1
|
Cui Z, Qiu J, Lin J, Fu Y, Lin L. Discovering genetically-supported drug targets for multisite chronic pain through multi-omics Mendelian randomization and single-cell RNA-sequencing. Neuroscience 2025; 572:254-268. [PMID: 39993665 DOI: 10.1016/j.neuroscience.2025.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/14/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Multisite chronic pain (MCP) is a highly prevalent disorder with substantial unmet therapeutic needs.We conducted multi-omics Mendelian randomization and Bayesian colocalization to identify potential therapeutic targets for MCP. Summary-level data of gene expressions and protein abundance levels were obtained from corresponding quantitative trait loci studies, respectively. Summary-level data for MCP was leveraged from the UK Biobank. The transcriptome-wide association study (TWAS), Mendelian randomization, and Bayesian colocalization approaches were applied to investigate the potential causal effects of gene expressions and protein levels on MCP in both blood and brain tissues. Phenome-wide Mendelian randomization analysis (MR-PheWAS), single-cell sequencing data, protein-protein interaction (PPI), and reaction pathway analysis were further conducted to digging the underlying mechanisms. Our analysis identified and validated two plasma targets for MCP, namely KLC1 and LANCL1, at both gene expression levels and protein levels across multi-methodologies. Moreover, MR-PheWAS observed additional benefits associated with these two targets. Through analyses based on single-cell sequencing data, we identified critical cell types for KLC1, primarily megakaryocytes, and neurons, notably linked to the axon guidance pathway, while LANCL1 showed associations with B lymphocytes, neurons, and the electron transport pathway. In dorsal root ganglions, we identified enrichments of both LANCL1 and KLC1 in putative silent nociceptors. The effects are possibly mediated through axonal transport and the activation of NMDARs, supported by PPI and reaction pathway analysis. Our multi-dimensional analysis suggests that genetically determined KLC1 and LANCL1 are causally linked to MCP risk, holding promise as appealing drug targets for MCP.
Collapse
Affiliation(s)
- Ziyang Cui
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jianwei Lin
- Big Data Laboratory, Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China.
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Liling Lin
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Jáuregui GV, Parpura V. Neuron-Astrocyte Interactions in Aging and Alzheimer's Disease: Dysregulation of Amyloid Precursor Protein. AGEING & LONGEVITY 2025; 6:117-128. [PMID: 40098995 PMCID: PMC11911455 DOI: 10.47855/jal9020-2025-2-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Amyloid precursor protein (APP) is central to Alzheimer's disease (AD) by its role in Aβ build-up and in neuronal and astrocytic malfunction. The major risk factor for late-onset AD is aging, which increases APP processing in both neurons and astrocytes, and consequently increases Aβ production. This focused review covers the subjects of how aging and AD affect APP dynamics within the both cell types and how astrocytes dysfunction can enhance neuroinflammation and neuronal dysfunction and injury. We discuss the interplay between neurons and astrocytes in aging and AD brains, where bi-directional cellular interactions accelerate neurodegeneration.
Collapse
Affiliation(s)
- Gretsen Velezmoro Jáuregui
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Vladimir Parpura
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
3
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-z] [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: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
4
|
Duswald T, Breitwieser L, Thorne T, Wohlmuth B, Bauer R. Calibration of stochastic, agent-based neuron growth models with approximate Bayesian computation. J Math Biol 2024; 89:50. [PMID: 39379537 PMCID: PMC11461709 DOI: 10.1007/s00285-024-02144-2] [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: 05/22/2024] [Revised: 05/22/2024] [Accepted: 08/31/2024] [Indexed: 10/10/2024]
Abstract
Understanding how genetically encoded rules drive and guide complex neuronal growth processes is essential to comprehending the brain's architecture, and agent-based models (ABMs) offer a powerful simulation approach to further develop this understanding. However, accurately calibrating these models remains a challenge. Here, we present a novel application of Approximate Bayesian Computation (ABC) to address this issue. ABMs are based on parametrized stochastic rules that describe the time evolution of small components-the so-called agents-discretizing the system, leading to stochastic simulations that require appropriate treatment. Mathematically, the calibration defines a stochastic inverse problem. We propose to address it in a Bayesian setting using ABC. We facilitate the repeated comparison between data and simulations by quantifying the morphological information of single neurons with so-called morphometrics and resort to statistical distances to measure discrepancies between populations thereof. We conduct experiments on synthetic as well as experimental data. We find that ABC utilizing Sequential Monte Carlo sampling and the Wasserstein distance finds accurate posterior parameter distributions for representative ABMs. We further demonstrate that these ABMs capture specific features of pyramidal cells of the hippocampus (CA1). Overall, this work establishes a robust framework for calibrating agent-based neuronal growth models and opens the door for future investigations using Bayesian techniques for model building, verification, and adequacy assessment.
Collapse
Affiliation(s)
- Tobias Duswald
- CERN, Geneva, Switzerland.
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany.
| | - Lukas Breitwieser
- Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Thomas Thorne
- School of Computer Science and Electronic Engineering, University of Surrey, Guildford, UK
| | - Barbara Wohlmuth
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany
| | - Roman Bauer
- School of Computer Science and Electronic Engineering, University of Surrey, Guildford, UK
| |
Collapse
|
5
|
Vaillant-Beuchot L, Eysert F, Duval B, Kinoshita PF, Pardossi-Piquard R, Bauer C, Eddarkaoui S, Buée L, Checler F, Chami M. The amyloid precursor protein and its derived fragments concomitantly contribute to the alterations of mitochondrial transport machinery in Alzheimer's disease. Cell Death Dis 2024; 15:367. [PMID: 38806484 PMCID: PMC11133367 DOI: 10.1038/s41419-024-06742-2] [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: 08/04/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Mitochondria dysfunctions and mitophagy failure have been associated with several Alzheimer's disease (AD) related molecular actors including amyloid beta (Aβ) and recently the amyloid precursor protein-C terminal fragments (APP-CTFs). The efficacy of the mitophagy process in neurons relies on regulated mitochondrial transport along axons involving a complex molecular machinery. The contribution of the amyloid precursor protein (APP) and its derived fragments to the mitochondrial transport machinery alterations in AD have not been investigated before. We report herein a change of the expression of mitochondrial transport proteins (SNPH and Miro1), motor adapters (TRANK1 and TRAK2), and components of the dynein and kinesin motors (i.e., IC1,2 and Kif5 (A, B, C) isoforms) by endogenous APP and by overexpression of APP carrying the familial Swedish mutation (APPswe). We show that APP-CTFs and Aβ concomitantly regulate the expression of a set of transport proteins as demonstrated in APPswe cells treated with β- and γ-secretase inhibitors and in cells Knock-down for presenilin 1 and 2. We further report the impact of APP-CTFs on the expression of transport proteins in AAV-injected C99 mice brains. Our data also indicate that both Aβ oligomers (Aβo) and APP-CTFs impair the colocalization of mitochondria and transport proteins. This has been demonstrated in differentiated SH-SY5Y naive cells treated with Aβo and in differentiated SH-SY5Y and murine primary neurons expressing APPswe and treated with the γ-secretase inhibitor. Importantly, we uncover that the expression of a set of transport proteins is modulated in a disease-dependent manner in 3xTgAD mice and in human sporadic AD brains. This study highlights molecular mechanisms underlying mitochondrial transport defects in AD that likely contribute to mitophagy failure and disease progression.
