVries D, Gogate A, Aiken A, Kaur K, Chahrour MH. Disruption of the autism gene and chromatin regulator KDM5A alters hippocampal cell identity. SCIENCE ADVANCES 2023; 9:eadi0074. [PMID: 37992166 PMCID: PMC10664992 DOI: 10.1126/sciadv.adi0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Abstract
Chromatin regulation plays a pivotal role in establishing and maintaining cellular identity and is one of the top pathways disrupted in autism spectrum disorder (ASD). The hippocampus, composed of distinct cell types, is often affected in patients with ASD. However, the specific hippocampal cell types and their transcriptional programs that are dysregulated in ASD are unknown. Using single-nucleus RNA sequencing, we show that the ASD gene, lysine demethylase 5A (KDM5A), regulates the development of specific subtypes of excitatory and inhibitory neurons. We found that KDM5A is essential for establishing hippocampal cell identity by controlling a differentiation switch early in development. Our findings define a role for the chromatin regulator KDM5A in establishing hippocampal cell identity and contribute to the emerging convergent mechanisms across ASD.
Collapse
Affiliation(s)
- Lauretta El Hayek
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Darlene DeVries
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashlesha Gogate
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ariel Aiken
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kiran Kaur
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Maria H. Chahrour
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
4
|
Ohyama K, Shinohara HM, Omura S, Kawachi T, Sato T, Toda K. PSmad3+/Olig2- expression defines a subpopulation of gfap-GFP+/Sox9+ neural progenitors and radial glia-like cells in mouse dentate gyrus through embryonic and postnatal development. Front Neurosci 2023; 17:1204012. [PMID: 37795190 PMCID: PMC10547214 DOI: 10.3389/fnins.2023.1204012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/29/2023] [Indexed: 10/06/2023] Open
Abstract
In mouse dentate gyrus, radial glia-like cells (RGLs) persist throughout life and play a critical role in the generation of granule neurons. A large body of evidence has shown that the combinatorial expression of transcription factors (TFs) defines cell types in the developing central nervous system (CNS). As yet, the identification of specific TFs that exclusively define RGLs in the developing mouse dentate gyrus (DG) remains elusive. Here we show that phospho-Smad3 (PSmad3) is expressed in a subpopulation of neural progenitors in the DG. During embryonic stage (E14-15), PSmad3 was predominantly expressed in gfap-GFP-positive (GFP+)/Sox2+ progenitors located at the lower dentate notch (LDN). As the development proceeds (E16-17), the vast majority of PSmad3+ cells were GFP+/Sox2+/Prox1low+/Ki67+ proliferative progenitors that eventually differentiated into granule neurons. During postnatal stage (P1-P6) PSmad3 expression was observed in GFP+ progenitors and astrocytes. Subsequently, at P14-P60, PSmad3 expression was found both in GFP+ RGLs in the subgranular zone (SGZ) and astrocytes in the molecular layer (ML) and hilus. Notably, PSmad3+ SGZ cells did not express proliferation markers such as PCNA and phospho-vimentin, suggesting that they are predominantly quiescent from P14 onwards. Significantly PSmad3+/GFP+ astrocytes, but not SGZ cells, co-expressed Olig2 and S100β. Together, PSmad3+/Olig2- expression serves as an exclusive marker for a specific subpopulation of GFP+ neural progenitors and RGLs in the mouse DG during both embryonic and postnatal period.
Collapse
Affiliation(s)
- Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
5
|
Raus AM, Fuller TD, Nelson NE, Valientes DA, Bayat A, Ivy AS. Early-life exercise primes the murine neural epigenome to facilitate gene expression and hippocampal memory consolidation. Commun Biol 2023; 6:18. [PMID: 36611093 PMCID: PMC9825372 DOI: 10.1038/s42003-022-04393-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
Aerobic exercise is well known to promote neuroplasticity and hippocampal memory. In the developing brain, early-life exercise (ELE) can lead to persistent improvements in hippocampal function, yet molecular mechanisms underlying this phenomenon have not been fully explored. In this study, transgenic mice harboring the "NuTRAP" (Nuclear tagging and Translating Ribosome Affinity Purification) cassette in Emx1 expressing neurons ("Emx1-NuTRAP" mice) undergo ELE during adolescence. We then simultaneously isolate and sequence translating mRNA and nuclear chromatin from single hippocampal homogenates containing Emx1-expressing neurons. This approach allowed us to couple translatomic with epigenomic sequencing data to evaluate the influence of histone modifications H4K8ac and H3K27me3 on translating mRNA after ELE. A subset of ELE mice underwent a hippocampal learning task to determine the gene expression and epigenetic underpinnings of ELE's contribution to improved hippocampal memory performance. From this experiment, we discover gene expression - histone modification relationships that may play a critical role in facilitated memory after ELE. Our data reveal candidate gene-histone modification interactions and implicate gene regulatory pathways involved in ELE's impact on hippocampal memory.
