1
|
Conze C, Trushina NI, Monteiro-Abreu N, Singh L, Romero DV, Wienbeuker E, Schwarze AS, Holtmannspötter M, Bakota L, Brandt R. Redox signaling modulates axonal microtubule organization and induces a specific phosphorylation signature of microtubule-regulating proteins. Redox Biol 2025; 83:103626. [PMID: 40222271 PMCID: PMC12019850 DOI: 10.1016/j.redox.2025.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
Many life processes are regulated by physiological redox signaling, but excessive oxidative stress can damage biomolecules and contribute to disease. Neuronal microtubules are critically involved in axon homeostasis, regulation of axonal transport, and neurodegenerative processes. However, whether and how physiological redox signaling affects axonal microtubules is largely unknown. Using live cell imaging and super-resolution microscopy, we show that subtoxic concentrations of the central redox metabolite hydrogen peroxide increase axonal microtubule dynamics, alter the structure of the axonal microtubule array, and affect the efficiency of axonal transport. We report that the mitochondria-targeting antioxidant SkQ1 and the microtubule stabilizer EpoD abolish the increase in microtubule dynamics. We found that hydrogen peroxide specifically modulates the phosphorylation state of microtubule-regulating proteins, which differs from arsenite as an alternative stress inducer, and induces a largely non-overlapping phosphorylation pattern of MAP1B as a main target. Cell-wide phosphoproteome analysis revealed signaling pathways that are inversely activated by hydrogen peroxide and arsenite. In particular, hydrogen peroxide treatment was associated with kinases that suppress apoptosis and regulate brain metabolism (PRKDC, CK2, PDKs), suggesting that these pathways play a central role in physiological redox signaling and modulation of axonal microtubule organization. The results suggest that the redox metabolite and second messenger hydrogen peroxide induces rapid and local reorganization of the microtubule array in response to mitochondrial activity or as a messenger from neighboring cells by activating specific signaling cascades.
Collapse
Affiliation(s)
- Christian Conze
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Nataliya I Trushina
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Nanci Monteiro-Abreu
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Lisha Singh
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Daniel Villar Romero
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Eike Wienbeuker
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Anna-Sophie Schwarze
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | | | - Lidia Bakota
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany
| | - Roland Brandt
- Department of Neurobiology, School of Biology/Chemistry, Osnabrück University, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Germany; Institute of Cognitive Science, Osnabrück University, Germany.
| |
Collapse
|
2
|
Scaccini S, Cesaroni CA, Caraffi SG, Rizzi S, Rosato S, Peluso F, Spagnoli C, Cavalli A, Brugnoli C, Scandolo G, Pantani A, Ivanovski I, Garavelli L, Frattini D, Fusco C. Axonal motor polyneuropathy in a 13 years old Girl with a de Novo variant in ADNP. Neurogenetics 2025; 26:32. [PMID: 39992398 DOI: 10.1007/s10048-025-00815-w] [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: 10/31/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
ADNP-Related Disorder [previously known as Helsmoortel-Van der Aa syndrome (HVDAS)] is a rare genetic condition resulting from mutations in the activity-dependent neuroprotector homeobox (ADNP) gene. The ADNP protein has multiple functions, including serving as an essential transcription factor for brain development. In addition, pathogenic variants in ADNP have been recognized as one of the most frequent monogenic causes of autism spectrum disorder (ASD) and intellectual disability. Clinical features include craniofacial dysmorphisms, congenital heart defects, gastrointestinal problems such as feeding difficulties, gastroesophageal reflux and frequent vomiting, vision problems, recurrent infections and seizures. Here we describe the novel case of a girl who came to our attention in infancy because of poor and stereotyped motor repertoire, repetitive purposeless movements, and intellectual disability. Whole exome sequencing revealed a de novo heterozygous variant in the ADNP gene, leading to the diagnosis of HVDAS at age 5 years. At the age of 12, nerve conduction velocity testing showed severe four-limb axonal motor polyneuropathy. In this article, we would like to focus on the presence of peripheral nervous system involvement associated with the pathogenic ADNP de novo variant, which may contribute to the clinical characterization of ADNP-Related Disorder.
Collapse
Affiliation(s)
- Sara Scaccini
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Carlo Alberto Cesaroni
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy.
- Laboratorio di Neurofisiologia Pediatrica, Dipartimento Materno-Infantile, Azienda USL-IRCCS Di Reggio Emilia, 42123, Reggio Emilia, Italy.
| | | | - Susanna Rizzi
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Simonetta Rosato
- Clinical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Francesca Peluso
- Clinical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Carlotta Spagnoli
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Anna Cavalli
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Chiara Brugnoli
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Giulia Scandolo
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Agnese Pantani
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Ivan Ivanovski
- Clinical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Livia Garavelli
- Clinical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Daniele Frattini
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
| | - Carlo Fusco
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, 42123, Italy
- Laboratorio di Neurofisiologia Pediatrica, Dipartimento Materno-Infantile, Azienda USL-IRCCS Di Reggio Emilia, 42123, Reggio Emilia, Italy
| |
Collapse
|
3
|
Alsina FC, Lupan BM, Lin LJ, Musso CM, Mosti F, Newman CR, Wood LM, Suzuki A, Agostino M, Moore JK, Silver DL. The RNA-binding protein EIF4A3 promotes axon development by direct control of the cytoskeleton. Cell Rep 2024; 43:114666. [PMID: 39182224 PMCID: PMC11488691 DOI: 10.1016/j.celrep.2024.114666] [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: 03/30/2023] [Revised: 02/28/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
The exon junction complex (EJC), nucleated by EIF4A3, is indispensable for mRNA fate and function throughout eukaryotes. We discover that EIF4A3 directly controls microtubules, independent of RNA, which is critical for neural wiring. While neuronal survival in the developing mouse cerebral cortex depends upon an intact EJC, axonal tract development requires only Eif4a3. Using human cortical organoids, we show that EIF4A3 disease mutations also impair neuronal growth, highlighting conserved functions relevant for neurodevelopmental pathology. Live imaging of growing neurons shows that EIF4A3 is essential for microtubule dynamics. Employing biochemistry and competition experiments, we demonstrate that EIF4A3 directly binds to microtubules, mutually exclusive of the EJC. Finally, in vitro reconstitution assays and rescue experiments demonstrate that EIF4A3 is sufficient to promote microtubule polymerization and that EIF4A3-microtubule association is a major contributor to axon growth. This reveals a fundamental mechanism by which neurons re-utilize core gene expression machinery to directly control the cytoskeleton.
Collapse
Affiliation(s)
- Fernando C Alsina
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Bianca M Lupan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lydia J Lin
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Camila M Musso
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Federica Mosti
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Carly R Newman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lisa M Wood
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark Agostino
- Curtin Health Innovation Research Institute, Curtin Medical School, and Curtin Institute for Computation, Curtin University, Bentley, WA 6102, Australia
| | - Jeffrey K Moore
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences and Duke Regeneration Center, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
4
|
Richardson B, Goedert T, Quraishe S, Deinhardt K, Mudher A. How do neurons age? A focused review on the aging of the microtubular cytoskeleton. Neural Regen Res 2024; 19:1899-1907. [PMID: 38227514 DOI: 10.4103/1673-5374.390974] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024] Open
Abstract
Aging is the leading risk factor for Alzheimer's disease and other neurodegenerative diseases. We now understand that a breakdown in the neuronal cytoskeleton, mainly underpinned by protein modifications leading to the destabilization of microtubules, is central to the pathogenesis of Alzheimer's disease. This is accompanied by morphological defects across the somatodendritic compartment, axon, and synapse. However, knowledge of what occurs to the microtubule cytoskeleton and morphology of the neuron during physiological aging is comparatively poor. Several recent studies have suggested that there is an age-related increase in the phosphorylation of the key microtubule stabilizing protein tau, a modification, which is known to destabilize the cytoskeleton in Alzheimer's disease. This indicates that the cytoskeleton and potentially other neuronal structures reliant on the cytoskeleton become functionally compromised during normal physiological aging. The current literature shows age-related reductions in synaptic spine density and shifts in synaptic spine conformation which might explain age-related synaptic functional deficits. However, knowledge of what occurs to the microtubular and actin cytoskeleton, with increasing age is extremely limited. When considering the somatodendritic compartment, a regression in dendrites and loss of dendritic length and volume is reported whilst a reduction in soma volume/size is often seen. However, research into cytoskeletal change is limited to a handful of studies demonstrating reductions in and mislocalizations of microtubule-associated proteins with just one study directly exploring the integrity of the microtubules. In the axon, an increase in axonal diameter and age-related appearance of swellings is reported but like the dendrites, just one study investigates the microtubules directly with others reporting loss or mislocalization of microtubule-associated proteins. Though these are the general trends reported, there are clear disparities between model organisms and brain regions that are worthy of further investigation. Additionally, longitudinal studies of neuronal/cytoskeletal aging should also investigate whether these age-related changes contribute not just to vulnerability to disease but also to the decline in nervous system function and behavioral output that all organisms experience. This will highlight the utility, if any, of cytoskeletal fortification for the promotion of healthy neuronal aging and potential protection against age-related neurodegenerative disease. This review seeks to summarize what is currently known about the physiological aging of the neuron and microtubular cytoskeleton in the hope of uncovering mechanisms underpinning age-related risk to disease.
Collapse
Affiliation(s)
- Brad Richardson
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Thomas Goedert
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Shmma Quraishe
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Katrin Deinhardt
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Amritpal Mudher
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
5
|
Yang C, Huang YT, Yao YF, Fu JY, Long YS. Hippocampal proteome comparison of infant and adult Fmr1 deficiency mice reveals adult-related changes associated with postsynaptic density. J Proteomics 2024; 303:105202. [PMID: 38797434 DOI: 10.1016/j.jprot.2024.105202] [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: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
Deficiency in fragile X mental retardation 1 (Fmr1) leads to loss of its encoded protein FMRP and causes fragile X syndrome (FXS) by dysregulating its target gene expression in an age-related fashion. Using comparative proteomic analysis, this study identified 105 differentially expressed proteins (DEPs) in the hippocampus of postnatal day 7 (P7) Fmr1-/y mice and 306 DEPs of P90 Fmr1-/y mice. We found that most DEPs in P90 hippocampus were not changed in P7 hippocampus upon FMRP absence, and some P90 DEPs exhibited diverse proteophenotypes with abnormal expression of protein isoform or allele variants. Bioinformatic analyses showed that the P7 DEPs were mainly enriched in fatty acid metabolism and oxidoreductase activity and nutrient responses; whereas the P90 PEPs (especially down-regulated DEPs) were primarily enriched in postsynaptic density (PSD), neuronal projection development and synaptic plasticity. Interestingly, 25 of 30 down-regulated PSD proteins present in the most enriched protein to protein interaction network, and 6 of them (ANK3, ATP2B2, DST, GRIN1, SHANK2 and SYNGAP1) are both FMRP targets and autism candidates. Therefore, this study suggests age-dependent alterations in hippocampal proteomes upon loss of FMRP that may be associated with the pathogenesis of FXS and its related disorders. SIGNIFICANCE: It is well known that loss of FMRP resulted from Fmr1 deficiency leads to fragile X syndrome (FXS), a common neurodevelopmental disorder accompanied by intellectual disability and autism spectrum disorder (ASD). FMRP exhibits distinctly spatiotemporal patterns in the hippocampus between early development and adulthood, which lead to distinct dysregulations of gene expression upon loss of FMRP at the two age stages potentially linked to age-related phenotypes. Therefore, comparison of hippocampal proteomes between infancy and adulthood is valuable to provide insights into the early causations and adult-dependent consequences for FXS and ASD. Using a comparative proteomic analysis, this study identified 105 and 306 differentially expressed proteins (DEPs) in the hippocampi of postnatal day 7 (P7) and P90 Fmr1-/y mice, respectively. Few overlapping DEPs were identified between P7 and P90 stages, and the P7 DEPs were mainly enriched in the regulation of fatty acid metabolism and oxidoreduction, whereas the P90 DEPs were preferentially enriched in the regulation of synaptic formation and plasticity. Particularly, the up-regulated P90 proteins are primarily involved in immune responses and neurodegeneration, and the down-regulated P90 proteins are associated with postsynaptic density, neuron projection and synaptic plasticity. Our findings suggest that distinctly changed proteins in FMRP-absence hippocampus between infancy and adulthood may contribute to age-dependent pathogenesis of FXS and ASD.