Collapse
Affiliation(s)
- Loan Vaillant-Beuchot
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Fanny Eysert
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Blandine Duval
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Paula Fernanda Kinoshita
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
- Instituto de Ciências Biomédicas Department of Pharmacology, Universidade de São Paulo, São Paulo, Brazil
| | - Raphaëlle Pardossi-Piquard
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Charlotte Bauer
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, 59045, Lille, France
- Inserm UMR-S 1172, Laboratory of excellence DistALZ, 'Alzheimer and Tauopathies', Bâtiment Biserte, rue Polonovski, 59045, Lille, Cedex, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, 59045, Lille, France
- Inserm UMR-S 1172, Laboratory of excellence DistALZ, 'Alzheimer and Tauopathies', Bâtiment Biserte, rue Polonovski, 59045, Lille, Cedex, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France
| | - Mounia Chami
- Université Côte d'Azur, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of excellence DistALZ, 06560, Sophia-Antipolis, Valbonne, France.
| |
Collapse
|
6
|
Feole M, Pozo Devoto VM, Dragišić N, Arnaiz C, Bianchelli J, Texlová K, Kovačovicova K, Novotny JS, Havas D, Falzone TL, Stokin GB. Swedish Alzheimer's disease variant perturbs activity of retrograde molecular motors and causes widespread derangement of axonal transport pathways. J Biol Chem 2024; 300:107137. [PMID: 38447793 PMCID: PMC10997842 DOI: 10.1016/j.jbc.2024.107137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Experimental studies in flies, mice, and humans suggest a significant role of impaired axonal transport in the pathogenesis of Alzheimer's disease (AD). The mechanisms underlying these impairments in axonal transport, however, remain poorly understood. Here we report that the Swedish familial AD mutation causes a standstill of the amyloid precursor protein (APP) in the axons at the expense of its reduced anterograde transport. The standstill reflects the perturbed directionality of the axonal transport of APP, which spends significantly more time traveling in the retrograde direction. This ineffective movement is accompanied by an enhanced association of dynactin-1 with APP, which suggests that reduced anterograde transport of APP is the result of enhanced activation of the retrograde molecular motor dynein by dynactin-1. The impact of the Swedish mutation on axonal transport is not limited to the APP vesicles since it also reverses the directionality of a subset of early endosomes, which become enlarged and aberrantly accumulate in distal locations. In addition, it also reduces the trafficking of lysosomes due to their less effective retrograde movement. Altogether, our experiments suggest a pivotal involvement of retrograde molecular motors and transport in the mechanisms underlying impaired axonal transport in AD and reveal significantly more widespread derangement of axonal transport pathways in the pathogenesis of AD.
Collapse
Affiliation(s)
- Monica Feole
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Faculty of Medicine, Department of Biology, Masaryk University, Brno, Czech Republic; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Victorio M Pozo Devoto
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic
| | - Neda Dragišić
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic
| | - Cayetana Arnaiz
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Julieta Bianchelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Kateřina Texlová
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; PsychoGenics, Paramus, New Jersey, USA
| | | | - Jan S Novotny
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | | | - Tomas L Falzone
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA-CONICET-MPSP), Partner Institute of the Max Planck Society, Buenos Aires, Argentina; Instituto de Biología Celular y Neurociencia IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gorazd B Stokin
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St Anne's University Hospital, Brno, Czech Republic; Institute for Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic; Division of Neurology, University Medical Centre, Ljubljana, Slovenia; Department of Neurosciences, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
7
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. Mol Neurodegener 2024; 19:13. [PMID: 38282024 PMCID: PMC10823734 DOI: 10.1186/s13024-023-00690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Zhen-Xian Niou
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Andrea Enriquez
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
- Present address: Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Karen Ling
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Paymaan Jafar-Nejad
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Jonathan Gilley
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Michael P Coleman
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
8
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572105. [PMID: 38187564 PMCID: PMC10769303 DOI: 10.1101/2023.12.18.572105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent E2 treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p=1.6×10-51) and upregulation (p=3.8×10-3) of UBE2M across both brain regions, provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p=1.9×10-4; interaction p=3.5×10-2) of LTBR in the PFC, provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step towards understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Kip D. Zimmerman
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Department of Internal Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Larry Wilhelm
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Dongqin Zhu
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Jessica Bodie
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Steven G. Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
9
|
Shiraki Y, Mitsuma M, Takada R, Hata S, Kitamura A, Takada S, Kinjo M, Taru H, Müller UC, Yamamoto T, Sobu Y, Suzuki T. Axonal transport of Frizzled5 by Alcadein α-containing vesicles is associated with kinesin-1. Mol Biol Cell 2023; 34:ar110. [PMID: 37585286 PMCID: PMC10559311 DOI: 10.1091/mbc.e22-10-0495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
Alcadein α (Alcα) and amyloid-β protein precursor (APP) are cargo receptors that associate vesicles with kinesin-1. These vesicles, which contain either Alcα or APP, transport various proteins/cargo molecules into axon nerve terminals. Here, we analyzed immune-isolated Alcα- and APP-containing vesicles of adult mouse brains with LC-MS/MS and identified proteins present in vesicles that contained either Alcα or APP. Among these proteins, Frizzled-5 (Fzd5), a Wnt receptor, was detected mainly in Alcα vesicles. Although colocalization ratios of Fzd5 with Alcα are low in the neurites of differentiating neurons by a low expression of Fzd5 in embryonic brains, the suppression of Alcα expression decreased the localization of Fzd5 in neurites of primary cultured neurons. Furthermore, Fzd5-EGFP expressed in primary cultured neurons was preferentially transported in axons with the transport velocities of Alcα vesicles. In synaptosomal fractions of adult-mice brains that express higher levels of Fzd5, the amount of Fzd5 and the phosphorylation level of calcium/calmodulin-dependent protein kinase-II were reduced in the Alcα-deficient mice. These results suggest that reduced transport of Fzd5 by Alcα-containing vesicles associated with kinesin-1 in axon terminals may impair the response to Wnt ligands in the noncanonical Ca2+-dependent signal transduction pathway at nerve terminals of mature neurons.