Collapse
Affiliation(s)
- Anthony M Raus
- Physiology/Biophysics, Anatomy/Neurobiology, University of California- Irvine School of Medicine, Irvine, CA, USA
| | - Tyson D Fuller
- Pediatrics, University of California- Irvine School of Medicine, Irvine, CA, USA
| | - Nellie E Nelson
- Physiology/Biophysics, Anatomy/Neurobiology, University of California- Irvine School of Medicine, Irvine, CA, USA
| | - David A Valientes
- Pediatrics, University of California- Irvine School of Medicine, Irvine, CA, USA
| | - Anita Bayat
- Pediatrics, University of California- Irvine School of Medicine, Irvine, CA, USA
| | - Autumn S Ivy
- Physiology/Biophysics, Anatomy/Neurobiology, University of California- Irvine School of Medicine, Irvine, CA, USA.
- Pediatrics, University of California- Irvine School of Medicine, Irvine, CA, USA.
- Neurobiology/Behavior, University of California- Irvine School of Biological Sciences, Irvine, CA, USA.
- Anatomy/Neurobiology, University of California- Irvine School of Medicine, Irvine, CA, USA.
- Division of Neurology, Children's Hospital Orange County, Orange, CA, USA.
| |
Collapse
|
6
|
The promise of the TGF-β superfamily as a therapeutic target for Parkinson's disease. Neurobiol Dis 2022; 171:105805. [PMID: 35764291 DOI: 10.1016/j.nbd.2022.105805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
A large body of evidence underscore the regulatory role of TGF-β superfamily in the central nervous system. Components of the TGF-β superfamily modulate key events during embryonic brain development and adult brain tissue injury repair. With respect to Parkinson's disease (PD), TGF-ß signaling pathways are implicated in the differentiation, maintenance and synaptic function of the dopaminergic neurons, as well as in processes related to the activation state of astrocytes and microglia. In vitro and in vivo studies using toxin models, have interrogated on the dopaminotrophic and protective role of the TGF-β superfamily members. The evolution of genetic and animal models of PD that more closely recapitulate the disease condition has made possible the dissection of intracellular pathways in response to TGF-ß treatment. Although the first clinical trials using GDNF did not meet their primary endpoints, substantial work has been carried out to reappraise the TGF-β superfamily's clinical benefit.
Collapse
|
7
|
Gradari S, Herrera A, Tezanos P, Fontán-Lozano Á, Pons S, Trejo JL. The Role of Smad2 in Adult Neuroplasticity as Seen through Hippocampal-Dependent Spatial Learning/Memory and Neurogenesis. J Neurosci 2021; 41:6836-6849. [PMID: 34210778 PMCID: PMC8360684 DOI: 10.1523/jneurosci.2619-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 11/21/2022] Open
Abstract
Adult neural plasticity is an important and intriguing phenomenon in the brain, and adult hippocampal neurogenesis is directly involved in modulating neural plasticity by mechanisms that are only partially understood. We have performed gain-of-function and loss-of-function experiments to study Smad2, a transcription factor selected from genes that are demethylated after exercise through the analysis of an array of physical activity-induced factors, and their corresponding gene expression, and an efficient inducer of plasticity. In these studies, changes in cell number and morphology were analyzed in the hippocampal dentate gyrus (cell proliferation and survival, including regional distribution, and structural maturation/differentiation, including arborization, dendritic spines, and neurotransmitter-specific vesicles) of sedentary male mice, after evaluation in a battery of behavioral tests. As a result, we reveal a role for Smad2 in the balance of proliferation versus maturation of differentiating immature cells (Smad2 silencing increases both the proliferation and survival of cycling cells in the dentate granule cell layer), and in the plasticity of both newborn and mature neurons in mice (by decreasing dendritic arborization and dendritic spine number). Moreover, Smad2 silencing specifically compromises spatial learning in mice (through impairments of spatial tasks acquisition both in long-term learning and working memory). These data suggest that Smad2 participates in adult neural plasticity by influencing the proliferation and maturation of dentate gyrus neurons.SIGNIFICANCE STATEMENT Smad2 is one of the main components of the transforming growth factor-β (TGF-β) pathway. The commitment of cell fate in the nervous system is tightly coordinated by SMAD2 signaling, as are further differentiation steps (e.g., dendrite and axon growth, myelination, and synapse formation). However, there are no studies that have directly evaluated the role of Smad2 gene in hippocampus of adult animals. Modulation of these parameters in the adult hippocampus can affect hippocampal-dependent behaviors, which may shed light on the mechanisms that regulate adult neurogenesis and behavior. We demonstrate here a role for Smad2 in the maturation of differentiating immature cells and in the plasticity of mature neurons. Moreover, Smad2 silencing specifically compromises the spatial learning abilities of adult male mice.