Collapse
Affiliation(s)
- Cui Yang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yu-Ting Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yi-Fei Yao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jun-Yi Fu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Yue-Sheng Long
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
6
|
Gagnon J, Caron V, Tremblay A. SUMOylation of nuclear receptor Nor1/NR4A3 coordinates microtubule cytoskeletal dynamics and stability in neuronal cells. Cell Biosci 2024; 14:91. [PMID: 38997783 PMCID: PMC11245793 DOI: 10.1186/s13578-024-01273-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 07/05/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Nor1/NR4A3 is a member of the NR4A subfamily of nuclear receptors that play essential roles in regulating gene expression related to development, cell homeostasis and neurological functions. However, Nor1 is still considered an orphan receptor, as its natural ligand remains unclear for mediating transcriptional activation. Yet other activation signals may modulate Nor1 activity, although their precise role in the development and maintenance of the nervous system remains elusive. METHODS We used transcriptional reporter assays, gene expression profiling, protein turnover measurement, and cell growth assays to assess the functional relevance of Nor1 and SUMO-defective variants in neuronal cells. SUMO1 and SUMO2 conjugation to Nor1 were assessed by immunoprecipitation. Tubulin stability was determined by acetylation and polymerization assays, and live-cell fluorescent microscopy. RESULTS Here, we demonstrate that Nor1 undergoes SUMO1 conjugation at Lys-89 within a canonical ψKxE SUMOylation motif, contributing to the complex pattern of Nor1 SUMOylation, which also includes Lys-137. Disruption of Lys-89, thereby preventing SUMO1 conjugation, led to reduced Nor1 transcriptional competence and protein stability, as well as the downregulation of genes involved in cell growth and metabolism, such as ENO3, EN1, and CFLAR, and in microtubule cytoskeleton dynamics, including MAP2 and MAPT, which resulted in reduced survival of neuronal cells. Interestingly, Lys-89 SUMOylation was potentiated in response to nocodazole, a microtubule depolymerizing drug, although this was insufficient to rescue cells from microtubule disruption despite enhanced Nor1 gene expression. Instead, Lys-89 deSUMOylation reduced the expression of microtubule-severing genes like KATNA1, SPAST, and FIGN, and enhanced α-tubulin cellular levels, acetylation, and microfilament organization, promoting microtubule stability and resistance to nocodazole. These effects contrasted with Lys-137 SUMOylation, suggesting distinct regulatory mechanisms based on specific Nor1 input SUMOylation signals. CONCLUSIONS Our study provides novel insights into Nor1 transcriptional signaling competence and identifies a hierarchical mechanism whereby selective Nor1 SUMOylation may govern neuronal cytoskeleton network dynamics and resistance against microtubule disturbances, a condition strongly associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan Gagnon
- Research Center, CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, Québec, H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, Québec, H3T 1J4, Canada
| | - Véronique Caron
- Research Center, CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, Québec, H3T 1C5, Canada
| | - André Tremblay
- Research Center, CHU Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, Québec, H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montréal, Québec, H3T 1J4, Canada.
- Centre de Recherche en Reproduction et Fertilité, University of Montreal, Saint-Hyacinthe, Québec, J2S 7C6, Canada.
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Montreal, Montréal, Québec, H3T 1J4, Canada.
| |
Collapse
|
7
|
Pandey S, Miller CA. Targeting the cytoskeleton as a therapeutic approach to substance use disorders. Pharmacol Res 2024; 202:107143. [PMID: 38499081 PMCID: PMC11034636 DOI: 10.1016/j.phrs.2024.107143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.
Collapse
Affiliation(s)
- Surya Pandey
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States.
| |
Collapse
|
8
|
Palamarchuk IS, Slavich GM, Vaillancourt T, Rajji TK. Stress-related cellular pathophysiology as a crosstalk risk factor for neurocognitive and psychiatric disorders. BMC Neurosci 2023; 24:65. [PMID: 38087196 PMCID: PMC10714507 DOI: 10.1186/s12868-023-00831-2] [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: 06/13/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
In this narrative review, we examine biological processes linking psychological stress and cognition, with a focus on how psychological stress can activate multiple neurobiological mechanisms that drive cognitive decline and behavioral change. First, we describe the general neurobiology of the stress response to define neurocognitive stress reactivity. Second, we review aspects of epigenetic regulation, synaptic transmission, sex hormones, photoperiodic plasticity, and psychoneuroimmunological processes that can contribute to cognitive decline and neuropsychiatric conditions. Third, we explain mechanistic processes linking the stress response and neuropathology. Fourth, we discuss molecular nuances such as an interplay between kinases and proteins, as well as differential role of sex hormones, that can increase vulnerability to cognitive and emotional dysregulation following stress. Finally, we explicate several testable hypotheses for stress, neurocognitive, and neuropsychiatric research. Together, this work highlights how stress processes alter neurophysiology on multiple levels to increase individuals' risk for neurocognitive and psychiatric disorders, and points toward novel therapeutic targets for mitigating these effects. The resulting models can thus advance dementia and mental health research, and translational neuroscience, with an eye toward clinical application in cognitive and behavioral neurology, and psychiatry.
Collapse
Affiliation(s)
- Iryna S Palamarchuk
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Sunnybrook Health Sciences Centre, Division of Neurology, Toronto, ON, Canada.
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tracy Vaillancourt
- Counselling Psychology, Faculty of Education, University of Ottawa, Ottawa, ON, Canada
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, 1001 Queen Street West, Toronto, ON, M6J1H4, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Pfeifer LS, Schmitz J, Schwalvenberg M, Güntürkün O, Ocklenburg S. A deep phenotyping approach to assess the association of handedness, early life factors and mental health. Sci Rep 2023; 13:15348. [PMID: 37714904 PMCID: PMC10504248 DOI: 10.1038/s41598-023-42563-7] [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: 06/14/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
The development of handedness and other form of functional asymmetries is not yet understood in its critical determinants. Early life factors (e.g., birth weight, birth order) have been discussed to contribute to individual manifestations of functional asymmetries. However, large-scale data such as the UK Biobank suggest that the variance in handedness that is explained by early life factors is minimal. Additionally, atypical handedness has been linked to clinical outcomes such as neurodevelopmental and psychiatric disorders. Against the background of this triad, the current study investigated associations between different forms of functional asymmetries and (a) early life factors as well as (b) clinical outcomes. Functional asymmetries were determined by means of a deep phenotyping approach which notably extends previous work. In our final sample of N = 598 healthy participants, the different variables were tested for associations by means of linear regression models and group comparisons (i.e., ANOVAs and Chi-squared tests). Confirming previous findings from larger cohorts with shallow phenotyping, we found that birth factors do not explain a substantial amount of variance in functional asymmetries. Likewise, functional asymmetries did not seem to have comprehensive predictive power concerning clinical outcomes in our healthy participants. Future studies may further investigate postulated relations in healthy and clinical samples while acknowledging deep phenotyping of laterality.
Collapse
Affiliation(s)
- Lena Sophie Pfeifer
- Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| | - Judith Schmitz
- Biological Personality Psychology, Georg-August-University Goettingen, Göttingen, Germany
| | - Maike Schwalvenberg
- Biopsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Onur Güntürkün
- Biopsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Sebastian Ocklenburg
- Biopsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
- Department of Psychology, Medical School Hamburg, Hamburg, Germany
- Institute for Cognitive and Affective Neuroscience, Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
10
|
Naren P, Samim KS, Tryphena KP, Vora LK, Srivastava S, Singh SB, Khatri DK. Microtubule acetylation dyshomeostasis in Parkinson's disease. Transl Neurodegener 2023; 12:20. [PMID: 37150812 PMCID: PMC10165769 DOI: 10.1186/s40035-023-00354-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
The inter-neuronal communication occurring in extensively branched neuronal cells is achieved primarily through the microtubule (MT)-mediated axonal transport system. This mechanistically regulated system delivers cargos (proteins, mRNAs and organelles such as mitochondria) back and forth from the soma to the synapse. Motor proteins like kinesins and dynein mechanistically regulate polarized anterograde (from the soma to the synapse) and retrograde (from the synapse to the soma) commute of the cargos, respectively. Proficient axonal transport of such cargos is achieved by altering the microtubule stability via post-translational modifications (PTMs) of α- and β-tubulin heterodimers, core components constructing the MTs. Occurring within the lumen of MTs, K40 acetylation of α-tubulin via α-tubulin acetyl transferase and its subsequent deacetylation by HDAC6 and SIRT2 are widely scrutinized PTMs that make the MTs highly flexible, which in turn promotes their lifespan. The movement of various motor proteins, including kinesin-1 (responsible for axonal mitochondrial commute), is enhanced by this PTM, and dyshomeostasis of neuronal MT acetylation has been observed in a variety of neurodegenerative conditions, including Alzheimer's disease and Parkinson's disease (PD). PD is the second most common neurodegenerative condition and is closely associated with impaired MT dynamics and deregulated tubulin acetylation levels. Although the relationship between status of MT acetylation and progression of PD pathogenesis has become a chicken-and-egg question, our review aims to provide insights into the MT-mediated axonal commute of mitochondria and dyshomeostasis of MT acetylation in PD. The enzymatic regulators of MT acetylation along with their synthetic modulators have also been briefly explored. Moving towards a tubulin-based therapy that enhances MT acetylation could serve as a disease-modifying treatment in neurological conditions that lack it.
Collapse
Affiliation(s)
- Padmashri Naren
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Khan Sabiya Samim
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
11
|
Brandt R, Götz J. Special issue on "Cytoskeletal Proteins in Health and Neurodegenerative Disease: Concepts and Methods". Brain Res Bull 2023; 198:50-52. [PMID: 37084983 DOI: 10.1016/j.brainresbull.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
Since 2016, when we compiled a very well-received special issue on "Cytoskeletal Proteins in Health and Neurodegenerative Disease" for Brain Research Bulletin, the field has rapidly evolved, to a large part thanks to the development and maturation of new methods including super-resolution microscopy. Being asked to create a sequel, we therefore decided to keep the main topic, but focus on emerging concepts and novel methods. As before, we compiled nine articles on the role of the neuronal cytoskeleton in both physiological and pathological conditions. Seven of the contributions present current concepts and discuss how cytoskeletal components develop and are maintained throughout a neuron's long lifespan, and also, how they may contribute to physiology and neurodegenerative diseases. Two contributions focus on novel methodological developments and how these techniques can be used to analyze the structure and function of the neuronal cytoskeleton in new ways. The compilation of the articles makes it clear that future approaches must consider the functional relationships between the individual filament systems and the influence different signal transduction mechanisms have on the cytoskeleton and vice versa, in order to adequately explore the causes and consequences of the role of cytoskeletal proteins in health and disease. We hope that this compilation will help in the design of appropriate experiments, aided by new methods, to test critical hypotheses in the field.
Collapse
Affiliation(s)
- Roland Brandt
- Department of Neurobiology Osnabrück University, Barbarastrasse 11, D-49076 Osnabrück, Germany; Center for Cellular Nanosciences Osnabrück University, Barbarastrasse 11, D-49076 Osnabrück, Germany; Institute of Cognitive Sciences, Osnabrück University, Barbarastrasse 11, D-49076 Osnabrück, Germany.