Collapse
Affiliation(s)
- Yuzuha Shiraki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Monet Mitsuma
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ritsuko Takada
- Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi 444-8787, Japan
- National Institute for Basic Biology, National Institute of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Akira Kitamura
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
- AMED-PRIME, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004 Japan
| | - Shinji Takada
- Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi 444-8787, Japan
- National Institute for Basic Biology, National Institute of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo 001-0021, Japan
| | - Hidenori Taru
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ulrike C. Müller
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa 761-0793, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyotanabe 610-0394, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
10
|
Skeldal S, Voss LF, Lende J, Pedersen SB, Mølgaard S, Kaas M, Demange P, Bentsen AH, Fuglsang M, Sander MR, Buttenschøn H, Gustafsen C, Madsen P, Glerup S. Alternative splicing regulates adaptor protein binding, trafficking, and activity of the Vps10p domain receptor SorCS2 in neuronal development. J Biol Chem 2023; 299:105102. [PMID: 37507021 PMCID: PMC10463258 DOI: 10.1016/j.jbc.2023.105102] [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: 04/01/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The Vps10p domain receptor SorCS2 is crucial for the development and function of the nervous system and essential for brain-derived neurotrophic factor (BDNF)-induced changes in neuronal morphology and plasticity. SorCS2 regulates the subcellular trafficking of the BDNF signaling receptor TrkB as well as selected neurotransmitter receptors in a manner that is dependent on the SorCS2 intracellular domain (ICD). However, the cellular machinery and adaptor protein (AP) interactions that regulate receptor trafficking via the SorCS2 ICD are unknown. We here identify four splice variants of human SorCS2 differing in the insertion of an acidic cluster motif and/or a serine residue within the ICD. We show that each variant undergoes posttranslational proteolytic processing into a one- or two-chain receptor, giving rise to eight protein isoforms, the expression of which differs between neuronal and nonneuronal tissues and is affected by cellular stressors. We found that the only variants without the serine were able to rescue BDNF-induced branching of SorCS2 knockout hippocampal neurons, while variants without the acidic cluster showed increased interactions with clathrin-associated APs AP-1, AP-2, and AP-3. Using yeast two-hybrid screens, we further discovered that all variants bound dynein light chain Tctex-type 3; however, only variants with an acidic cluster motif bound kinesin light chain 1. Accordingly, splice variants showed markedly different trafficking properties and localized to different subcellular compartments. Taken together, our findings demonstrate the existence of eight functional SorCS2 isoforms with differential capacity for interactions with cytosolic ligands dynein light chain Tctex-type 3 and kinesin light chain 1, which potentially allows cell-type specific SorCS2 trafficking and BDNF signaling.
Collapse
Affiliation(s)
- Sune Skeldal
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Jonas Lende
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Simon Mølgaard
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Perline Demange
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Andreas Høiberg Bentsen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marie Fuglsang
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Henriette Buttenschøn
- NIDO | Centre for Research and Education, Gødstrup Hospital, Herning, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Peder Madsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
11
|
Banerjee R, Gunawardena S. Glycogen synthase kinase 3β (GSK3β) and presenilin (PS) are key regulators of kinesin-1-mediated cargo motility within axons. Front Cell Dev Biol 2023; 11:1202307. [PMID: 37363727 PMCID: PMC10288942 DOI: 10.3389/fcell.2023.1202307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
It has been a quarter century since the discovery that molecular motors are phosphorylated, but fundamental questions still remain as to how specific kinases contribute to particular motor functions, particularly in vivo, and to what extent these processes have been evolutionarily conserved. Such questions remain largely unanswered because there is no cohesive strategy to unravel the likely complex spatial and temporal mechanisms that control motility in vivo. Since diverse cargoes are transported simultaneously within cells and along narrow long neurons to maintain intracellular processes and cell viability, and disruptions in these processes can lead to cancer and neurodegeneration, there is a critical need to better understand how kinases regulate molecular motors. Here, we review our current understanding of how phosphorylation can control kinesin-1 motility and provide evidence for a novel regulatory mechanism that is governed by a specific kinase, glycogen synthase kinase 3β (GSK3β), and a scaffolding protein presenilin (PS).
Collapse
Affiliation(s)
- Rupkatha Banerjee
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
12
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. RESEARCH SQUARE 2023:rs.3.rs-2859584. [PMID: 37292715 PMCID: PMC10246254 DOI: 10.21203/rs.3.rs-2859584/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. Methods We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. Results We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. Conclusion NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
|
13
|
Báez-Flores J, Rodríguez-Martín M, Lacal J. The therapeutic potential of neurofibromin signaling pathways and binding partners. Commun Biol 2023; 6:436. [PMID: 37081086 PMCID: PMC10119308 DOI: 10.1038/s42003-023-04815-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Neurofibromin controls many cell processes, such as growth, learning, and memory. If neurofibromin is not working properly, it can lead to health problems, including issues with the nervous, skeletal, and cardiovascular systems and cancer. This review examines neurofibromin's binding partners, signaling pathways and potential therapeutic targets. In addition, it summarizes the different post-translational modifications that can affect neurofibromin's interactions with other molecules. It is essential to investigate the molecular mechanisms that underlie neurofibromin variants in order to provide with functional connections between neurofibromin and its associated proteins for possible therapeutic targets based on its biological function.
Collapse
Affiliation(s)
- Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
14
|
Qiao S, Jiang Y, Li N, Zhu X. The kinesin light chain-2, a target of mRNA stabilizing protein HuR, inhibits p53 protein phosphorylation to promote radioresistance in NSCLC. Thorac Cancer 2023. [PMID: 37055376 DOI: 10.1111/1759-7714.14886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Radioresistance hinders radiotherapy for the treatment of lung cancer. Kinesin light chain-2 (KLC2) has been found to be upregulated in lung cancer and also to be associated with poor prognosis. This study aimed to investigate the effect of KLC2 on radiosensitivity in lung cancer. METHODS The radioresistant role of KLC2 was determined by colony formation, neutral comet assay, and γH2AX immunofluorescent staining assay. We further verified the function of KLC2 in a xenograft tumor model. The downstream of KLC2 was identified through gene set enrichment analysis and validated by western blot. Finally, we analyzed clinical data from the TCGA database to reveal the upstream transcription factor of KLC2, which was validated by RNA binding protein immunoprecipitation assay. RESULTS Here, we found that downregulation of KLC2 could significantly reduce colony formation, increase γH2AX level, and double-stranded DNA breaks in vitro. Meanwhile, overexpressed KLC2 significantly increased the proportion of the S phase in lung cancer cells. KLC2 knockdown could activate P53 pathway, and ultimately promoting radiosensitivity. The mRNA of KLC2 was observed to bind with Hu-antigen R (HuR). The mRNA and protein expression of KLC2 in lung cancer cells was significantly reduced when combined with siRNA-HuR. Interestingly, KLC2 overexpression significantly increased the expression of HuR in lung cancer cells. CONCLUSION Taken together, these results indicated that HuR-KLC2 forms a positive feedback loop, which decreases the phosphorylation of p53 and thereby weaken the radiosensitivity of lung cancer cells. Our findings highlight the potential prognosis and therapeutic target value of KLC2 in lung cancer patients treated with radiotherapy.