Collapse
Affiliation(s)
- Simona Gradari
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | - Antonio Herrera
- Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - Patricia Tezanos
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | - Ángela Fontán-Lozano
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
- Department of Physiology, School of Biology, University of Sevilla, 41004 Sevilla, Spain
| | - Sebastián Pons
- Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas, 08028 Barcelona, Spain
| | - José Luis Trejo
- Cajal Institute, Translational Neuroscience Department, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| |
Collapse
|
8
|
TGF-β/Smad Signalling in Neurogenesis: Implications for Neuropsychiatric Diseases. Cells 2021; 10:cells10061382. [PMID: 34205102 PMCID: PMC8226492 DOI: 10.3390/cells10061382] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
TGF-β/Smad signalling has been the subject of extensive research due to its role in the cell cycle and carcinogenesis. Modifications to the TGF-β/Smad signalling pathway have been found to produce disparate effects on neurogenesis. We review the current research on canonical and non-canonical TGF-β/Smad signalling pathways and their functions in neurogenesis. We also examine the observed role of neurogenesis in neuropsychiatric disorders and the relationship between TGF-β/Smad signalling and neurogenesis in response to stressors. Overlapping mechanisms of cell proliferation, neurogenesis, and the development of mood disorders in response to stressors suggest that TGF-β/Smad signalling is an important regulator of stress response and is implicated in the behavioural outcomes of mood disorders.
Collapse
|
9
|
TGF-β/Smad3 Signalling Modulates GABA Neurotransmission: Implications in Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21020590. [PMID: 31963327 PMCID: PMC7013528 DOI: 10.3390/ijms21020590] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
γ-Aminobutiryc acid (GABA) is found extensively in different brain nuclei, including parts involved in Parkinson’s disease (PD), such as the basal ganglia and hippocampus. In PD and in different models of the disorder, an increase in GABA neurotransmission is observed and may promote bradykinesia or L-Dopa-induced side-effects. In addition, proteins involved in GABAA receptor (GABAAR) trafficking, such as GABARAP, Trak1 or PAELR, may participate in the aetiology of the disease. TGF-β/Smad3 signalling has been associated with several pathological features of PD, such as dopaminergic neurodegeneration; reduction of dopaminergic axons and dendrites; and α-synuclein aggregation. Moreover, TGF-β/Smad3 intracellular signalling was recently shown to modulate GABA neurotransmission in the context of parkinsonism and cognitive alterations. This review provides a summary of GABA neurotransmission and TGF-β signalling; their implications in PD; and the regulation of GABA neurotransmission by TGF-β/Smad3. There appear to be new possibilities to develop therapeutic approaches for the treatment of PD using GABA modulators.
Collapse
|
10
|
Fuchs C, Medici G, Trazzi S, Gennaccaro L, Galvani G, Berteotti C, Ren E, Loi M, Ciani E. CDKL5 deficiency predisposes neurons to cell death through the deregulation of SMAD3 signaling. Brain Pathol 2019; 29:658-674. [PMID: 30793413 DOI: 10.1111/bpa.12716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
CDKL5 deficiency disorder (CDD) is a rare encephalopathy characterized by early onset epilepsy and severe intellectual disability. CDD is caused by mutations in the X-linked cyclin-dependent kinase-like 5 (CDKL5) gene, a member of a highly conserved family of serine-threonine kinases. Only a few physiological substrates of CDKL5 are currently known, which hampers the discovery of therapeutic strategies for CDD. Here, we show that SMAD3, a primary mediator of TGF-β action, is a direct phosphorylation target of CDKL5 and that CDKL5-dependent phosphorylation promotes SMAD3 protein stability. Importantly, we found that restoration of the SMAD3 signaling through TGF-β1 treatment normalized defective neuronal survival and maturation in Cdkl5 knockout (KO) neurons. Moreover, we demonstrate that Cdkl5 KO neurons are more vulnerable to neurotoxic/excitotoxic stimuli. In vivo treatment with TGF-β1 prevents increased NMDA-induced cell death in hippocampal neurons from Cdkl5 KO mice, suggesting an involvement of the SMAD3 signaling deregulation in the neuronal susceptibility to excitotoxic injury of Cdkl5 KO mice. Our finding reveals a new function for CDKL5 in maintaining neuronal survival that could have important implications for susceptibility to neurodegeneration in patients with CDD.
Collapse
Affiliation(s)
- Claudia Fuchs
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Medici
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Laura Gennaccaro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Galvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisa Ren
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Manuela Loi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Smith K, Semënov MV. The impact of age on number and distribution of proliferating cells in subgranular zone in adult mouse brain. IBRO Rep 2018; 6:18-30. [PMID: 30582065 PMCID: PMC6297242 DOI: 10.1016/j.ibror.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/07/2018] [Indexed: 01/16/2023] Open
Abstract
The mouse brain retains an ability to produce hippocampal granule neurons during the mouse’s entire lifespan. The neurons are produced in the subgranular zone (SGZ) located on the inner surface of the granule cell layer in the dentate gyrus (DG). In our study, we used a point cloud approach to characterize how the production and distribution of neural precursors for new hippocampal neurons change in the mouse brain relative to age. We found that the production of neural precursors decreases 64 fold from the age of 30 days to the age of 2.5 years. Within the SGZ the decline of cell proliferation continues during entire mouse life. We reconstructed the distribution of proliferating cells along the longitudinal axis of the SGZ and found that the highest number of proliferating cells are located approximately 0.75 mm from the dorsomedial end of the SGZ that corresponds to the most dorsal part of the DG in the mouse brain. The distribution of proliferating cells in the SGZ showed no apparent aggregations, periodicity or any other readily identifiable spatial characteristics. Proliferating cells in the SGZ tended to be located separately from other proliferating cells. About two thirds of them have no closely located other proliferating cells, and the remaining third is located in small clusters comprised of 2 or 3 proliferating cells. Based on our measurements, we calculated that from the age of 30 days to the age of 2.5 years 1.5 million neural precursors are produced in the SGZ.