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia.
| |
Collapse
|
12
|
Rierola M, Trushina NI, Holtmannspötter M, Kurre R, Bakota L. Lattice light-sheet microscopy and evaluation of dendritic transport in cultured hippocampal tissue reveal high variability in mobility of the KIF1A motor domain and entry into dendritic spines. Brain Res Bull 2023; 194:13-22. [PMID: 36626968 DOI: 10.1016/j.brainresbull.2022.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/15/2022] [Accepted: 12/27/2022] [Indexed: 01/09/2023]
Abstract
The unique morphology of neurons consists of a long axon and a highly variable arbour of dendritic processes, which assort neuronal cells into the main classes. The dendritic tree serves as the main domain for receiving synaptic input. Therefore, to maintain the structure and to be able to plastically change according to the incoming stimuli, molecules and organelles need to be readily available. This is achieved mainly via bi-directional transport of cargo along the microtubule lattices. Analysis of dendritic transport is lagging behind the investigation of axonal transport. Moreover, addressing transport mechanisms in tissue environment is very challenging and, therefore, rare. We employed high-speed volumetric lattice light-sheet microscopy and single particle tracking of truncated KIF1A motor protein lacking the cargo-binding domain. We focused our analysis on dendritic processes of CA1 pyramidal neurons in cultured hippocampal tissue. Analysis of individual trajectories revealed detailed information about stalling and high variability in movement and speed, and biased directionality of KIF1A. Furthermore, we could also observe KIF1A shortly entering into dendritic spines. We provide a workflow to analyse variations in the speed and direction of motor protein movement in dendrites that are either intrinsic properties of the motor domain or depend on the structure and modification of the microtubule trails.
Collapse
Affiliation(s)
- Marina Rierola
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | | | - Michael Holtmannspötter
- Integrated Bioimaging Facility iBiOs, Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Rainer Kurre
- Integrated Bioimaging Facility iBiOs, Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany.
| |
Collapse
|
13
|
Wang H, Liang L, Yang C, Xiao L, Wang H, Wang G, Zhu Z. The protective role of hippocampal LRP1 knockdown involves synaptic plasticity through the promoting microtubule dynamics and activation of Akt/GSK-3β pathway in depressive rats. J Affect Disord 2023; 322:63-75. [PMID: 36372121 DOI: 10.1016/j.jad.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/14/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND The mechanism by which synaptic plasticity mediates the occurrence of depression is unknown. Low-density lipoprotein receptor-related protein 1 (LRP1) affects axon growth and neurogenesis in the brain, but its role in depressive-like behaviors is poorly understood. METHODS Adeno-associated virus-mediated small interfering RNA was injected into the bilateral hippocampus 14 days before chronic unpredicted mild stress (CUMS). Behavior performance was assessed for depressive-like behaviors. Western blot was conducted to detect levels of LRP1, neurogenesis-related proteins, synaptic markers, microtubule system molecules and Akt/GSK-3β signaling-related proteins. Immunohistochemical staining was performed for LRP1 protein, immunofluorescence staining was conducted to determine the Sox2 protein, Nissl's staining and transmission electron microscope staining were used to observe hippocampal morphological features. RESULTS The expression of hippocampal LRP1 was positively correlated with depressive-like behaviors. Treatment with iAAV-LRP1 exerted protective effects on depressive-like behaviors. LRP1 Knockdown relieved the inhibition of synaptic plasticity induced by CUMS. Expression of Sox2, GluR2 and SYP was significantly increased in iAAV-LRP1 CUMS rats. LRP1 knockdown reduced the p-tau (Ser262 and Thr404) and Acet-tubule levels in depressed rats. Finally, we found that LRP1 knockdown activated the PI3K/Akt pathway and inhibited GSK-3β signal transduction. LIMITATIONS More neurogenesis markers would be considered, and stereotactic injection into hippocampal DG region could be performed to investigate the effects of LRP1. CONCLUSIONS These findings indicated that hippocampal LRP1 deficiency in stressed rats plays an important protective role in depressive-like behavior by increasing synaptic plasticity mediated by microtubule dynamic and activating Akt/GSK-3β signaling pathway. Therefore, LRP1 may represent a potential therapeutic target for depression.
Collapse
Affiliation(s)
- Hui Wang
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Liang Liang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Can Yang
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| | - Zhixian Zhu
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
14
|
Conze C, Trushina NI, Holtmannspötter M, Rierola M, Attanasio S, Bakota L, Piehler J, Brandt R. Super-resolution imaging and quantitative analysis of microtubule arrays in model neurons show that epothilone D increases the density but decreases the length and straightness of microtubules in axon-like processes. Brain Res Bull 2022; 190:234-243. [PMID: 36244582 PMCID: PMC9634454 DOI: 10.1016/j.brainresbull.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/29/2022]
Abstract
Microtubules are essential for the development of neurons and the regulation of their structural plasticity. Microtubules also provide the structural basis for the long-distance transport of cargo. Various factors influence the organization and dynamics of neuronal microtubules, and disturbance of microtubule regulation is thought to play a central role in neurodegenerative diseases. However, imaging and quantitative assessment of the microtubule organization in the densely packed neuronal processes is challenging. The development of super-resolution techniques combined with the use of nanobodies offers new possibilities to visualize microtubules in neurites in high resolution. In combination with recently developed computational analysis tools, this allows automated quantification of neuronal microtubule organization with high precision. Here we have implemented three-dimensional DNA-PAINT (Point Accumulation in Nanoscale Topography), a single-molecule localization microscopy (SMLM) technique, which allows us to acquire 3D arrays of the microtubule lattice in axons of model neurons (neuronally differentiated PC12 cells) and dendrites of primary neurons. For the quantitative analysis of the microtubule organization, we used the open-source software package SMLM image filament extractor (SIFNE). We found that treatment with nanomolar concentrations of the microtubule-targeting drug epothilone D (EpoD) increased microtubule density in axon-like processes of model neurons and shifted the microtubule length distribution to shorter ones, with a mean microtubule length of 2.39 µm (without EpoD) and 1.98 µm (with EpoD). We also observed a significant decrease in microtubule straightness after EpoD treatment. The changes in microtubule density were consistent with live-cell imaging measurements of ensemble microtubule dynamics using a previously established Fluorescence Decay After Photoactivation (FDAP) assay. For comparison, we determined the organization of the microtubule array in dendrites of primary hippocampal neurons. We observed that dendritic microtubules have a very similar length distribution and straightness compared to microtubules in axon-like processes of a neuronal cell line. Our data show that super-resolution imaging of microtubules followed by algorithm-based image analysis represents a powerful tool to quantitatively assess changes in microtubule organization in neuronal processes, useful to determine the effect of microtubule-modulating conditions. We also provide evidence that the approach is robust and can be applied to neuronal cell lines or primary neurons, both after incorporation of labeled tubulin and by anti-tubulin antibody staining.
Collapse
Affiliation(s)
- Christian Conze
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | | | | | - Marina Rierola
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Simone Attanasio
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Jacob Piehler
- Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany; Division of Biophysics, Osnabrück University, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany; Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany.
| |
Collapse
|
15
|
Lipid metabolism and ageing in Caenorhabditis elegans: a complex interplay. Biogerontology 2022; 23:541-557. [PMID: 36048312 DOI: 10.1007/s10522-022-09989-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/25/2022] [Indexed: 11/02/2022]
Abstract
Life expectancy in Western countries is increasing, with concomitant rise in ageing-related pathologies, including Parkinson's and Alzheimer's disease, as well as other neurodegenerative diseases. Consequently, the medical, psychological and economic burden to society is increasing. Thus, understanding the cellular and molecular mechanisms underlying the association of ageing with elevated vulnerability to disease is crucial towards promoting quality of life in old age. Caenorhabditis elegans has emerged as a versatile model to study ageing, due to its simplicity, fast life cycle, and the availability of a wide range of biological tools to target specific genes and cells. Indeed, recent studies in C. elegans have revealed that lipid metabolism plays a key role in controlling longevity by impinging on a plethora of molecular pathways and cell types. Here, we summarise findings relevant to the interplay between lipid metabolism and ageing in C. elegans, and discuss the implications for the pathogenesis of age-related disorders in humans.
Collapse
|
16
|
Synaptic branch stability is mediated by non-enzymatic functions of MEC-17/αTAT1 and ATAT-2. Sci Rep 2022; 12:14003. [PMID: 35977998 PMCID: PMC9385713 DOI: 10.1038/s41598-022-18333-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
Microtubules are fundamental elements of neuronal structure and function. They are dynamic structures formed from protofilament chains of α- and β-tubulin heterodimers. Acetylation of the lysine 40 (K40) residue of α-tubulin protects microtubules from mechanical stresses by imparting structural elasticity. The enzyme responsible for this acetylation event is MEC-17/αTAT1. Despite its functional importance, however, the consequences of altered MEC-17/αTAT1 levels on neuronal structure and function are incompletely defined. Here we demonstrate that overexpression or loss of MEC-17, or of its functional paralogue ATAT-2, causes a delay in synaptic branch extension, and defective synaptogenesis in the mechanosensory neurons of Caenorhabditis elegans. Strikingly, by adulthood, the synaptic branches in these animals are lost, while the main axon shaft remains mostly intact. We show that MEC-17 and ATAT-2 regulate the stability of the synaptic branches largely independently from their acetyltransferase domains. Genetic analyses reveals novel interactions between both mec-17 and atat-2 with the focal adhesion gene zyx-1/Zyxin, which has previously been implicated in actin remodelling. Together, our results reveal new, acetylation-independent roles for MEC-17 and ATAT-2 in the development and maintenance of neuronal architecture.
Collapse
|
17
|
Sun C, Dong Y, Wei J, Cai M, Liang D, Fu Y, Zhou Y, Sui Y, Wu F, Mikhaylov R, Wang H, Fan F, Xie Z, Stringer M, Yang Z, Wu Z, Tian L, Yang X. Acoustically Accelerated Neural Differentiation of Human Embryonic Stem Cells. Acta Biomater 2022; 151:333-345. [DOI: 10.1016/j.actbio.2022.07.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/14/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
18
|
Soliman A, Bakota L, Brandt R. Microtubule-modulating Agents in the Fight Against Neurodegeneration: Will it ever Work? Curr Neuropharmacol 2022; 20:782-798. [PMID: 34852744 PMCID: PMC9878958 DOI: 10.2174/1570159x19666211201101020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022] Open
Abstract
The microtubule skeleton plays an essential role in nerve cells as the most important structural determinant of morphology and as a highway for axonal transport processes. Many neurodegenerative diseases are characterized by changes in the structure and organization of microtubules and microtubule-regulating proteins such as the microtubule-associated protein tau, which exhibits characteristic changes in a whole class of diseases collectively referred to as tauopathies. Changes in the dynamics of microtubules appear to occur early under neurodegenerative conditions and are also likely to contribute to age-related dysfunction of neurons. Thus, modulating microtubule dynamics and correcting impaired microtubule stability can be a useful neuroprotective strategy to counteract the disruption of the microtubule system in disease and aging. In this article, we review current microtubule- directed approaches for the treatment of neurodegenerative diseases with microtubules as a drug target, tau as a drug target, and post-translational modifications as potential modifiers of the microtubule system. We discuss limitations of the approaches that can be traced back to the rather unspecific mechanism of action, which causes undesirable side effects in non-neuronal cell types or which are due to the disruption of non-microtubule-related interactions. We also develop some thoughts on how the specificity of the approaches can be improved and what further targets could be used for modulating substances.