Collapse
Affiliation(s)
- Simiao Qiao
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhang Jiang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Na Li
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Zhu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Gicking AM, Ma TC, Feng Q, Jiang R, Badieyan S, Cianfrocco MA, Hancock WO. Kinesin-1, -2, and -3 motors use family-specific mechanochemical strategies to effectively compete with dynein during bidirectional transport. eLife 2022; 11:e82228. [PMID: 36125250 PMCID: PMC9545524 DOI: 10.7554/elife.82228] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/19/2022] [Indexed: 12/03/2022] Open
Abstract
Bidirectional cargo transport in neurons requires competing activity of motors from the kinesin-1, -2, and -3 superfamilies against cytoplasmic dynein-1. Previous studies demonstrated that when kinesin-1 attached to dynein-dynactin-BicD2 (DDB) complex, the tethered motors move slowly with a slight plus-end bias, suggesting kinesin-1 overpowers DDB but DDB generates a substantial hindering load. Compared to kinesin-1, motors from the kinesin-2 and -3 families display a higher sensitivity to load in single-molecule assays and are thus predicted to be overpowered by dynein complexes in cargo transport. To test this prediction, we used a DNA scaffold to pair DDB with members of the kinesin-1, -2, and -3 families to recreate bidirectional transport in vitro, and tracked the motor pairs using two-channel TIRF microscopy. Unexpectedly, we find that when both kinesin and dynein are engaged and stepping on the microtubule, kinesin-1, -2, and -3 motors are able to effectively withstand hindering loads generated by DDB. Stochastic stepping simulations reveal that kinesin-2 and -3 motors compensate for their faster detachment rates under load with faster reattachment kinetics. The similar performance between the three kinesin transport families highlights how motor kinetics play critical roles in balancing forces between kinesin and dynein, and emphasizes the importance of motor regulation by cargo adaptors, regulatory proteins, and the microtubule track for tuning the speed and directionality of cargo transport in cells.
Collapse
Affiliation(s)
- Allison M Gicking
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Tzu-Chen Ma
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Qingzhou Feng
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Rui Jiang
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Somayesadat Badieyan
- Department of Biological Chemistry and the Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Michael A Cianfrocco
- Department of Biological Chemistry and the Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
16
|
Weible AP, Wehr M. Amyloid Pathology in the Central Auditory Pathway of 5XFAD Mice Appears First in Auditory Cortex. J Alzheimers Dis 2022; 89:1385-1402. [PMID: 36031901 PMCID: PMC10097438 DOI: 10.3233/jad-220538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Effective treatment of Alzheimer’s disease (AD) will hinge on early detection. This has led to the search for early biomarkers that use non-invasive testing. One possible early biomarker is auditory temporal processing deficits, which reflect central auditory pathway dysfunction and precede cognitive and memory declines in AD. Gap detection is a measure of auditory temporal processing, is impaired in human AD, and is also impaired in the 5XFAD mouse model of AD. Gap detection deficits appear as early as postnatal day 60 in 5XFAD mice, months before cognitive deficits or cell death, supporting gap detection as an early biomarker. However, it remains unclear how gap detection deficits relate to the progression of amyloid pathology in the auditory system. Objective: To determine the progression of amyloid pathology throughout the central auditory system and across age in 5XFAD mice. Methods: We quantified intracellular and extracellular antibody labelling of Aβ 42 in 6 regions of the central auditory system from p14 to p150. Results: Pathology appeared first in primary auditory cortex (A1) as intracellular accumulation of Aβ 42 in layer 5 pyramidal neurons by age p21. Extracellular plaques appeared later, by age p90, in A1, medial geniculate body, and inferior colliculus. Auditory brainstem structures showed minimal amyloid pathology. We also observed pathology in the caudal pontine reticular nucleus, a brainstem structure that is outside of the central auditory pathway but which is involved in the acoustic startle reflex. Conclusion: These results suggest that Aβ 42 accumulation, but not plaques, may impair gap detection.
Collapse
Affiliation(s)
- Aldis P. Weible
- Department of Psychology, Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Michael Wehr
- Department of Psychology, Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| |
Collapse
|
17
|
Kumar PP, Bawani SS, Anandhi DU, Prashanth KVH. Rotenone mediated developmental toxicity in Drosophila melanogaster. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 93:103892. [PMID: 35654372 DOI: 10.1016/j.etap.2022.103892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/21/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Rotenone (ROT) is a widely used natural pesticide, and its effect on growth and developmental toxicity remain unclear. In the present study, the effects of ROT exposure on the reproductive structure and function of the female Drosophila melanogaster and third instar larvae were investigated. ROT exposure on female Drosophila melanogaster resulted in developmental inhibition and ovarian abnormality, which were evident from the disruptive growth of border cells as well as morphological changes in the orientation of nurse cells during the 9th-10th stage of developing egg chamber of in the Drosophila ovary. Other abnormalities, such as, altered developmental gene expression (Osk, Grk, Nos, Bic-d), inhibition in the kinesin motor protein level (KIF-5B), increased caspases activities (Caspase 3, 8, & 9) and apoptosis were also observed. Subsequently, ROT treated larvae exhibited behavioral deficits and delay in developmental time. The above findings demonstrate that the exposure of ROT causes developmental toxicity in Drosophila melanogaster.