Collapse
Affiliation(s)
- Karen Smith
- New England Geriatric Research Education and Clinical Center, Bedford Division, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Mikhail V Semënov
- New England Geriatric Research Education and Clinical Center, Bedford Division, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States.,The Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Meng F, Li J, Yang X, Yuan X, Tang X. Role of Smad3 signaling in the epithelial‑mesenchymal transition of the lens epithelium following injury. Int J Mol Med 2018; 42:851-860. [PMID: 29750298 PMCID: PMC6034923 DOI: 10.3892/ijmm.2018.3662] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/04/2018] [Indexed: 11/26/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is important in the development of posterior capsule opacification (PCO), and inhibition of the TGF-β pathway may represent a novel method of treating PCO. Drosophila protein, mothers against decapentaplegic homolog 3 (Smad3) is a phosphorylated receptor-activated Smad required for the transmission of TGF-β signals. Smad3 knockout (KO) disturbs the activation of TGF-β signaling, thus inhibiting the onset of PCO. In the present study, lens epithelial cell (LEC) damage induced by extracapsular cataract extraction was simulated by puncture of the anterior capsule using a 26-gauge hypodermic needle. The effect of Smad3 in the trauma-induced epithelial-mesenchymal transition (EMT) of the lens epithelium in Smad3-KO and wild-type (WT) mice was then observed. The expression levels of EMT markers and extracellular matrix components were measured in the two groups by reverse transcription-quantitative polymerase chain reaction analysis, western blot analysis and immunofluorescence staining. Apoptosis was also detected in the punctured anterior capsule. The Smad3-KO mice exhibited lower expression levels of α-smooth muscle actin, lumican, osteopontin, fibronectin and collagen, compared with the WT mice. Additionally, the Smad3-KO mice exhibited a higher percentage of apoptotic cells than the WT mice. Smad3 signaling was associated with the induction of trauma-induced EMT, and Smad3 KO interfered with TGF-β signaling pathway activation, but did not completely inhibit the trauma-induced EMT in LECs. Therefore, Smad3 may be a target in the treatment of PCO and other fibrosis-related diseases.
Collapse
Affiliation(s)
- Fanlan Meng
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, P.R. China
| | - Jun Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, P.R. China
| | - Xiao Yang
- The State Key Laboratory of Proteomics, Genetics Laboratory of Development and Disease, Institute of Biotechnology, AMMS, Beijing 100071, P.R. China
| | - Xiaoyong Yuan
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, P.R. China
| | - Xin Tang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300020, P.R. China
| |
Collapse
|
13
|
Hsu WL, Ma YL, Liu YC, Lee EHY. Smad4 SUMOylation is essential for memory formation through upregulation of the skeletal myopathy gene TPM2. BMC Biol 2017; 15:112. [PMID: 29183317 PMCID: PMC5706330 DOI: 10.1186/s12915-017-0452-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 11/07/2017] [Indexed: 11/22/2022] Open
Abstract
Background Smad4 is a critical effector of TGF-β signaling that regulates a variety of cellular functions. However, its role in the brain has rarely been studied. Here, we examined the molecular mechanisms underlying the post-translational regulation of Smad4 function by SUMOylation, and its role in spatial memory formation. Results In the hippocampus, Smad4 is SUMOylated by the E3 ligase PIAS1 at Lys-113 and Lys-159. Both spatial training and NMDA injection enhanced Smad4 SUMOylation. Inhibition of Smad4 SUMOylation impaired spatial learning and memory in rats by downregulating TPM2, a gene associated with skeletal myopathies. Similarly, knockdown of TPM2 expression impaired spatial learning and memory, while TPM2 mRNA and protein expression increased after spatial training. Among the TPM2 mutations associated with skeletal myopathies, the TPM2E122K mutation was found to reduce TPM2 expression and impair spatial learning and memory in rats. Conclusions We have identified a novel role of Smad4 SUMOylation and TPM2 in learning and memory formation. These results suggest that patients with skeletal myopathies who carry the TPM2E122K mutation may also have deficits in learning and memory functions. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0452-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei L Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yun L Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yen C Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan. .,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|
14
|