Collapse
Affiliation(s)
- Ahmed Soliman
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, Osnabrück University, Osnabrück, Germany;,Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany;,Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany,Address correspondence to this author at the Department of Neurobiology, Osnabrück University, Osnabrück, Germany; Tel: +49 541 969 2338; E-mail:
| |
Collapse
|
19
|
Peña-Ortega F, Robles-Gómez ÁA, Xolalpa-Cueva L. Microtubules as Regulators of Neural Network Shape and Function: Focus on Excitability, Plasticity and Memory. Cells 2022; 11:cells11060923. [PMID: 35326374 PMCID: PMC8946818 DOI: 10.3390/cells11060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
Neuronal microtubules (MTs) are complex cytoskeletal protein arrays that undergo activity-dependent changes in their structure and function as a response to physiological demands throughout the lifespan of neurons. Many factors shape the allostatic dynamics of MTs and tubulin dimers in the cytosolic microenvironment, such as protein–protein interactions and activity-dependent shifts in these interactions that are responsible for their plastic capabilities. Recently, several findings have reinforced the role of MTs in behavioral and cognitive processes in normal and pathological conditions. In this review, we summarize the bidirectional relationships between MTs dynamics, neuronal processes, and brain and behavioral states. The outcomes of manipulating the dynamicity of MTs by genetic or pharmacological approaches on neuronal morphology, intrinsic and synaptic excitability, the state of the network, and behaviors are heterogeneous. We discuss the critical position of MTs as responders and adaptative elements of basic neuronal function whose impact on brain function is not fully understood, and we highlight the dilemma of artificially modulating MT dynamics for therapeutic purposes.
Collapse
|
20
|
Huang L, Peng Y, Tao X, Ding X, Li R, Jiang Y, Zuo W. Microtubule Organization Is Essential for Maintaining Cellular Morphology and Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1623181. [PMID: 35295719 PMCID: PMC8920689 DOI: 10.1155/2022/1623181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/10/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microtubules (MTs) are highly dynamic polymers essential for a wide range of cellular physiologies, such as acting as directional railways for intracellular transport and position, guiding chromosome segregation during cell division, and controlling cell polarity and morphogenesis. Evidence has established that maintaining microtubule (MT) stability in neurons is vital for fundamental cellular and developmental processes, such as neurodevelopment, degeneration, and regeneration. To fulfill these diverse functions, the nervous system employs an arsenal of microtubule-associated proteins (MAPs) to control MT organization and function. Subsequent studies have identified that the disruption of MT function in neurons is one of the most prevalent and important pathological features of traumatic nerve damage and neurodegenerative diseases and that this disruption manifests as a reduction in MT polymerization and concomitant deregulation of the MT cytoskeleton, as well as downregulation of microtubule-associated protein (MAP) expression. A variety of MT-targeting agents that reverse this pathological condition, which is regarded as a therapeutic opportunity to intervene the onset and development of these nervous system abnormalities, is currently under development. Here, we provide an overview of the MT-intrinsic organization process and how MAPs interact with the MT cytoskeleton to promote MT polymerization, stabilization, and bundling. We also highlight recent advances in MT-targeting therapeutic agents applied to various neurological disorders. Together, these findings increase our current understanding of the function and regulation of MT organization in nerve growth and regeneration.
Collapse
Affiliation(s)
- Lijiang Huang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| | - Yan Peng
- Hangzhou Institute for Food and Drug Control, Hangzhou, Zhejiang, China
| | - Xuetao Tao
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xiaoxiao Ding
- Department of Pharmacy, The People's Hospital of Beilun District, Ningbo, Zhejiang 315807, China
| | - Rui Li
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongsheng Jiang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| | - Wei Zuo
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| |
Collapse
|
21
|
Saunders HAJ, Johnson-Schlitz DM, Jenkins BV, Volkert PJ, Yang SZ, Wildonger J. Acetylated α-tubulin K394 regulates microtubule stability to shape the growth of axon terminals. Curr Biol 2022; 32:614-630.e5. [PMID: 35081332 PMCID: PMC8843987 DOI: 10.1016/j.cub.2021.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/19/2021] [Accepted: 12/07/2021] [Indexed: 02/09/2023]
Abstract
Microtubules are essential to neuron shape and function. Acetylation of tubulin has the potential to directly tune the behavior and function of microtubules in cells. Although proteomic studies have identified several acetylation sites in α-tubulin, the effects of acetylation at these sites remains largely unknown. This includes the highly conserved residue lysine 394 (K394), which is located at the αβ-tubulin dimer interface. Using a fly model, we show that α-tubulin K394 is acetylated in the nervous system and is an essential residue. We found that an acetylation-blocking mutation in endogenous α-tubulin, K394R, perturbs the synaptic morphogenesis of motoneurons and reduces microtubule stability. Intriguingly, the K394R mutation has opposite effects on the growth of two functionally and morphologically distinct motoneurons, revealing neuron-type-specific responses when microtubule stability is altered. Eliminating the deacetylase HDAC6 increases K394 acetylation, and the over-expression of HDAC6 reduces microtubule stability similar to the K394R mutant. Thus, our findings implicate α-tubulin K394 and its acetylation in the regulation of microtubule stability and suggest that HDAC6 regulates K394 acetylation during synaptic morphogenesis.
Collapse
Affiliation(s)
- Harriet A. J. Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Dena M. Johnson-Schlitz
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Brian V. Jenkins
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Peter J. Volkert
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Biochemistry Scholars Program, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Sihui Z. Yang
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Cellular & Molecular Biology Graduate Program, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Current address: Pediatrics Department and Biological Sciences Division, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA,Lead and author for correspondence:
| |
Collapse
|
22
|
Liang C, Carrel D, Singh NK, Hiester LL, Fanget I, Kim H, Firestein BL. Carboxypeptidase E Independently Changes Microtubule Glutamylation, Dendritic Branching, and Neuronal Migration. ASN Neuro 2022; 14:17590914211062765. [PMID: 35014548 PMCID: PMC8755936 DOI: 10.1177/17590914211062765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Neuronal migration and dendritogenesis are dependent on dynamic changes to the microtubule (MT) network. Among various factors that regulate MT dynamics and stability, post-translational modifications (PTMs) of MTs play a critical role in conferring specificity of regulatory protein binding to MTs. Thus, it is important to understand the regulation of PTMs during brain development as multiple developmental processes are dependent on MTs. In this study, we identified that carboxypeptidase E (CPE) changes tubulin polyglutamylation, a major PTM in the brain, and we examine the impact of CPE-mediated changes to polyglutamylation on cortical neuron migration and dendrite morphology. We show, for the first time, that overexpression of CPE increases the level of polyglutamylated α-tubulin while knockdown decreases the level of polyglutamylation. We also demonstrate that CPE-mediated changes to polyglutamylation are dependent on the CPE zinc-binding motif and that this motif is necessary for CPE action on p150Glued localization. However, overexpression of a CPE mutant that does not increase MT glutamylation mimics the effects of overexpression of wild type CPE on dendrite branching. Furthermore, although overexpression of wild type CPE does not alter cortical neuron migration, overexpression of the mutant may act in a dominant-negative manner as it decreases the number of neurons that reach the cortical plate (CP), as we previously reported for CPE knockdown. Overall, our data suggest that CPE changes MT glutamylation and redistribution of p150Glued and that this function of CPE is independent of its role in shaping dendrite development but plays a partial role in regulating cortical neuron migration.
Collapse
Affiliation(s)
- Chen Liang
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA.,Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Damien Carrel
- SPPIN Laboratory, 555089Université de Paris, Centre National de la Recherche Scientifique UMR 8003, Paris, France
| | - Nisha K Singh
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA.,Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Liam L Hiester
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| | - Isabelle Fanget
- SPPIN Laboratory, 555089Université de Paris, Centre National de la Recherche Scientifique UMR 8003, Paris, France
| | - Hyuck Kim
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, 242612The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
23
|
Elsayed LEO, Eltazi IZ, Ahmed AE, Stevanin G. Insights into Clinical, Genetic, and Pathological Aspects of Hereditary Spastic Paraplegias: A Comprehensive Overview. Front Mol Biosci 2021; 8:690899. [PMID: 34901147 PMCID: PMC8662366 DOI: 10.3389/fmolb.2021.690899] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 10/19/2021] [Indexed: 12/31/2022] Open
Abstract
Hereditary spastic paraplegias (HSP) are a heterogeneous group of motor neurodegenerative disorders that have the core clinical presentation of pyramidal syndrome which starts typically in the lower limbs. They can present as pure or complex forms with all classical modes of monogenic inheritance reported. To date, there are more than 100 loci/88 spastic paraplegia genes (SPG) involved in the pathogenesis of HSP. New patterns of inheritance are being increasingly identified in this era of huge advances in genetic and functional studies. A wide range of clinical symptoms and signs are now reported to complicate HSP with increasing overall complexity of the clinical presentations considered as HSP. This is especially true with the emergence of multiple HSP phenotypes that are situated in the borderline zone with other neurogenetic disorders. The genetic diagnostic approaches and the utilized techniques leave a diagnostic gap of 25% in the best studies. In this review, we summarize the known types of HSP with special focus on those in which spasticity is the principal clinical phenotype ("SPGn" designation). We discuss their modes of inheritance, clinical phenotypes, underlying genetics, and molecular pathways, providing some observations about therapeutic opportunities gained from animal models and functional studies. This review may pave the way for more analytic approaches that take into consideration the overall picture of HSP. It will shed light on subtle associations that can explain the occurrence of the disease and allow a better understanding of its observed variations. This should help in the identification of future biomarkers, predictors of disease onset and progression, and treatments for both better functional outcomes and quality of life.
Collapse
Affiliation(s)
- Liena E. O. Elsayed
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University [PNU], Riyadh, Saudi Arabia
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Ammar E. Ahmed
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Giovanni Stevanin
- Institut du Cerveau – Paris Brain Institute - ICM, Sorbonne Université, INSERM, CNRS, APHP, Paris, France
- CNRS, INCIA, Université de Bordeaux, Bordeaux, France
- Ecole Pratique des Hautes Etudes, EPHE, PSL Research University, Paris, France
| |
Collapse
|
24
|
Yin BK, Wang ZQ. Beyond HAT Adaptor: TRRAP Liaisons with Sp1-Mediated Transcription. Int J Mol Sci 2021; 22:12445. [PMID: 34830324 PMCID: PMC8625110 DOI: 10.3390/ijms222212445] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
The members of the phosphatidylinositol 3-kinase-related kinase (PIKK) family play vital roles in multiple biological processes, including DNA damage response, metabolism, cell growth, mRNA decay, and transcription. TRRAP, as the only member lacking the enzymatic activity in this family, is an adaptor protein for several histone acetyltransferase (HAT) complexes and a scaffold protein for multiple transcription factors. TRRAP has been demonstrated to regulate various cellular functions in cell cycle progression, cell stemness maintenance and differentiation, as well as neural homeostasis. TRRAP is known to be an important orchestrator of many molecular machineries in gene transcription by modulating the activity of some key transcription factors, including E2F1, c-Myc, p53, and recently, Sp1. This review summarizes the biological and biochemical studies on the action mode of TRRAP together with the transcription factors, focusing on how TRRAP-HAT mediates the transactivation of Sp1-governing biological processes, including neurodegeneration.