Collapse
Affiliation(s)
- P Pramod Kumar
- Functional Biopolymer Lab, Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru 570020, Karnataka, India
| | - Saliya S Bawani
- Reproductive Physiology Unit, Department of Zoology, Bangalore University, 560056 Karnataka, India
| | - Duraiswamy Usha Anandhi
- Reproductive Physiology Unit, Department of Zoology, Bangalore University, 560056 Karnataka, India
| | - K V Harish Prashanth
- Functional Biopolymer Lab, Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru 570020, Karnataka, India.
| |
Collapse
|
18
|
Seki E, Komori T, Arai N. Distribution of amyloid-β precursor protein-immunoreactive axons differs according to the severity of cerebral ischemia in autopsy brains. Neuropathology 2022; 42:269-273. [PMID: 35534990 DOI: 10.1111/neup.12809] [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: 11/04/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022]
Abstract
Amyloid-β precursor protein (APP) immunohistochemistry has been used to detect axonal injury in forensic neuropathology. However, axonal injury caused by cerebral ischemia has not been investigated by APP immunohistochemistry in detail. In particular, it is unknown if there is a correlation between the prognosis of cerebral ischemia and the distribution of axonal injury detected by APP immunohistochemistry. To address this issue, we compared the distribution of APP-immunoreactive axons in autopsy brains including lesions of acute phase of cerebral infarction in the territory of the middle cerebral artery (MCA) or internal carotid artery (ICA) with the degree of severity. The presence or absence of a midline shift was used as an indicator of the severity of cerebral ischemia. We identified a difference in the distribution of APP-immunoreactive axons between cases with and without a midline shift. In both the groups, APP-immunoreactive axons were detected at the margin of the ischemic lesions; however, only in cases with a midline shift, intense APP-immunoreactive axons were also found in areas other than the MCA and ICA territories, including the white matter of the cerebral hemispheres ipsilateral and contralateral to the ischemic lesions. This distribution was different from that of acute global cerebral ischemia cases reported previously. Our results indicate that the distribution of APP-immunoreactive axons differs according to the severity and type of cerebral ischemia, suggesting that the distribution of APP-immunoreactive axons is associated with the prognosis of cerebral ischemia.
Collapse
Affiliation(s)
- Erika Seki
- Laboratory of Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Komori
- Department of Laboratory Medicine and Pathology, Tokyo Metropolitan Neurological Hospital, Tokyo, Japan
| | - Nobutaka Arai
- Laboratory of Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
19
|
Haynes EM, Burnett KH, He J, Jean-Pierre MW, Jarzyna M, Eliceiri KW, Huisken J, Halloran MC. KLC4 shapes axon arbors during development and mediates adult behavior. eLife 2022; 11:74270. [PMID: 36222498 PMCID: PMC9596160 DOI: 10.7554/elife.74270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Development of elaborate and polarized neuronal morphology requires precisely regulated transport of cellular cargos by motor proteins such as kinesin-1. Kinesin-1 has numerous cellular cargos which must be delivered to unique neuronal compartments. The process by which this motor selectively transports and delivers cargo to regulate neuronal morphogenesis is poorly understood, although the cargo-binding kinesin light chain (KLC) subunits contribute to specificity. Our work implicates one such subunit, KLC4, as an essential regulator of axon branching and arborization pattern of sensory neurons during development. Using live imaging approaches in klc4 mutant zebrafish, we show that KLC4 is required for stabilization of nascent axon branches, proper microtubule (MT) dynamics, and endosomal transport. Furthermore, KLC4 is required for proper tiling of peripheral axon arbors: in klc4 mutants, peripheral axons showed abnormal fasciculation, a behavior characteristic of central axons. This result suggests that KLC4 patterns axonal compartments and helps establish molecular differences between central and peripheral axons. Finally, we find that klc4 mutant larva are hypersensitive to touch and adults show anxiety-like behavior in a novel tank test, implicating klc4 as a new gene involved in stress response circuits.
Collapse
Affiliation(s)
- Elizabeth M Haynes
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Center for Quantitative Cell Imaging, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States
| | - Korri H Burnett
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Jiaye He
- Morgridge Institute for ResearchMadisonUnited States,National Innovation Center for Advanced Medical DevicesShenzenChina
| | - Marcel W Jean-Pierre
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Martin Jarzyna
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Kevin W Eliceiri
- Center for Quantitative Cell Imaging, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States
| | - Jan Huisken
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Morgridge Institute for ResearchMadisonUnited States,Department of Biology and Psychology, Georg-August-UniversityGöttingenGermany
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin-MadisonMadisonUnited States,Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
20
|
DeGiorgis JA, Jang M, Bearer EL. The Giant Axon of the Squid: A Simple System for Axonal Transport Studies. Methods Mol Biol 2022; 2431:3-22. [PMID: 35412269 DOI: 10.1007/978-1-0716-1990-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The squid giant axon has a long history of being a superb experimental system in which to investigate a wide range of questions concerning intracellular transport. In this protocol we describe the method used for dissecting the axon to preserve its viability in vitro, and the technique for injecting exogenous materials into the living axon. Now that the squid genome is emerging, and the CRISPR/cas9 system has been successfully applied to knock-out squid genes, the giant axon will resume its place in the scientific pantheon of powerful experimental systems in which to address biological questions pertaining to all eukaryotes.
Collapse
Affiliation(s)
- Joseph A DeGiorgis
- Biology Department, Providence College, Providence, RI, USA
- Marine Biological Laboratory, Woods Hole, MA, USA
- Brown University, Providence, RI, USA
| | | | - Elaine L Bearer
- Marine Biological Laboratory, Woods Hole, MA, USA.
- Brown University, Providence, RI, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
21
|
Song XJ, Zhou HY, Sun YY, Huang HC. Phosphorylation and Glycosylation of Amyloid-β Protein Precursor: The Relationship to Trafficking and Cleavage in Alzheimer's Disease. J Alzheimers Dis 2021; 84:937-957. [PMID: 34602469 DOI: 10.3233/jad-210337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder in the central nervous system, and this disease is characterized by extracellular senile plaques and intracellular neurofibrillary tangles. Amyloid-β (Aβ) peptide is the main constituent of senile plaques, and this peptide is derived from the amyloid-β protein precursor (AβPP) through the successive cleaving by β-site AβPP-cleavage enzyme 1 (BACE1) and γ-secretase. AβPP undergoes the progress of post-translational modifications, such as phosphorylation and glycosylation, which might affect the trafficking and the cleavage of AβPP. In the recent years, about 10 phosphorylation sites of AβPP were identified, and they play complex roles in glycosylation modification and cleavage of AβPP. In this article, we introduced the transport and the cleavage pathways of AβPP, then summarized the phosphorylation and glycosylation sites of AβPP, and further discussed the links and relationship between phosphorylation and glycosylation on the pathways of AβPP trafficking and cleavage in order to provide theoretical basis for AD research.