Jafarzadeh M, Soltani BM. Hsa-miR-590-5p Interaction with SMAD3 Transcript Supports Its Regulatory Effect on The TGFβ Signaling Pathway. CELL JOURNAL 2016; 18:7-12. [PMID: 27054113 PMCID: PMC4819388 DOI: 10.22074/cellj.2016.3981] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE SMAD proteins are the core players of the transforming growth factor-beta (TGFβ) signaling pathway, a pathway which is involved in cell proliferation, differentiation and migration. On the other hand, hsa-miRNA-590-5p (miR-590-5p) is known to have a negative regulatory effect on TGFβ signaling pathway receptors. Since, RNAhybrid analy- sis suggested SMAD3 as a bona fide target gene for miR-590, we intended to investigate the effect of miR-590-5p on SMAD3 transcription. MATERIALS AND METHODS In this experimental study, miR-590-5p was overexpressed in different cell lines and its increased expression was detected through quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Western blot analysis was then used to investigate the effect of miR-590-5p overexpression on SMAD3 protein level. Next, the direct interaction of miR-590-5p with the 3´-UTR sequence of SMAD3 transcript was investigated using the dual luciferase assay. Finally, flow cytometery was used to inves- tigate the effect of miR-590-5p overexpression on cell cycle progression in HeLa and SW480 cell lines. RESULTS miR-590-5p was overexpressed in the SW480 cell line and its overexpression resulted in significant reduction of the SMAD3 protein level. Consistently, direct interaction of miR-590-5p with 3´-UTR sequence of SMAD3 was detected. Finally, miR-590-5p over- expression did not show a significant effect on cell cycle progression of Hela and SW480 cell lines. CONCLUSION Consistent with previous reports about the negative regulatory effect of miR-590 on TGFβ receptors, our data suggest that miR-590-5p also attenuates the TGFβ signaling pathway through down-regulation of SMAD3.
Collapse
Affiliation(s)
- Meisam Jafarzadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M Soltani
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Transcriptional Profiling of Newly Generated Dentate Granule Cells Using TU Tagging Reveals Pattern Shifts in Gene Expression during Circuit Integration. eNeuro 2016; 3:eN-NWR-0024-16. [PMID: 27011954 PMCID: PMC4797955 DOI: 10.1523/eneuro.0024-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 11/21/2022] Open
Abstract
Despite representing only a small fraction of hippocampal granule cells, adult-generated newborn granule cells have been implicated in learning and memory (Aimone et al., 2011). Newborn granule cells undergo functional maturation and circuit integration over a period of weeks. However, it is difficult to assess the accompanying gene expression profiles in vivo with high spatial and temporal resolution using traditional methods. Here we used a novel method ["thiouracil (TU) tagging"] to map the profiles of nascent mRNAs in mouse immature newborn granule cells compared with mature granule cells. We targeted a nonmammalian uracil salvage enzyme, uracil phosphoribosyltransferase, to newborn neurons and mature granule cells using retroviral and lentiviral constructs, respectively. Subsequent injection of 4-TU tagged nascent RNAs for analysis by RNA sequencing. Several hundred genes were significantly enhanced in the retroviral dataset compared with the lentiviral dataset. We compared a selection of the enriched genes with steady-state levels of mRNAs using quantitative PCR. Ontology analysis revealed distinct patterns of nascent mRNA expression, with newly generated immature neurons showing enhanced expression for genes involved in synaptic function, and neural differentiation and development, as well as genes not previously associated with granule cell maturation. Surprisingly, the nascent mRNAs enriched in mature cells were related to energy homeostasis and metabolism, presumably indicative of the increased energy demands of synaptic transmission and their complex dendritic architecture. The high spatial and temporal resolution of our modified TU-tagging method provides a foundation for comparison with steady-state RNA analyses by traditional transcriptomic approaches in defining the functional roles of newborn neurons.
Collapse
|
16
|
Liu L, Liu X, Ren X, Tian Y, Chen Z, Xu X, Du Y, Jiang C, Fang Y, Liu Z, Fan B, Zhang Q, Jin G, Yang X, Zhang X. Smad2 and Smad3 have differential sensitivity in relaying TGFβ signaling and inversely regulate early lineage specification. Sci Rep 2016; 6:21602. [PMID: 26905010 PMCID: PMC4764856 DOI: 10.1038/srep21602] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/27/2016] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor beta (TGFβ) related signaling is one of the most important signaling pathways regulating early developmental events. Smad2 and Smad3 are structurally similar and it is mostly considered that they are equally important in mediating TGFβ signals. Here, we show that Smad3 is an insensitive TGFβ transducer as compared with Smad2. Smad3 preferentially localizes within the nucleus and is thus sequestered from membrane signaling. The ability of Smad3 in oligomerization with Smad4 upon agonist stimulation is also impaired given its unique linker region. Smad2 mediated TGFβ signaling plays a crucial role in epiblast development and patterning of three germ layers. However, signaling unrelated nuclear localized Smad3 is dispensable for TGFβ signaling-mediated epiblast specification, but important for early neural development, an event blocked by TGFβ/Smad2 signaling. Both Smad2 and Smad3 bind to the conserved Smads binding element (SBE), but they show nonoverlapped target gene binding specificity and differential transcriptional activity. We conclude that Smad2 and Smad3 possess differential sensitivities in relaying TGFβ signaling and have distinct roles in regulating early developmental events.