Collapse
Affiliation(s)
- Bo-Kun Yin
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany;
| | - Zhao-Qi Wang
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany;
- Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
25
|
Santiago-Mujika E, Luthi-Carter R, Giorgini F, Kalaria RN, Mukaetova-Ladinska EB. Tubulin and Tubulin Posttranslational Modifications in Alzheimer's Disease and Vascular Dementia. Front Aging Neurosci 2021; 13:730107. [PMID: 34776926 PMCID: PMC8586541 DOI: 10.3389/fnagi.2021.730107] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 01/26/2023] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are the two most common forms of dementia in older people. Although these two dementia types differ in their etiology, they share many pathophysiological and morphological features, including neuronal loss, which is associated with the microtubule (MT) destabilization. Stabilization of MTs is achieved in different ways: through interactions with MT binding proteins (MTBP) or by posttranslational modifications (PTMs) of tubulin. Polyglutamylation and tyrosination are two foremost PTMs that regulate the interaction between MTs and MTBPs, and play, therefore, a role in neurodegeneration. In this review, we summarize key information on tubulin PTMs in relation to AD and VaD and address the importance of studying further the tubulin code to reveal sites of potential intervention in development of novel and effective dementia therapy.
Collapse
Affiliation(s)
- Estibaliz Santiago-Mujika
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Ruth Luthi-Carter
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Elizabeta B. Mukaetova-Ladinska
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
- Evington Centre, Leicester General Hospital, Leicester, United Kingdom
| |
Collapse
|
26
|
Franke K, Bublak P, Hoyer D, Billiet T, Gaser C, Witte OW, Schwab M. In vivo biomarkers of structural and functional brain development and aging in humans. Neurosci Biobehav Rev 2021; 117:142-164. [PMID: 33308708 DOI: 10.1016/j.neubiorev.2017.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/25/2022]
Abstract
Brain aging is a major determinant of aging. Along with the aging population, prevalence of neurodegenerative diseases is increasing, therewith placing economic and social burden on individuals and society. Individual rates of brain aging are shaped by genetics, epigenetics, and prenatal environmental. Biomarkers of biological brain aging are needed to predict individual trajectories of aging and the risk for age-associated neurological impairments for developing early preventive and interventional measures. We review current advances of in vivo biomarkers predicting individual brain age. Telomere length and epigenetic clock, two important biomarkers that are closely related to the mechanistic aging process, have only poor deterministic and predictive accuracy regarding individual brain aging due to their high intra- and interindividual variability. Phenotype-related biomarkers of global cognitive function and brain structure provide a much closer correlation to age at the individual level. During fetal and perinatal life, autonomic activity is a unique functional marker of brain development. The cognitive and structural biomarkers also boast high diagnostic specificity for determining individual risks for neurodegenerative diseases.
Collapse
Affiliation(s)
- K Franke
- Department of Neurology, Jena University Hospital, Jena, Germany.
| | - P Bublak
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - D Hoyer
- Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - C Gaser
- Department of Neurology, Jena University Hospital, Jena, Germany; Department of Psychiatry, Jena University Hospital, Jena, Germany
| | - O W Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - M Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
27
|
Wu Z, Wang G, Zhou L, Sun L, Xie Y, Xiao L. Neuroinflammation decreased hippocampal microtubule dynamics in the acute behavioral deficits induced by intracerebroventricular injection of lipopolysaccharide in male adult rats. Neuroreport 2021; 32:603-611. [PMID: 33850084 DOI: 10.1097/wnr.0000000000001638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroinflammation plays a vital role in the pathology of depression. Microtubule dynamics produces an immediate response to stress, but the effect of microtubule dynamics in the rats with acute behavioral deficits following a central immune challenge remains elusive. Adult male Sprague-Dawley rats were subjected to the intracerebroventricular (icv) injection of lipopolysaccharide (. Behavioral tests, including bodyweight, sucrose preference test (SPT), forced swimming test (FST) and open field test (OFT), were performed to evaluate anxiety-like and depressive-like phenotypes at 24 h after injection, and some neuroinflammation biomarkers and microtubule dynamics in the hippocampus were detected. Lipopolysaccharide decreased the bodyweight, sucrose preference in SPT (depressive-like behavior), spontaneous activity in OFT (anxiety-like behavior) and increased the immobility time in FST (depressive-like behavior). Besides, lipopolysaccharide increased the mRNA levels of hippocampal CD11b and ionized calcium binding adaptor molecule (Iba1), which suggest microglial activation, and also upregulated hippocampal NLR Family Pyrin Domain Containing 3 inflammasome/interleukin-18/nuclear factor kappa-B mRNA. Lipopolysaccharide injection(icv) reduced the ratio of Tyr-/Acet-tubulin, an important marker of microtubule dynamics, in the acute behavioral deficit rats. Specifically, a decrease in Tyr-tubulin and an increase in the expression of Acet-tubulin were observed, indicating weakened microtubule dynamics. Pearson correlation analysis further showed that there was a significant negative correlation between hippocampal microtubule dynamics and neuroinflammatory activity. This study confirmed that hippocampal microtubule dynamics was decreased in the rats with acute behavioral deficits following a central immune challenge.
Collapse
Affiliation(s)
- Zuotian Wu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang, Wuhan, China
| | | | | | | | | | | |
Collapse
|
28
|
Jangampalli Adi P, Reddy PH. Phosphorylated tau targeted small-molecule PROTACs for the treatment of Alzheimer's disease and tauopathies. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166162. [PMID: 33940164 DOI: 10.1016/j.bbadis.2021.166162] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Tau is a microtubule-stabilizing protein that plays an important role in the formation of axonal microtubules in neurons. Phosphorylated tau (p-Tau) has received great attention in the field of Alzheimer's disease (AD) as a potential therapeutic target due to its involvement with synaptic damage and neuronal dysfunction. Mounting evidence suggests that amyloid beta (Aβ)-targeted clinical trials continuously failed; therefore, it is important to consider alternative therapeutic strategies such as p-tau-PROTACs targeted small molecules for AD and other tauopathies. The present article describes the characteristics of tau biology, structure, and function in both healthy and pathological states in AD. It also explains data from studies that have identified the involvement of p-tau in neuronal damage and synaptic and cognitive functions in AD. Current article also covers several aspects, including small molecule inhibitors, and the development of p-tau-PROTACs targeted drug molecules to treat patients with AD and other tauopathies.
Collapse
Affiliation(s)
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
29
|
Cuellar-Partida G, Tung JY, Eriksson N, Albrecht E, Aliev F, Andreassen OA, Barroso I, Beckmann JS, Boks MP, Boomsma DI, Boyd HA, Breteler MMB, Campbell H, Chasman DI, Cherkas LF, Davies G, de Geus EJC, Deary IJ, Deloukas P, Dick DM, Duffy DL, Eriksson JG, Esko T, Feenstra B, Geller F, Gieger C, Giegling I, Gordon SD, Han J, Hansen TF, Hartmann AM, Hayward C, Heikkilä K, Hicks AA, Hirschhorn JN, Hottenga JJ, Huffman JE, Hwang LD, Ikram MA, Kaprio J, Kemp JP, Khaw KT, Klopp N, Konte B, Kutalik Z, Lahti J, Li X, Loos RJF, Luciano M, Magnusson SH, Mangino M, Marques-Vidal P, Martin NG, McArdle WL, McCarthy MI, Medina-Gomez C, Melbye M, Melville SA, Metspalu A, Milani L, Mooser V, Nelis M, Nyholt DR, O'Connell KS, Ophoff RA, Palmer C, Palotie A, Palviainen T, Pare G, Paternoster L, Peltonen L, Penninx BWJH, Polasek O, Pramstaller PP, Prokopenko I, Raikkonen K, Ripatti S, Rivadeneira F, Rudan I, Rujescu D, Smit JH, Smith GD, Smoller JW, Soranzo N, Spector TD, Pourcain BS, Starr JM, Stefánsson H, Steinberg S, Teder-Laving M, Thorleifsson G, Stefánsson K, Timpson NJ, Uitterlinden AG, van Duijn CM, van Rooij FJA, Vink JM, Vollenweider P, Vuoksimaa E, Waeber G, et alCuellar-Partida G, Tung JY, Eriksson N, Albrecht E, Aliev F, Andreassen OA, Barroso I, Beckmann JS, Boks MP, Boomsma DI, Boyd HA, Breteler MMB, Campbell H, Chasman DI, Cherkas LF, Davies G, de Geus EJC, Deary IJ, Deloukas P, Dick DM, Duffy DL, Eriksson JG, Esko T, Feenstra B, Geller F, Gieger C, Giegling I, Gordon SD, Han J, Hansen TF, Hartmann AM, Hayward C, Heikkilä K, Hicks AA, Hirschhorn JN, Hottenga JJ, Huffman JE, Hwang LD, Ikram MA, Kaprio J, Kemp JP, Khaw KT, Klopp N, Konte B, Kutalik Z, Lahti J, Li X, Loos RJF, Luciano M, Magnusson SH, Mangino M, Marques-Vidal P, Martin NG, McArdle WL, McCarthy MI, Medina-Gomez C, Melbye M, Melville SA, Metspalu A, Milani L, Mooser V, Nelis M, Nyholt DR, O'Connell KS, Ophoff RA, Palmer C, Palotie A, Palviainen T, Pare G, Paternoster L, Peltonen L, Penninx BWJH, Polasek O, Pramstaller PP, Prokopenko I, Raikkonen K, Ripatti S, Rivadeneira F, Rudan I, Rujescu D, Smit JH, Smith GD, Smoller JW, Soranzo N, Spector TD, Pourcain BS, Starr JM, Stefánsson H, Steinberg S, Teder-Laving M, Thorleifsson G, Stefánsson K, Timpson NJ, Uitterlinden AG, van Duijn CM, van Rooij FJA, Vink JM, Vollenweider P, Vuoksimaa E, Waeber G, Wareham NJ, Warrington N, Waterworth D, Werge T, Wichmann HE, Widen E, Willemsen G, Wright AF, Wright MJ, Xu M, Zhao JH, Kraft P, Hinds DA, Lindgren CM, Mägi R, Neale BM, Evans DM, Medland SE. Genome-wide association study identifies 48 common genetic variants associated with handedness. Nat Hum Behav 2021; 5:59-70. [PMID: 32989287 PMCID: PMC7116623 DOI: 10.1038/s41562-020-00956-y] [Show More Authors] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023]
Abstract
Handedness has been extensively studied because of its relationship with language and the over-representation of left-handers in some neurodevelopmental disorders. Using data from the UK Biobank, 23andMe and the International Handedness Consortium, we conducted a genome-wide association meta-analysis of handedness (N = 1,766,671). We found 41 loci associated (P < 5 × 10-8) with left-handedness and 7 associated with ambidexterity. Tissue-enrichment analysis implicated the CNS in the aetiology of handedness. Pathways including regulation of microtubules and brain morphology were also highlighted. We found suggestive positive genetic correlations between left-handedness and neuropsychiatric traits, including schizophrenia and bipolar disorder. Furthermore, the genetic correlation between left-handedness and ambidexterity is low (rG = 0.26), which implies that these traits are largely influenced by different genetic mechanisms. Our findings suggest that handedness is highly polygenic and that the genetic variants that predispose to left-handedness may underlie part of the association with some psychiatric disorders.