Collapse
Affiliation(s)
- Xi-Jun Song
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - He-Yan Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - Yu-Ying Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| |
Collapse
|
22
|
Chebli J, Rahmati M, Lashley T, Edeman B, Oldfors A, Zetterberg H, Abramsson A. The localization of amyloid precursor protein to ependymal cilia in vertebrates and its role in ciliogenesis and brain development in zebrafish. Sci Rep 2021; 11:19115. [PMID: 34580355 PMCID: PMC8476544 DOI: 10.1038/s41598-021-98487-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Amyloid precursor protein (APP) is expressed in many tissues in human, mice and in zebrafish. In zebrafish, there are two orthologues, Appa and Appb. Interestingly, some cellular processes associated with APP overlap with cilia-mediated functions. Whereas the localization of APP to primary cilia of in vitro-cultured cells has been reported, we addressed the presence of APP in motile and in non-motile sensory cilia and its potential implication for ciliogenesis using zebrafish, mouse, and human samples. We report that Appa and Appb are expressed by ciliated cells and become localized at the membrane of cilia in the olfactory epithelium, otic vesicle and in the brain ventricles of zebrafish embryos. App in ependymal cilia persisted in adult zebrafish and was also detected in mouse and human brain. Finally, we found morphologically abnormal ependymal cilia and smaller brain ventricles in appa−/−appb−/− mutant zebrafish. Our findings demonstrate an evolutionary conserved localisation of APP to cilia and suggest a role of App in ciliogenesis and cilia-related functions.
Collapse
Affiliation(s)
- Jasmine Chebli
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Maryam Rahmati
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden
| | - Tammaryn Lashley
- Department of Clinical and Movement Neurosciences, Queen Square Brain Bank for Neurological Disorders, Queen Square Institute of Neurology, University College London, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Brigitta Edeman
- Department of Laboratory Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anders Oldfors
- Department of Laboratory Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute, London, UK
| | - Alexandra Abramsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 41345, Gothenburg, Sweden.
| |
Collapse
|
23
|
Toupenet Marchesi L, Leblanc M, Stevanin G. Current Knowledge of Endolysosomal and Autophagy Defects in Hereditary Spastic Paraplegia. Cells 2021; 10:cells10071678. [PMID: 34359848 PMCID: PMC8307360 DOI: 10.3390/cells10071678] [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: 05/07/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) refers to a group of neurological disorders involving the degeneration of motor neurons. Due to their clinical and genetic heterogeneity, finding common effective therapeutics is difficult. Therefore, a better understanding of the common pathological mechanisms is necessary. The role of several HSP genes/proteins is linked to the endolysosomal and autophagic pathways, suggesting a functional convergence. Furthermore, impairment of these pathways is particularly interesting since it has been linked to other neurodegenerative diseases, which would suggest that the nervous system is particularly sensitive to the disruption of the endolysosomal and autophagic systems. In this review, we will summarize the involvement of HSP proteins in the endolysosomal and autophagic pathways in order to clarify their functioning and decipher some of the pathological mechanisms leading to HSP.
Collapse
Affiliation(s)
- Liriopé Toupenet Marchesi
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
| | - Marion Leblanc
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
| | - Giovanni Stevanin
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
- Correspondence:
| |
Collapse
|
24
|
Richards A, Berth SH, Brady S, Morfini G. Engagement of Neurotropic Viruses in Fast Axonal Transport: Mechanisms, Potential Role of Host Kinases and Implications for Neuronal Dysfunction. Front Cell Neurosci 2021; 15:684762. [PMID: 34234649 PMCID: PMC8255969 DOI: 10.3389/fncel.2021.684762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/17/2021] [Indexed: 11/28/2022] Open
Abstract
Much remains unknown about mechanisms sustaining the various stages in the life cycle of neurotropic viruses. An understanding of those mechanisms operating before their replication and propagation could advance the development of effective anti-viral strategies. Here, we review our current knowledge of strategies used by neurotropic viruses to undergo bidirectional movement along axons. We discuss how the invasion strategies used by specific viruses might influence their mode of interaction with selected components of the host’s fast axonal transport (FAT) machinery, including specialized membrane-bounded organelles and microtubule-based motor proteins. As part of this discussion, we provide a critical evaluation of various reported interactions among viral and motor proteins and highlight limitations of some in vitro approaches that led to their identification. Based on a large body of evidence documenting activation of host kinases by neurotropic viruses, and on recent work revealing regulation of FAT through phosphorylation-based mechanisms, we posit a potential role of host kinases on the engagement of viruses in retrograde FAT. Finally, we briefly describe recent evidence linking aberrant activation of kinase pathways to deficits in FAT and neuronal degeneration in the context of human neurodegenerative diseases. Based on these findings, we speculate that neurotoxicity elicited by viral infection may involve deregulation of host kinases involved in the regulation of FAT and other cellular processes sustaining neuronal function and survival.
Collapse
Affiliation(s)
- Alexsia Richards
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States
| | - Sarah H Berth
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Scott Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
25
|
Aiken J, Holzbaur ELF. Cytoskeletal regulation guides neuronal trafficking to effectively supply the synapse. Curr Biol 2021; 31:R633-R650. [PMID: 34033795 DOI: 10.1016/j.cub.2021.02.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The development and proper function of the brain requires the formation of highly complex neuronal circuitry. These circuits are shaped from synaptic connections between neurons and must be maintained over a lifetime. The formation and continued maintenance of synapses requires accurate trafficking of presynaptic and postsynaptic components along the axon and dendrite, respectively, necessitating deliberate and specialized delivery strategies to replenish essential synaptic components. Maintenance of synaptic transmission also requires readily accessible energy stores, produced in part by localized mitochondria, that are tightly regulated with activity level. In this review, we focus on recent developments in our understanding of the cytoskeletal environment of axons and dendrites, examining how local regulation of cytoskeletal dynamics and organelle trafficking promotes synapse-specific delivery and plasticity. These new insights shed light on the complex and coordinated role that cytoskeletal elements play in establishing and maintaining neuronal circuitry.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Mutations and Protein Interaction Landscape Reveal Key Cellular Events Perturbed in Upper Motor Neurons with HSP and PLS. Brain Sci 2021; 11:brainsci11050578. [PMID: 33947096 PMCID: PMC8146506 DOI: 10.3390/brainsci11050578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) and primary lateral sclerosis (PLS) are rare motor neuron diseases, which affect mostly the upper motor neurons (UMNs) in patients. The UMNs display early vulnerability and progressive degeneration, while other cortical neurons mostly remain functional. Identification of numerous mutations either directly linked or associated with HSP and PLS begins to reveal the genetic component of UMN diseases. Since each of these mutations are identified on genes that code for a protein, and because cellular functions mostly depend on protein-protein interactions, we hypothesized that the mutations detected in patients and the alterations in protein interaction domains would hold the key to unravel the underlying causes of their vulnerability. In an effort to bring a mechanistic insight, we utilized computational analyses to identify interaction partners of proteins and developed the protein-protein interaction landscape with respect to HSP and PLS. Protein-protein interaction domains, upstream regulators and canonical pathways begin to highlight key cellular events. Here we report that proteins involved in maintaining lipid homeostasis and cytoarchitectural dynamics and their interactions are of great importance for UMN health and stability. Their perturbation may result in neuronal vulnerability, and thus maintaining their balance could offer therapeutic interventions.