Collapse
Affiliation(s)
- Ling Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China.,Tongji University Advanced Institute of Translational Medicine, Shanghai 200092, China
| | - Xu Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Xudong Ren
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yue Tian
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhenyu Chen
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Xiangjie Xu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yanhua Du
- The School of Life Sciences and Technology, Tongji University, Shanghai 200092
| | - Cizhong Jiang
- The School of Life Sciences and Technology, Tongji University, Shanghai 200092
| | - Yujiang Fang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhongliang Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Beibei Fan
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Quanbin Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Guohua Jin
- Department of Anatomy and Neurobiology, the Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Jiangsu 226001, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing 100071, China
| | - Xiaoqing Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China.,Tongji University Advanced Institute of Translational Medicine, Shanghai 200092, China.,The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| |
Collapse
|
17
|
Muñoz MD, Antolín-Vallespín M, Tapia-González S, Sánchez-Capelo A. Smad3 deficiency inhibits dentate gyrus LTP by enhancing GABAA neurotransmission. J Neurochem 2016; 137:190-9. [PMID: 26826552 DOI: 10.1111/jnc.13558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/07/2016] [Accepted: 01/20/2016] [Indexed: 11/29/2022]
Abstract
Transforming growth factor-β signaling through intracellular Smad3 has been implicated in Parkinson's disease (PD) and it fulfills an important role in the neurogenesis and synaptic plasticity that occurs in the adult dentate gyrus (DG). The long-term potentiation (LTP) induced in the DG by high-frequency stimulation of the medial perforant pathway is abolished in the DG of Smad3-deficient mice, but not in the CA1 hippocampal region. Here, we show that NMDA- and AMPA-type glutamate receptors do not participate in the inhibition of LTP associated with Smad3 deficiency. Moreover, there is no difference in the hippocampal GAD65 and GAD67 content, suggesting that GABA biosynthesis remains unaffected. Increased conductance and higher action potential firing thresholds were evident in intracellular recordings of granule cells from Smad3 deficient mice. Interestingly, phasic and tonic GABAA receptor (GABAA R)-mediated neurotransmission is enhanced in the DG of Smad3-deficient mice, and LTP induction can be rescued by inhibiting GABAA R with picrotoxin. Hence, Smad3 signaling in the DG appears to be necessary to induce LTP by regulating GABAA neurotransmission, suggesting a central role of this intracellular signaling pathway in the hippocampal brain plasticity related to learning and memory. Smad3 deficient mice represent a new and interesting model of Parkinson's disease, displaying hippocampal dysfunctions that include decreased neurogenesis and the failure to induce LTP in the dentate gyrus. Here we show that Smad3 deficiency inhibits LTP induction by enhancing phasic and tonic GABAA receptor-mediated neurotransmission, while LTP induction can be rescued with a GABAA receptor antagonist. Alteration of GABA neurotransmission is thought to produce hippocampal cognitive dysfunction in Down's syndrome or Alzheimer's disease, and here we provide new insights into the hippocampal changes in an animal model of Parkinson's disease.
Collapse
Affiliation(s)
- M Dolores Muñoz
- Unidad de Neurología Experimental, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Mónica Antolín-Vallespín
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Silvia Tapia-González
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| | - Amelia Sánchez-Capelo
- CIBERNED - Ser. Neurobiología - Investigación, Hospital Universitario Ramón y Cajal - IRYCIS, Madrid, Spain
| |
Collapse
|
18
|
Ataxin-1 regulates proliferation of hippocampal neural precursors. Neuroscience 2016; 322:54-65. [PMID: 26876606 DOI: 10.1016/j.neuroscience.2016.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/11/2016] [Accepted: 02/03/2016] [Indexed: 02/01/2023]
Abstract
Polyglutamine expansion in the protein ATAXIN-1 (ATXN1) causes spinocerebellar ataxia type 1 (SCA1), an inherited neurodegenerative disease characterized by motor deficits, cognitive impairment and depression. Although ubiquitously expressed, mutant ATXN1 causes neurodegeneration primarily in the cerebellum, which is responsible for the observed motor deficits. The role of ATXN1 outside of the cerebellum and the causes of cognitive deficits and depression in SCA1 are less understood. In this study, we demonstrate a novel role of ATXN1 in the hippocampus as a regulator of adult neurogenesis. Adult hippocampal neurogenesis is the process of generating new hippocampal neurons and is linked to cognition and mood. We found that loss of ATXN1 causes a decrease in hippocampal neurogenesis in ATXN1 null (Atxn1(-/-)) mice. This decrease was caused by reduced proliferation of neural precursors in the hippocampus of Atxn1(-/-) mice, and persisted even when Atxn1(-/-) hippocampal neural precursors were removed from their natural environment and grown in vitro, suggesting that ATXN1 affects proliferation in a cell-autonomous manner. Moreover, expression of ATXN1 with a pathological polyglutamine (polyQ) expansion in wild-type neural precursor cells inhibited their proliferation. Our data establish a novel role for ATXN1 in the hippocampus as an intrinsic regulator of precursor cell proliferation, and suggest a mechanism by which polyQ expansion and loss of ATXN1 affect hippocampal function, potentially contributing to cognitive deficits and depression. These results indicate that while depletion of ATXN1 is a promising therapeutic approach to treat the cerebellar aspects of SCA1, this approach should be employed with caution given the potential for side effects on hippocampal function with loss of wild-type ATXN1.