Collapse
Affiliation(s)
- Gabriel Cuellar-Partida
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
- 23andMe, Inc., Sunnyvale, CA, USA
| | | | | | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Fazil Aliev
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
- Karabuk University, Faculty of Business, Karabük, Turkey
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Inês Barroso
- Human Genetics, Wellcome Sanger Institute, Hinxton, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jacques S Beckmann
- Service of Medical Genetics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Marco P Boks
- Department of Psychiatry, UMC Utrecht Brain Center, University Utrecht, Utrecht, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Heather A Boyd
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lynn F Cherkas
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Gail Davies
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London Medical School, and the Centre for Genomic Health, Queen Mary University of London, London, UK
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - David L Duffy
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Neuherberg, Germany
| | - Ina Giegling
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Scott D Gordon
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Thomas F Hansen
- Institute of Biological Psychiatry, Mental Health Services of Copenhagen, Copenhagen, Denmark
- Danish Headache Center, Copenhagen University Hospital, Glostrup, Denmark
| | - Annette M Hartmann
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Kauko Heikkilä
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Andrew A Hicks
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jennifer E Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Liang-Dar Hwang
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - John P Kemp
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Kay-Tee Khaw
- Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Norman Klopp
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Bettina Konte
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Zoltan Kutalik
- Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Ruth J F Loos
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | | | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Human Genetics, Genentech, South San Francisco, CA, USA
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Vincent Mooser
- Service of Clinical Chemistry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mari Nelis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Dale R Nyholt
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kevin S O'Connell
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Roel A Ophoff
- Department of Human Genetics, University California Los Angeles, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cameron Palmer
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Aarno Palotie
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Teemu Palviainen
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Guillaume Pare
- Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Leena Peltonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Brenda W J H Penninx
- Amsterdam Public Health research institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Ozren Polasek
- Department of Public Health, University of Split School of Medicine, Split, Croatia
- Research Unit, Psychiatric Hospital Sveti Ivan, Zagreb, Croatia
| | | | - Inga Prokopenko
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford, UK
- Section of Genomics of Common Disease, Department of Medicine, Imperial College London, London, UK
| | - Katri Raikkonen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Igor Rudan
- Centre for Global Health Research, The Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Dan Rujescu
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Johannes H Smit
- Amsterdam Public Health research institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | | | - Jordan W Smoller
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | | | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Max Planck Institute for Psycholinguistics, Wundtlaan, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - John M Starr
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemilogy, University of Edinburgh, Edinburgh, UK
| | | | | | - Maris Teder-Laving
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | | | | | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Frank J A van Rooij
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jaqueline M Vink
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eero Vuoksimaa
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Gérard Waeber
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Nicole Warrington
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | | | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Lundbeck Foundation's IPSYCH Initiative, Copenhagen, Denmark
| | | | - Elisabeth Widen
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Alan F Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Mousheng Xu
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard Medical School, Boston, MA, USA
| | - Jing Hua Zhao
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Peter Kraft
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard Medical School, Boston, MA, USA
| | | | - Cecilia M Lindgren
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Benjamin M Neale
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David M Evans
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| | - Sarah E Medland
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia.
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
30
|
Ageta-Ishihara N, Kinoshita M. Developmental and postdevelopmental roles of septins in the brain. Neurosci Res 2020; 170:6-12. [PMID: 33159992 DOI: 10.1016/j.neures.2020.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 11/25/2022]
Abstract
Morphogenetic processes during brain development and postdevelopmental remodeling of neural architecture depend on the exquisite interplay between the microtubule- and actin-based cytoskeletal systems. Accumulation of evidence indicates cooperative roles of another cytoskeletal system composed of the septin family. Here we overview experimental findings on mammalian septins and their hypothetical roles in the proliferation of neural progenitor cells, neurite development, synapse formation and regulations. The diverse, mostly unexpected phenotypes obtained from gain- and loss-of-function mutants point to unknown molecular network to be elucidated, which may underlie pathogenetic processes of infectious diseases and neuropsychiatric disorders in humans.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| | - Makoto Kinoshita
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| |
Collapse
|
31
|
Sai KKS, Prabhakaran J, Damuka N, Craft S, Rajagopal SA, Mintz A, Mann J, Kumar D. Synthesis and Initial In Vivo Evaluations of [
11
C]WX‐132‐18B, a Microtubule PET Imaging Agent. ChemistrySelect 2020. [DOI: 10.1002/slct.202001827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | - Jaya Prabhakaran
- Department of PsychiatryColumbia University Irving Medical Center 1051 Riverside Drive New York 10032 USA
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
| | - Naresh Damuka
- Department of RadiologyWake Forest University School of Medicine Winston-Salem North Carolina 27157 USA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of Medicine Winston Salem North Carolina 27157 USA
| | - Shamyaa A. Rajagopal
- Department of RadiologyWake Forest University School of Medicine Winston-Salem North Carolina 27157 USA
| | - Akiva Mintz
- Department of RadiologyColumbia University Irving Medical Center New York 10032 USA
| | - John Mann
- Department of PsychiatryColumbia University Irving Medical Center 1051 Riverside Drive New York 10032 USA
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
- Department of RadiologyColumbia University Irving Medical Center New York 10032 USA
| | - Dileep Kumar
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
| |
Collapse
|
32
|
Mori T, Kitani Y, Hatakeyama D, Machida K, Goto-Inoue N, Hayakawa S, Yamamoto N, Kashiwagi K, Kashiwagi A. Predation threats for a 24-h period activated the extension of axons in the brains of Xenopus tadpoles. Sci Rep 2020; 10:11737. [PMID: 32678123 PMCID: PMC7367293 DOI: 10.1038/s41598-020-67975-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/24/2020] [Indexed: 11/24/2022] Open
Abstract
The threat of predation is a driving force in the evolution of animals. We have previously reported that Xenopus laevis enhanced their tail muscles and increased their swimming speeds in the presence of Japanese larval salamander predators. Herein, we investigated the induced gene expression changes in the brains of tadpoles under the threat of predation using 3′-tag digital gene expression profiling. We found that many muscle genes were expressed after 24 h of exposure to predation. Ingenuity pathway analysis further showed that after 24 h of a predation threat, various signal transduction genes were stimulated, such as those affecting the actin cytoskeleton and CREB pathways, and that these might increase microtubule dynamics, axonogenesis, cognition, and memory. To verify the increase in microtubule dynamics, DiI was inserted through the tadpole nostrils. Extension of the axons was clearly observed from the nostril to the diencephalon and was significantly increased (P ≤ 0.0001) after 24 h of exposure to predation, compared with that of the control. The dynamic changes in the signal transductions appeared to bring about new connections in the neural networks, as suggested by the microtubule dynamics. These connections may result in improved memory and cognition abilities, and subsequently increase survivability.
Collapse
Affiliation(s)
- Tsukasa Mori
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan.
| | - Yoichiro Kitani
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan.,Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, Japan
| | - Den Hatakeyama
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Kazumasa Machida
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Naoko Goto-Inoue
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University, Kameino 1866, Fujisawa, 252-0880, Japan
| | - Satoshi Hayakawa
- Department of Pathology and Microbiology, School of Medicine, Nihon University, Tokyo, Japan
| | - Naoyuki Yamamoto
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Keiko Kashiwagi
- Amphibian Research Center (Building M), Hiroshima University, Hiroshima, Japan
| | - Akihiko Kashiwagi
- Amphibian Research Center (Building M), Hiroshima University, Hiroshima, Japan
| |
Collapse
|
33
|
Taran AS, Shuvalova LD, Lagarkova MA, Alieva IB. Huntington's Disease-An Outlook on the Interplay of the HTT Protein, Microtubules and Actin Cytoskeletal Components. Cells 2020; 9:E1514. [PMID: 32580314 PMCID: PMC7348758 DOI: 10.3390/cells9061514] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease is a severe and currently incurable neurodegenerative disease. An autosomal dominant mutation in the Huntingtin gene (HTT) causes an increase in the polyglutamine fragment length at the protein N-terminus. The consequence of the mutation is the death of neurons, mostly striatal neurons, leading to the occurrence of a complex of motor, cognitive and emotional-volitional personality sphere disorders in carriers. Despite intensive studies, the functions of both mutant and wild-type huntingtin remain poorly understood. Surprisingly, there is the selective effect of the mutant form of HTT even on nervous tissue, whereas the protein is expressed ubiquitously. Huntingtin plays a role in cell physiology and affects cell transport, endocytosis, protein degradation and other cellular and molecular processes. Our experimental data mining let us conclude that a significant part of the Huntingtin-involved cellular processes is mediated by microtubules and other cytoskeletal cell structures. The review attempts to look at unresolved issues in the study of the huntingtin and its mutant form, including their functions affecting microtubules and other components of the cell cytoskeleton.
Collapse
Affiliation(s)
- Aleksandra S. Taran
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninsky Gory, 119992 Moscow, Russia; (A.S.T.); (L.D.S.)
| | - Lilia D. Shuvalova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninsky Gory, 119992 Moscow, Russia; (A.S.T.); (L.D.S.)
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Maria A. Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Irina B. Alieva
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninsky Gory, 119992 Moscow, Russia
| |
Collapse
|
34
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Lüders J. Nucleating microtubules in neurons: Challenges and solutions. Dev Neurobiol 2020; 81:273-283. [PMID: 32324945 DOI: 10.1002/dneu.22751] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/08/2020] [Accepted: 04/20/2020] [Indexed: 11/08/2022]
Abstract
The highly polarized morphology of neurons is crucial for their function and involves formation of two distinct types of cellular extensions, the axonal and dendritic compartments. An important effector required for the morphogenesis and maintenance and thus the identity of axons and dendrites is the microtubule cytoskeleton. Microtubules in axons and dendrites are arranged with distinct polarities, to allow motor-dependent, compartment-specific sorting of cargo. Despite the importance of the microtubule cytoskeleton in neurons, the molecular mechanisms that generate the intricate compartment-specific microtubule configurations remain largely obscure. Work in other cell types has identified microtubule nucleation, the de novo formation of microtubules, and its spatio-temporal regulation as essential for the proper organization of the microtubule cytoskeleton. Whereas regulation of microtubule nucleation usually involves microtubule organizing centers such as the centrosome, neurons seem to rely largely on decentralized nucleation mechanisms. In this review, I will discuss recent advances in deciphering nucleation mechanisms in neurons, how they contribute to the arrangement of microtubules with specific polarities, and how this affects neuron morphogenesis. While this work has shed some light on these important processes, we are far from a comprehensive understanding. Thus, to provide a coherent model, my discussion will include both well-established mechanisms and mechanisms with more limited supporting data. Finally, I will also highlight important outstanding questions for future investigation.
Collapse
Affiliation(s)
- Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| |
Collapse
|
36
|
Wu Z, Wang G, Wang H, Xiao L, Wei Y, Yang C. Fluoxetine exposure for more than 2 days decreases the neuronal plasticity mediated by CRMP2 in differentiated PC12 cells. Brain Res Bull 2020; 158:99-107. [PMID: 32070769 DOI: 10.1016/j.brainresbull.2020.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/22/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Recent studies indicate that antidepressants treatment restores neuronal plasticity. In contrast, some researchers claim that serotonergic antidepressants, including fluoxetine (FLU), may exacerbate neuronal plasticity, which is contradictory and rarely studied. Since almost those studies exposed cells with drugs for 1-2 days as treatment models of antidepressants, it is possible that FLU exposure for longer periods would have opposite effects on neuronal plasticity. RESULTS In the present study, we examined the effects of FLU exposure (up to 3 days) on the neuronal plasticity in differentiated PC12 cells. The cell viability shown a slight decrease at day 2 (93.5 ± 3.5 %), followed by a highly significant decrease at day 3(71.4 ± 4.4 %). As previously reported, neuronal plasticity was significantly upregulated by FLU exposure at day 1. However, the neurite length, activity-regulated cytoskeleton-associated protein (Arc) and c-Fos mRNA were inhibited with FLU exposure at day 3. Similarly, the expression of tubulin, which play important roles in the neuronal plasticity, was the same result. Furthermore, we found α-tubulin interacted with collapsing response mediator protein 2(CRMP2), which is related to neuronal plasticity, and the regulation of CRMP2 activity influenced the neurite length, Arc, c-Fos and tubulin expression. CONCLUSIONS The results demonstrated that neuronal plasticity was increased by FLU exposure at day 1, but exposure with FLU for more than 2 days had opposite effect on it. The reduction in neuronal plasticity with FLU exposure for more than 2 days might be involved in some aspects of the therapeutic effect of antidepressant on depression.