Collapse
|
27
|
Chen XQ, Das U, Park G, Mobley WC. Normal levels of KIF5 but reduced KLC1 levels in both Alzheimer disease and Alzheimer disease in Down syndrome: evidence suggesting defects in anterograde transport. Alzheimers Res Ther 2021; 13:59. [PMID: 33691783 PMCID: PMC7945332 DOI: 10.1186/s13195-021-00796-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/22/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Impaired axonal transport may contribute to the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD) and Down syndrome (DS). Axonal transport is a complex process in which specific motor proteins move cargoes to and from neuronal cell bodies and their processes. Inconsistent reports point to the changes in AD in the levels of the classical anterograde motor protein kinesin family member 5 (KIF5) and the primary neuronal KIF regulator kinesin light chain 1 (KLC1), raising the possibility that anterograde transport is compromised in AD. METHODS AND MATERIALS To address inconsistencies and determine if the shared pathologies in AD and elderly DS subjects with dementia (AD in DS; AD-DS) extend to the changes in KIF5 and KLC1, we measured the levels of all the three KIF5 family members and KLC1 in the AD and AD-DS frontal cortex and AD temporal cortex and cerebellum in samples taken with a short postmortem interval. To support future studies to explore the cell biological basis for any changes detected, we also examined the levels of these proteins in the brains of young and aged adult mice in the Dp (16)1Yey/+ (Dp16) mouse model of DS and J20 mouse model of AD. RESULTS There were no changes in comparison with controls in KIF5 family members in either the AD or AD-DS samples when normalized to either β-actin or glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Interestingly, however, samples from control brains as well as from AD and AD-DS demonstrated strong positive correlations between the levels of KIF5 family members, suggesting positive co-regulated expression. Importantly, while earlier reports pointed to a negative correlation between the levels of the amyloid precursor protein (APP) and KIF5A levels, we found the opposite to be true in AD-DS; this was especially striking given triplication of the APP gene, with increased APP protein levels. AD and control samples showed positive correlations between fl-hAPP and KIF5 members, but they were less consistent. In contrast to the findings for KIF5, the levels of KLC1 were downregulated in the frontal cortex of both AD and AD-DS brains; interestingly, this change was not seen in the AD temporal cortex or cerebellum. As postmortem interval has a negative effect on the levels of KLC1, but not KIF5 members, we analyzed a subset of samples with a very short postmortem interval (PMI) (≤ 6 h), a PMI that was not significantly correlated with the levels of KLC1 in either AD or AD-DS samples; we confirmed the presence of a statistically significant reduction of KLC1 in AD and AD-DS brains as compared with control brains. Studies comparing Dp16 to its euploid control recapitulated human studies in demonstrating no change in KIF5 levels and a positive correlation between the levels of KIF5 family members. J20 mice also showed normal KIF5 levels. However, unlike the AD and AD-DS frontal cortex, KLC1 levels were not reduced in the brains of Dp16 or J20 mice. CONCLUSION These data point to significant reductions in KLC1 in AD and AD-DS. In so doing, they raise the possibility of compromised KLC1-mediated axonal transport in these conditions, a posit that can now be pursued in model systems in which KLC1 expression is reduced.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California San Diego, La Jolla, CA 92093 USA
| | - Utpal Das
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California San Diego, La Jolla, CA 92093 USA
| | - Gooho Park
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California San Diego, La Jolla, CA 92093 USA
| | - William C. Mobley
- grid.266100.30000 0001 2107 4242Department of Neurosciences, University of California San Diego, La Jolla, CA 92093 USA
| |
Collapse
|
28
|
Twelvetrees AE. The lifecycle of the neuronal microtubule transport machinery. Semin Cell Dev Biol 2020; 107:74-81. [DOI: 10.1016/j.semcdb.2020.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/08/2023]
|
29
|
Bergoug M, Doudeau M, Godin F, Mosrin C, Vallée B, Bénédetti H. Neurofibromin Structure, Functions and Regulation. Cells 2020; 9:cells9112365. [PMID: 33121128 PMCID: PMC7692384 DOI: 10.3390/cells9112365] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Neurofibromin is a large and multifunctional protein encoded by the tumor suppressor gene NF1, mutations of which cause the tumor predisposition syndrome neurofibromatosis type 1 (NF1). Over the last three decades, studies of neurofibromin structure, interacting partners, and functions have shown that it is involved in several cell signaling pathways, including the Ras/MAPK, Akt/mTOR, ROCK/LIMK/cofilin, and cAMP/PKA pathways, and regulates many fundamental cellular processes, such as proliferation and migration, cytoskeletal dynamics, neurite outgrowth, dendritic-spine density, and dopamine levels. The crystallographic structure has been resolved for two of its functional domains, GRD (GAP-related (GTPase-activating protein) domain) and SecPH, and its post-translational modifications studied, showing it to be localized to several cell compartments. These findings have been of particular interest in the identification of many therapeutic targets and in the proposal of various therapeutic strategies to treat the symptoms of NF1. In this review, we provide an overview of the literature on neurofibromin structure, function, interactions, and regulation and highlight the relationships between them.
Collapse
|
30
|
Vasudevan A, Koushika SP. Molecular mechanisms governing axonal transport: a C. elegans perspective. J Neurogenet 2020; 34:282-297. [PMID: 33030066 DOI: 10.1080/01677063.2020.1823385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axonal transport is integral for maintaining neuronal form and function, and defects in axonal transport have been correlated with several neurological diseases, making it a subject of extensive research over the past several years. The anterograde and retrograde transport machineries are crucial for the delivery and distribution of several cytoskeletal elements, growth factors, organelles and other synaptic cargo. Molecular motors and the neuronal cytoskeleton function as effectors for multiple neuronal processes such as axon outgrowth and synapse formation. This review examines the molecular mechanisms governing axonal transport, specifically highlighting the contribution of studies conducted in C. elegans, which has proved to be a tractable model system in which to identify both novel and conserved regulatory mechanisms of axonal transport.