Collapse
|
19
|
Hohman TJ, Bush WS, Jiang L, Brown-Gentry KD, Torstenson ES, Dudek SM, Mukherjee S, Naj A, Kunkle BW, Ritchie MD, Martin ER, Schellenberg GD, Mayeux R, Farrer LA, Pericak-Vance MA, Haines JL, Thornton-Wells TA. Discovery of gene-gene interactions across multiple independent data sets of late onset Alzheimer disease from the Alzheimer Disease Genetics Consortium. Neurobiol Aging 2016; 38:141-150. [PMID: 26827652 PMCID: PMC4735733 DOI: 10.1016/j.neurobiolaging.2015.10.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022]
Abstract
Late-onset Alzheimer disease (AD) has a complex genetic etiology, involving locus heterogeneity, polygenic inheritance, and gene-gene interactions; however, the investigation of interactions in recent genome-wide association studies has been limited. We used a biological knowledge-driven approach to evaluate gene-gene interactions for consistency across 13 data sets from the Alzheimer Disease Genetics Consortium. Fifteen single nucleotide polymorphism (SNP)-SNP pairs within 3 gene-gene combinations were identified: SIRT1 × ABCB1, PSAP × PEBP4, and GRIN2B × ADRA1A. In addition, we extend a previously identified interaction from an endophenotype analysis between RYR3 × CACNA1C. Finally, post hoc gene expression analyses of the implicated SNPs further implicate SIRT1 and ABCB1, and implicate CDH23 which was most recently identified as an AD risk locus in an epigenetic analysis of AD. The observed interactions in this article highlight ways in which genotypic variation related to disease may depend on the genetic context in which it occurs. Further, our results highlight the utility of evaluating genetic interactions to explain additional variance in AD risk and identify novel molecular mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William S Bush
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Lan Jiang
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Eric S Torstenson
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Scott M Dudek
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Adam Naj
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian W Kunkle
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Marylyn D Ritchie
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Eden R Martin
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Richard Mayeux
- Gertrude H. Sergievsky Center, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University, Boston, MA, USA; Department of Neurology, Boston University, Boston, MA, USA; Department of Ophthalmology, Boston University, Boston, MA, USA; Department of Epidemiology, Boston University, Boston, MA, USA; Department of Biostatistics, Boston University, Boston, MA, USA
| | - Margaret A Pericak-Vance
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Tricia A Thornton-Wells
- Vanderbilt Genetics Institute, Department of Molecular Physiology & Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
von Bernhardi R, Cornejo F, Parada GE, Eugenín J. Role of TGFβ signaling in the pathogenesis of Alzheimer's disease. Front Cell Neurosci 2015; 9:426. [PMID: 26578886 PMCID: PMC4623426 DOI: 10.3389/fncel.2015.00426] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/09/2015] [Indexed: 12/19/2022] Open
Abstract
Aging is the main risk factor for Alzheimer’s disease (AD); being associated with conspicuous changes on microglia activation. Aged microglia exhibit an increased expression of cytokines, exacerbated reactivity to various stimuli, oxidative stress, and reduced phagocytosis of β-amyloid (Aβ). Whereas normal inflammation is protective, it becomes dysregulated in the presence of a persistent stimulus, or in the context of an inflammatory environment, as observed in aging. Thus, neuroinflammation can be a self-perpetuating deleterious response, becoming a source of additional injury to host cells in neurodegenerative diseases. In aged individuals, although transforming growth factor β (TGFβ) is upregulated, its canonical Smad3 signaling is greatly reduced and neuroinflammation persists. This age-related Smad3 impairment reduces protective activation while facilitating cytotoxic activation of microglia through several cellular mechanisms, potentiating microglia-mediated neurodegeneration. Here, we critically discuss the role of TGFβ-Smad signaling on the cytotoxic activation of microglia and its relevance in the pathogenesis of AD. Other protective functions, such as phagocytosis, although observed in aged animals, are not further induced by inflammatory stimuli and TGFβ1. Analysis in silico revealed that increased expression of receptor scavenger receptor (SR)-A, involved in Aβ uptake and cell activation, by microglia exposed to TGFβ, through a Smad3-dependent mechanism could be mediated by transcriptional co-factors Smad2/3 over the MSR1 gene. We discuss that changes of TGFβ-mediated regulation could at least partially mediate age-associated microglia changes, and, together with other changes on inflammatory response, could result in the reduction of protective activation and the potentiation of cytotoxicity of microglia, resulting in the promotion of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Francisca Cornejo
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Guillermo E Parada