Collapse
Affiliation(s)
- Zuotian Wu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Yanyan Wei
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| | - Can Yang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China.
| |
Collapse
|
37
|
Calogero AM, Mazzetti S, Pezzoli G, Cappelletti G. Neuronal microtubules and proteins linked to Parkinson's disease: a relevant interaction? Biol Chem 2020; 400:1099-1112. [PMID: 31256059 DOI: 10.1515/hsz-2019-0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022]
Abstract
Neuronal microtubules are key determinants of cell morphology, differentiation, migration and polarity, and contribute to intracellular trafficking along axons and dendrites. Microtubules are strictly regulated and alterations in their dynamics can lead to catastrophic effects in the neuron. Indeed, the importance of the microtubule cytoskeleton in many human diseases is emerging. Remarkably, a growing body of evidence indicates that microtubule defects could be linked to Parkinson's disease pathogenesis. Only a few of the causes of the progressive neuronal loss underlying this disorder have been identified. They include gene mutations and toxin exposure, but the trigger leading to neurodegeneration is still unknown. In this scenario, the evidence showing that mutated proteins in Parkinson's disease are involved in the regulation of the microtubule cytoskeleton is intriguing. Here, we focus on α-Synuclein, Parkin and Leucine-rich repeat kinase 2 (LRRK2), the three main proteins linked to the familial forms of the disease. The aim is to dissect their interaction with tubulin and microtubules in both physiological and pathological conditions, in which these proteins are overexpressed, mutated or absent. We highlight the relevance of such an interaction and suggest that these proteins could trigger neurodegeneration via defective regulation of the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Alessandra M Calogero
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy.,Parkinson Institute, ASST "G.Pini-CTO", via Bignami 1, I-20133 Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti, I-20133 Milan, Italy
| |
Collapse
|
38
|
Trushina NI, Mulkidjanian AY, Brandt R. The microtubule skeleton and the evolution of neuronal complexity in vertebrates. Biol Chem 2020; 400:1163-1179. [PMID: 31116700 DOI: 10.1515/hsz-2019-0149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022]
Abstract
The evolution of a highly developed nervous system is mirrored by the ability of individual neurons to develop increased morphological complexity. As microtubules (MTs) are crucially involved in neuronal development, we tested the hypothesis that the evolution of complexity is driven by an increasing capacity of the MT system for regulated molecular interactions as it may be implemented by a higher number of molecular players and a greater ability of the individual molecules to interact. We performed bioinformatics analysis on different classes of components of the vertebrate neuronal MT cytoskeleton. We show that the number of orthologs of tubulin structure proteins, MT-binding proteins and tubulin-sequestering proteins expanded during vertebrate evolution. We observed that protein diversity of MT-binding and tubulin-sequestering proteins increased by alternative splicing. In addition, we found that regions of the MT-binding protein tau and MAP6 displayed a clear increase in disorder extent during evolution. The data provide evidence that vertebrate evolution is paralleled by gene expansions, changes in alternative splicing and evolution of coding sequences of components of the MT system. The results suggest that in particular evolutionary changes in tubulin-structure proteins, MT-binding proteins and tubulin-sequestering proteins were prominent drivers for the development of increased neuronal complexity.
Collapse
Affiliation(s)
- Nataliya I Trushina
- Department of Neurobiology, University of Osnabrück, Barbarastraße 11, D-49076 Osnabrück, Germany
| | - Armen Y Mulkidjanian
- Department of Physics, University of Osnabrück, Barbarastraße 7, D-49076 Osnabrück, Germany.,A.N. Belozersky Institute of Physico-Chemical Biology and School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Barbarastraße 11, D-49076 Osnabrück, Germany.,Center for Cellular Nanoanalytics, University of Osnabrück, Barbarastraße 11, D-49076 Osnabrück, Germany.,Institute of Cognitive Science, University of Osnabrück, Barbarastraße 11, D-49076 Osnabrück, Germany
| |
Collapse
|
39
|
Coombes CE, Saunders HAJ, Mannava AG, Johnson-Schlitz DM, Reid TA, Parmar S, McClellan M, Yan C, Rogers SL, Parrish JZ, Wagenbach M, Wordeman L, Wildonger J, Gardner MK. Non-enzymatic Activity of the α-Tubulin Acetyltransferase αTAT Limits Synaptic Bouton Growth in Neurons. Curr Biol 2020; 30:610-623.e5. [PMID: 31928876 DOI: 10.1016/j.cub.2019.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/16/2019] [Accepted: 12/06/2019] [Indexed: 10/25/2022]
Abstract
Neuronal axons terminate as synaptic boutons that form stable yet plastic connections with their targets. Synaptic bouton development relies on an underlying network of both long-lived and dynamic microtubules that provide structural stability for the boutons while also allowing for their growth and remodeling. However, a molecular-scale mechanism that explains how neurons appropriately balance these two microtubule populations remains a mystery. We hypothesized that α-tubulin acetyltransferase (αTAT), which both stabilizes long-lived microtubules against mechanical stress via acetylation and has been implicated in promoting microtubule dynamics, could play a role in this process. Using the Drosophila neuromuscular junction as a model, we found that non-enzymatic dαTAT activity limits the growth of synaptic boutons by affecting dynamic, but not stable, microtubules. Loss of dαTAT results in the formation of ectopic boutons. These ectopic boutons can be similarly suppressed by resupplying enzyme-inactive dαTAT or by treatment with a low concentration of the microtubule-targeting agent vinblastine, which acts to suppress microtubule dynamics. Biophysical reconstitution experiments revealed that non-enzymatic αTAT1 activity destabilizes dynamic microtubules but does not substantially impact the stability of long-lived microtubules. Further, during microtubule growth, non-enzymatic αTAT1 activity results in increasingly extended tip structures, consistent with an increased rate of acceleration of catastrophe frequency with microtubule age, perhaps via tip structure remodeling. Through these mechanisms, αTAT enriches for stable microtubules at the expense of dynamic ones. We propose that the specific suppression of dynamic microtubules by non-enzymatic αTAT activity regulates the remodeling of microtubule networks during synaptic bouton development.
Collapse
Affiliation(s)
- Courtney E Coombes
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Harriet A J Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anirudh G Mannava
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Taylor A Reid
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sneha Parmar
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Connie Yan
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Stephen L Rogers
- Department of Biology, Integrative Program for Biological and Genome Sciences, and Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Michael Wagenbach
- Department of Physiology and Biophysics, The University of Washington, Seattle, WA 98195, USA
| | - Linda Wordeman
- Department of Physiology and Biophysics, The University of Washington, Seattle, WA 98195, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
40
|
Hahn I, Voelzmann A, Liew YT, Costa-Gomes B, Prokop A. The model of local axon homeostasis - explaining the role and regulation of microtubule bundles in axon maintenance and pathology. Neural Dev 2019; 14:11. [PMID: 31706327 PMCID: PMC6842214 DOI: 10.1186/s13064-019-0134-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Axons are the slender, cable-like, up to meter-long projections of neurons that electrically wire our brains and bodies. In spite of their challenging morphology, they usually need to be maintained for an organism's lifetime. This makes them key lesion sites in pathological processes of ageing, injury and neurodegeneration. The morphology and physiology of axons crucially depends on the parallel bundles of microtubules (MTs), running all along to serve as their structural backbones and highways for life-sustaining cargo transport and organelle dynamics. Understanding how these bundles are formed and then maintained will provide important explanations for axon biology and pathology. Currently, much is known about MTs and the proteins that bind and regulate them, but very little about how these factors functionally integrate to regulate axon biology. As an attempt to bridge between molecular mechanisms and their cellular relevance, we explain here the model of local axon homeostasis, based on our own experiments in Drosophila and published data primarily from vertebrates/mammals as well as C. elegans. The model proposes that (1) the physical forces imposed by motor protein-driven transport and dynamics in the confined axonal space, are a life-sustaining necessity, but pose a strong bias for MT bundles to become disorganised. (2) To counterbalance this risk, MT-binding and -regulating proteins of different classes work together to maintain and protect MT bundles as necessary transport highways. Loss of balance between these two fundamental processes can explain the development of axonopathies, in particular those linking to MT-regulating proteins, motors and transport defects. With this perspective in mind, we hope that more researchers incorporate MTs into their work, thus enhancing our chances of deciphering the complex regulatory networks that underpin axon biology and pathology.
Collapse
Affiliation(s)
- Ines Hahn
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - André Voelzmann
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Yu-Ting Liew
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Beatriz Costa-Gomes
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK.
| |
Collapse
|
41
|
Maraldi T, Beretti F, Anselmi L, Franchin C, Arrigoni G, Braglia L, Mandrioli J, Vinceti M, Marmiroli S. Influence of selenium on the emergence of neuro tubule defects in a neuron-like cell line and its implications for amyotrophic lateral sclerosis. Neurotoxicology 2019; 75:209-220. [PMID: 31585128 DOI: 10.1016/j.neuro.2019.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
Impairment of the axonal transport system mediated by intracellular microtubules (MTs) is known to be a major drawback in neurodegenerative processes. Due to a growing interest on the neurotoxic effects of selenium in environmental health, our study aimed to assess the relationship between selenium and MTs perturbation, that may favour disease onset over a genetic predisposition to amyotrophic lateral sclerosis. We treated a neuron-like cell line with sodium selenite, sodium selenate and seleno-methionine and observed that the whole cytoskeleton was affected. We then investigated the protein interactome of cells overexpressing αTubulin-4A (TUBA4A) and found that selenium increases the interaction of TUBA4A with DNA- and RNA-binding proteins. TUBA4A ubiquitination and glutathionylation were also observed, possibly due to a selenium-dependent increase of ROS, leading to perturbation and degradation of MTs. Remarkably, the TUBA4A mutants R320C and A383 T, previously described in ALS patients, showed the same post-translational modifications to a similar extent. In conclusion this study gives insights into a specific mechanism characterizing selenium neurotoxicity.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy.
| | - Francesca Beretti
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via Del Pozzo 71, 41124, Modena, Italy.
| | - Laura Anselmi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Cinzia Franchin
- Department of Biomedical Sciences, University of Padova, via G. Basso 58/B, 35131, Padova, Italy; Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, via G. Orus 2/B, 35129, Padova, Italy.
| | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padova, via G. Basso 58/B, 35131, Padova, Italy; Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, via G. Orus 2/B, 35129, Padova, Italy.
| | - Luca Braglia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Jessica Mandrioli
- Neurology Unit, Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy.
| | - Marco Vinceti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy; Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States.
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| |
Collapse
|
42
|
Apple E, Chen L. Neuronal microtubules impact lifespan. Aging (Albany NY) 2019; 11:6616-6617. [PMID: 31492827 PMCID: PMC6756882 DOI: 10.18632/aging.102224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/05/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Ellen Apple
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, UTHSCSA, San Antonio, TX 7822, USA
| | - Lizhen Chen
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, UTHSCSA, San Antonio, TX 7822, USA
| |
Collapse
|
43
|
Gattoni G, Bernocchi G. Calcium-Binding Proteins in the Nervous System during Hibernation: Neuroprotective Strategies in Hypometabolic Conditions? Int J Mol Sci 2019; 20:E2364. [PMID: 31086053 PMCID: PMC6540041 DOI: 10.3390/ijms20092364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/06/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium-binding proteins (CBPs) can influence and react to Ca2+ transients and modulate the activity of proteins involved in both maintaining homeostatic conditions and protecting cells in harsh environmental conditions. Hibernation is a strategy that evolved in vertebrate and invertebrate species to survive in cold environments; it relies on molecular, cellular, and behavioral adaptations guided by the neuroendocrine system that together ensure unmatched tolerance to hypothermia, hypometabolism, and hypoxia. Therefore, hibernation is a useful model to study molecular neuroprotective adaptations to extreme conditions, and can reveal useful applications to human pathological conditions. In this review, we describe the known changes in Ca2+-signaling and the detection and activity of CBPs in the nervous system of vertebrate and invertebrate models during hibernation, focusing on cytosolic Ca2+ buffers and calmodulin. Then, we discuss these findings in the context of the neuroprotective and neural plasticity mechanisms in the central nervous system: in particular, those associated with cytoskeletal proteins. Finally, we compare the expression of CBPs in the hibernating nervous system with two different conditions of neurodegeneration, i.e., platinum-induced neurotoxicity and Alzheimer's disease, to highlight the similarities and differences and demonstrate the potential of hibernation to shed light into part of the molecular mechanisms behind neurodegenerative diseases.
Collapse
Affiliation(s)
- Giacomo Gattoni
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Graziella Bernocchi
- Former Full Professor of Zoology, Neurogenesis and Comparative Neuromorphology, (Residence address) Viale Matteotti 73, I-27100 Pavia, Italy.
| |
Collapse
|
44
|
Tang M, Luo Z, Wu Y, Zhuang J, Li K, Hu D, Rong H, Xian B, Ge J. BAM15 attenuates transportation-induced apoptosis in iPS-differentiated retinal tissue. Stem Cell Res Ther 2019; 10:64. [PMID: 30795805 PMCID: PMC6387563 DOI: 10.1186/s13287-019-1151-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 02/08/2023] Open
Abstract
Background BAM15 is a novel mitochondrial protonophore uncoupler capable of protecting mammals from acute renal ischemic-reperfusion injury and cold-induced microtubule damage. The purpose of our study was to investigate the effect of BAM15 on apoptosis during 5-day transportation of human-induced pluripotent stem (hiPS)-differentiated retinal tissue. Methods Retinal tissues of 30 days and 60 days were transported with or without BAM15 for 5 days in the laboratory or by real express. Immunofluorescence staining of apoptosis marker cleaved caspase3, proliferation marker Ki67, and neural axon marker NEFL was performed. And expression of apoptotic-related factors p53, NFkappaB, and TNF-a was detected by real-time PCR. Also, location of ganglion cells, photoreceptor cells, amacrine cells, and precursors of neuronal cell types in retinal tissue was stained by immunofluorescence after transportation. Furthermore, cell viability was assessed by CCK8 assay. Results Results showed transportation remarkably intensified expression of apoptotic factor cleaved caspase3, p53, NFkappaB, and TNF-a, which could be reduced by supplement of BAM15. In addition, neurons were severely injured after transportation, with axons manifesting disrupted and tortuous by staining NEFL. And the addition of BAM15 in transportation was able to protect neuronal structure and increase cell viability without affecting subtypes cells location of retinal tissue. Conclusions BAM15 might be used as a protective reagent on apoptosis during transporting retinal tissues, holding great potential in research and clinical applications. Electronic supplementary material The online version of this article (10.1186/s13287-019-1151-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mingjun Tang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Ziming Luo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Yihui Wu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Dongpeng Hu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Huifeng Rong
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Bikun Xian
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China.
| |
Collapse
|
45
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
46
|
Paternoster V, Svanborg M, Edhager AV, Rajkumar AP, Eickhardt EA, Pallesen J, Grove J, Qvist P, Fryland T, Wegener G, Nyengaard JR, Mors O, Palmfeldt J, Børglum AD, Christensen JH. Brain proteome changes in female Brd1 +/- mice unmask dendritic spine pathology and show enrichment for schizophrenia risk. Neurobiol Dis 2018; 124:479-488. [PMID: 30590179 DOI: 10.1016/j.nbd.2018.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/23/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Genetic and molecular studies have implicated the Bromodomain containing 1 (BRD1) gene in the pathogenesis of schizophrenia and bipolar disorder. Accordingly, mice heterozygous for a targeted deletion of Brd1 (Brd1+/- mice) show behavioral phenotypes with broad translational relevance to psychiatric disorders. BRD1 encodes a scaffold protein that affects the expression of many genes through modulation of histone acetylation. BRD1 target genes have been identified in cell lines; however the impact of reduced Brd1 levels on the brain proteome is largely unknown. In this study, we applied label-based quantitative mass spectrometry to profile the frontal cortex, hippocampus and striatum proteome and synaptosomal proteome of female Brd1+/- mice. We successfully quantified between 1537 and 2196 proteins and show widespread changes in protein abundancies and compartmentalization. By integrative analysis of human genetic data, we find that the differentially abundant proteins in frontal cortex and hippocampus are enriched for schizophrenia risk further linking the actions of BRD1 to psychiatric disorders. Affected proteins were further enriched for proteins involved in processes known to influence neuronal and dendritic spine morphology e.g. regulation of cytoskeleton dynamics and mitochondrial function. Directly prompted in these findings, we investigated dendritic spine morphology of pyramidal neurons in anterior cingulate cortex and found them significantly altered, including reduced size of small dendritic spines and decreased number of the mature mushroom type. Collectively, our study describes known as well as new mechanisms related to BRD1 dysfunction and its role in psychiatric disorders, and provides evidence for the molecular and cellular dysfunctions underlying altered neurosignalling and cognition in Brd1+/- mice.
Collapse
Affiliation(s)
- Veerle Paternoster
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.
| | - Maria Svanborg
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Anto P Rajkumar
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Mental Health of Older Adults and Dementia Clinical Academic Group, South London and Maudsley NHS Foundation Trust, London, UK; Department of Old Age Psychiatry, Psychology, & Neuroscience, King's College London, Institute of Psychiatry, London, UK
| | - Esben Ahlburg Eickhardt
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jonatan Pallesen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Bioinformatics Research Centre, BiRC, Aarhus University, Aarhus, Denmark
| | - Per Qvist
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tue Fryland
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Jens Randel Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Dupont Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jane Hvarregaard Christensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Pradeepkiran JA, Reddy AP, Reddy PH. Pharmacophore-based models for therapeutic drugs against phosphorylated tau in Alzheimer's disease. Drug Discov Today 2018; 24:616-623. [PMID: 30453058 DOI: 10.1016/j.drudis.2018.11.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 09/22/2018] [Accepted: 11/07/2018] [Indexed: 10/27/2022]
Abstract
Phosphorylated tau (P-tau) has received much attention in the field of Alzheimer's disease (AD), as a potential therapeutic target owing to its involvement with synaptic damage and neuronal dysfunction. The continuous failure of amyloid β (Aβ)-targeted therapeutics highlights the urgency to consider alternative therapeutic strategies for AD. The present review describes the latest developments in tau biology and function. It also explains abnormal interactions between P-tau with Aβ and the mitochondrial fission protein Drp1, leading to excessive mitochondrial fragmentation and synaptic damage in AD neurons. This article also addresses 3D pharmacophore-based drug models designed to treat patients with AD and other tauopathies.
Collapse
Affiliation(s)
- Jangampalli Adi Pradeepkiran
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA; Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA; Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, USA; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, USA.
| |
Collapse
|
48
|
Hibernation induces changes in the metacerebral neurons of Cornu aspersum: distribution and co-localization of cytoskeletal and calcium-binding proteins. INVERTEBRATE NEUROSCIENCE 2018; 18:13. [DOI: 10.1007/s10158-018-0217-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/08/2018] [Indexed: 01/05/2023]
|
49
|
Schverer M, Lanfumey L, Baulieu EE, Froger N, Villey I. Neurosteroids: non-genomic pathways in neuroplasticity and involvement in neurological diseases. Pharmacol Ther 2018; 191:190-206. [PMID: 29953900 DOI: 10.1016/j.pharmthera.2018.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neurosteroids are neuroactive brain-born steroids. They can act through non-genomic and/or through genomic pathways. Genomic pathways are largely described for steroid hormones: the binding to nuclear receptors leads to transcription regulation. Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone have no corresponding nuclear receptor identified so far whereas some of their non-genomic targets have been identified. Neuroplasticity is the capacity that neuronal networks have to change their structure and function in response to biological and/or environmental signals; it is regulated by several mechanisms, including those that involve neurosteroids. In this review, after a description of their biosynthesis, the effects of Pregnenolone, Dehydroepiandrosterone, their respective sulfate esters and Allopregnanolone on their targets will be exposed. We then shall highlight that neurosteroids, by acting on these targets, can regulate neurogenesis, structural and functional plasticity. Finally, we will discuss the therapeutic potential of neurosteroids in the pathophysiology of neurological diseases in which alterations of neuroplasticity are associated with changes in neurosteroid levels.
Collapse
Affiliation(s)
- Marina Schverer
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France
| | - Laurence Lanfumey
- Inserm U894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014 Paris, France.
| | - Etienne-Emile Baulieu
- MAPREG SAS, Le Kremlin-Bicêtre, France; Inserm UMR 1195, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | | | | |
Collapse
|
50
|
Banerjee A, Paluh JL, Mukherjee A, Kumar KG, Ghosh A, Naskar MK. Modeling the neuron as a nanocommunication system to identify spatiotemporal molecular events in neurodegenerative disease. Int J Nanomedicine 2018; 13:3105-3128. [PMID: 29872297 PMCID: PMC5975603 DOI: 10.2147/ijn.s152664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM In tauopathies such as Alzheimer's disease (AD), molecular changes spanning multiple subcellular compartments of the neuron contribute to neurodegeneration and altered axonal signaling. Computational modeling of end-to-end linked events benefit mechanistic analysis and can be informative to understand disease progression and accelerate development of effective therapies. In the calcium-amyloid beta model of AD, calcium ions that are an important regulator of neuronal function undergo dysregulated homeostasis that disrupts cargo loading for neurotrophic signaling along axonal microtubules (MTs). The aim of the present study was to develop a computational model of the neuron using a layered architecture simulation that enables us to evaluate the functionalities of several interlinked components in the calcium-amyloid beta model. METHODS The elevation of intracellular calcium levels is modeled upon binding of amyloid beta oligomers (AβOs) to calcium channels or as a result of membrane insertion of oligomeric Aβ1-42 to form pores/channels. The resulting subsequent Ca2+ disruption of dense core vesicle (DCV)-kinesin cargo loading and transport of brain-derived neurotrophic factor (BDNF) on axonal MTs are then evaluated. Our model applies published experimental data on calcium channel manipulation of DCV-BDNF and incorporates organizational complexity of the axon as bundled polar and discontinuous MTs. The interoperability simulation is based on the Institute of Electrical and Electronics Engineers standard association P1906.1 framework for nanoscale and molecular communication. RESULTS Our analysis provides new spatiotemporal insights into the end-to-end signaling events linking calcium dysregulation and BDNF transport and by simulation compares the relative impact of dysregulation of calcium levels by AβO-channel interactions, oligomeric Aβ1-42 pores/channel formation, and release of calcium by internal stores. The flexible platform of our model allows continued expansion of molecular details including mechanistic and morphological parameters of axonal cytoskeleton networks as they become available to test disease and treatment predictions. CONCLUSION The present model will benefit future drug studies on calcium homeostasis and dysregulation linked to measurable neural functional outcomes. The algorithms used can also link to other multiscale multi-cellular modeling platforms to fill in molecular gaps that we believe will assist in broadening and refining multiscale computational maps of neurodegeneration.
Collapse
Affiliation(s)
- Arunima Banerjee
- Department of Electronics and Tele-Communication Engineering, Jadavpur University, Kolkata, India
| | - Janet L Paluh
- College of Nanoscale Science, Nanobioscience Constellation, State University of New York Polytechnic Institute, Albany, NY, USA
| | | | - K Gaurav Kumar
- Department of Electronics and Tele-Communication Engineering, Jadavpur University, Kolkata, India
| | - Archisman Ghosh
- Department of Electronics and Tele-Communication Engineering, Jadavpur University, Kolkata, India
| | - Mrinal K Naskar
- Department of Electronics and Tele-Communication Engineering, Jadavpur University, Kolkata, India
| |
Collapse
|