- Laboratory of Neuroscience, Faculty of Medicine, Department of Neurology, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Jaime Eugenín
- Laboratory of Neural Systems, Faculty of Chemistry and Biology, Department of Biology, Universidad de Santiago de Chile Santiago, Chile
| |
Collapse
|
21
|
Giráldez-Pérez RM, Antolín-Vallespín M, Muñoz MD, Sánchez-Capelo A. Models of α-synuclein aggregation in Parkinson's disease. Acta Neuropathol Commun 2014; 2:176. [PMID: 25497491 PMCID: PMC4272812 DOI: 10.1186/s40478-014-0176-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is not only characterized by motor disturbances but also, by cognitive, sensory, psychiatric and autonomic dysfunction. It has been proposed that some of these symptoms might be related to the widespread pathology of α-synuclein (α-syn) aggregation in different nuclei of the central and peripheral nervous system. However, the pathogenic formation of α-syn aggregates in different brain areas of PD patients is poorly understood. Most experimental models of PD are valuable to assess specific aspects of its pathogenesis, such as toxin-induced dopaminergic neurodegeneration. However, new models are required that reflect the widespread and progressive formation of α-syn aggregates in different brain areas. Such α-syn aggregation is induced in only a few animal models, for example perikaryon inclusions are found in rats administered rotenone, aggregates with a neuritic morphology develop in mice overexpressing either mutated or wild-type α-syn, and in Smad3 deficient mice, aggregates form extensively in the perikaryon and neurites of specific brain nuclei. In this review we focus on α-syn aggregation in the human disorder, its genetics and the availability of experimental models. Indeed, evidences show that dopamine (DA) metabolism may be related to α-syn and its conformational plasticity, suggesting an interesting link between the two pathological hallmarks of PD: dopaminergic neurodegeneration and Lewy body (LB) formation.
Collapse
Affiliation(s)
- Rosa María Giráldez-Pérez
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
- />Departamento Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Mónica Antolín-Vallespín
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| | - María Dolores Muñoz
- />Unidad de Neurología Experimental, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| | - Amelia Sánchez-Capelo
- />CIBERNED - Ser. Neurobiología – Investigación, Hospital Universitario Ramón y Cajal – IRYCIS, Ctra. Colmenar Viejo Km 9, 28034 Madrid, Spain
| |
Collapse
|
22
|
Morales-Garcia JA, Aguilar-Morante D, Hernandez-Encinas E, Alonso-Gil S, Gil C, Martinez A, Santos A, Perez-Castillo A. Silencing phosphodiesterase 7B gene by lentiviral-shRNA interference attenuates neurodegeneration and motor deficits in hemiparkinsonian mice. Neurobiol Aging 2014; 36:1160-73. [PMID: 25457552 DOI: 10.1016/j.neurobiolaging.2014.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 09/16/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
Abstract
Different studies have suggested that the nucleotide cyclic adenosine 3', 5'-monophosphate can actively play an important role as a neuroprotective and anti-inflammatory agent after a brain injury. The phosphodiesterase 7 (PDE7) enzyme is one of the enzymes responsible for controlling specifically the intracellular levels of cyclic adenosine 3', 5'-monophosphate in the immune and central nervous systems. Therefore, this enzyme could play an important role in brain inflammation and neurodegeneration. In this regard, using different chemical inhibitors of PDE7 we have demonstrated their neuroprotective and anti-inflammatory activity in different models of neurodegenerative disorders, including Parkinson's disease (PD). In the present study, we have used the toxin 6-hydroxydopamine and lipopolysaccharide to model PD and explore the protective effects of PDE7B deficiency in dopaminergic neurons cell death. Lentivirus-mediated PDE7B deprivation conferred marked in vitro and in vivo neuroprotection against 6-hydroxydopamine and lipopolysaccharide toxicity in dopaminergic neurons and preserved motor function involving the dopamine system in mouse. Our results substantiate previous data and provide a validation of PDE7B enzyme as a valuable new target for therapeutic development in the treatment of PD.
Collapse
Affiliation(s)
- Jose A Morales-Garcia
- Instituto de Investigaciones Biomédicas, (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Diana Aguilar-Morante
- Instituto de Investigaciones Biomédicas, (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Elena Hernandez-Encinas
- Instituto de Investigaciones Biomédicas, (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sandra Alonso-Gil
- Instituto de Investigaciones Biomédicas, (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biologicas, Biología FisicoQuimica, CSIC, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biologicas, Biología FisicoQuimica, CSIC, Madrid, Spain
| | - Angel Santos
- Departamento de Bioquímica y Biologia Molecular, Facultad de Medicina, UCM, Madrid, Spain
| | - Ana Perez-Castillo
- Instituto de Investigaciones Biomédicas, (CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|