Collapse
Affiliation(s)
- Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
31
|
Lee D, Choi YH, Seo J, Kim JK, Lee SB. Discovery of new epigenomics-based biomarkers and the early diagnosis of neurodegenerative diseases. Ageing Res Rev 2020; 61:101069. [PMID: 32416267 DOI: 10.1016/j.arr.2020.101069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 03/02/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
Abstract
Treatment options for many neurodegenerative diseases are limited due to the lack of early diagnostic procedures that allow timely delivery of therapeutic agents to affected neurons prior to cell death. While notable advances have been made in neurodegenerative disease biomarkers, whether or not the biomarkers discovered to date are useful for early diagnosis remains an open question. Additionally, the reliability of these biomarkers has been disappointing, due in part to the large dissimilarities between the tissues traditionally used to source biomarkers and primarily diseased neurons. In this article, we review the potential viability of atypical epigenetic and/or consequent transcriptional alterations (ETAs) as biomarkers of early-stage neurodegenerative disease, and present our perspectives on the discovery and practical use of such biomarkers in patient-derived neural samples using single-cell level analyses, thereby greatly enhancing the reliability of biomarker application.
Collapse
|
32
|
Fourriere L, Jimenez AJ, Perez F, Boncompain G. The role of microtubules in secretory protein transport. J Cell Sci 2020; 133:133/2/jcs237016. [PMID: 31996399 DOI: 10.1242/jcs.237016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Microtubules are part of the dynamic cytoskeleton network and composed of tubulin dimers. They are the main tracks used in cells to organize organelle positioning and trafficking of cargos. In this Review, we compile recent findings on the involvement of microtubules in anterograde protein transport. First, we highlight the importance of microtubules in organelle positioning. Second, we discuss the involvement of microtubules within different trafficking steps, in particular between the endoplasmic reticulum and the Golgi complex, traffic through the Golgi complex itself and in post-Golgi processes. A large number of studies have assessed the involvement of microtubules in transport of cargo from the Golgi complex to the cell surface. We focus here on the role of kinesin motor proteins and protein interactions in post-Golgi transport, as well as the impact of tubulin post-translational modifications. Last, in light of recent findings, we highlight the role microtubules have in exocytosis, the final step of secretory protein transport, occurring close to focal adhesions.
Collapse
Affiliation(s)
- Lou Fourriere
- Dynamics of Intracellular Organization Laboratory, Institut Curie, PSL Research University, CNRS UMR 144, Sorbonne Université, 75005 Paris, France
| | - Ana Joaquina Jimenez
- Dynamics of Intracellular Organization Laboratory, Institut Curie, PSL Research University, CNRS UMR 144, Sorbonne Université, 75005 Paris, France
| | - Franck Perez
- Dynamics of Intracellular Organization Laboratory, Institut Curie, PSL Research University, CNRS UMR 144, Sorbonne Université, 75005 Paris, France
| | - Gaelle Boncompain
- Dynamics of Intracellular Organization Laboratory, Institut Curie, PSL Research University, CNRS UMR 144, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
33
|
Zhao J, Fok AHK, Fan R, Kwan PY, Chan HL, Lo LHY, Chan YS, Yung WH, Huang J, Lai CSW, Lai KO. Specific depletion of the motor protein KIF5B leads to deficits in dendritic transport, synaptic plasticity and memory. eLife 2020; 9:53456. [PMID: 31961321 PMCID: PMC7028368 DOI: 10.7554/elife.53456] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
The kinesin I family of motor proteins are crucial for axonal transport, but their roles in dendritic transport and postsynaptic function are not well-defined. Gene duplication and subsequent diversification give rise to three homologous kinesin I proteins (KIF5A, KIF5B and KIF5C) in vertebrates, but it is not clear whether and how they exhibit functional specificity. Here we show that knockdown of KIF5A or KIF5B differentially affects excitatory synapses and dendritic transport in hippocampal neurons. The functional specificities of the two kinesins are determined by their diverse carboxyl-termini, where arginine methylation occurs in KIF5B and regulates its function. KIF5B conditional knockout mice exhibit deficits in dendritic spine morphogenesis, synaptic plasticity and memory formation. Our findings provide insights into how expansion of the kinesin I family during evolution leads to diversification and specialization of motor proteins in regulating postsynaptic function. Transporting molecules within a cell becomes a daunting task when the cell is a neuron, with fibers called axons and dendrites that can stretch as long as a meter. Neurons use many different molecules to send messages across the body and store memories in the brain. If the right molecules cannot be delivered along the length of nerve cells, connections to neighboring neurons may decay, which may impair learning and memory. Motor proteins are responsible for transporting molecules within cells. Kinesins are a type of motor protein that typically transports materials from the body of a neuron to the cell’s periphery, including the dendrites, which is where a neuron receives messages from other nerve cells. Each cell has up to 45 different kinesin motors, but it is not known whether each one performs a distinct task or if they have overlapping roles. Now, Zhao, Fok et al. have studied two similar kinesins, called KIF5A and KIF5B, in rodent neurons to determine their roles. First, it was shown that both proteins were found at dendritic spines, which are small outgrowths on dendrites where contact with other cells occurs. Next, KIF5A and KIF5B were depleted, one at a time, from neurons extracted from a brain region called the hippocampus. Removing KIF5B interfered with the formation of dendritic spines, but removing KIF5A did not have an effect. Dendritic spines are essential for learning and memory, so several behavioral tests were conducted on mice that had been genetically modified to express less KIF5B in the forebrain. These tests revealed that the mice performed poorly in tasks that tested their memory recall. This work opens a new area of research studying the specific roles of different kinesin motor proteins in nerve cells. This could have important implications because certain kinesin motor proteins such as KIF5A are known to be defective in some inherited neurodegenerative diseases.
Collapse
Affiliation(s)
- Junjun Zhao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Albert Hiu Ka Fok
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Ruolin Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Pui-Yi Kwan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Hei-Lok Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Louisa Hoi-Ying Lo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Ying-Shing Chan
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Wing-Ho Yung
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jiandong Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
34
|
Mórotz GM, Glennon EB, Greig J, Lau DHW, Bhembre N, Mattedi F, Muschalik N, Noble W, Vagnoni A, Miller CCJ. Kinesin light chain-1 serine-460 phosphorylation is altered in Alzheimer's disease and regulates axonal transport and processing of the amyloid precursor protein. Acta Neuropathol Commun 2019; 7:200. [PMID: 31806024 PMCID: PMC6896704 DOI: 10.1186/s40478-019-0857-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Damage to axonal transport is an early pathogenic event in Alzheimer’s disease. The amyloid precursor protein (APP) is a key axonal transport cargo since disruption to APP transport promotes amyloidogenic processing of APP. Moreover, altered APP processing itself disrupts axonal transport. The mechanisms that regulate axonal transport of APP are therefore directly relevant to Alzheimer’s disease pathogenesis. APP is transported anterogradely through axons on kinesin-1 motors and one route for this transport involves calsyntenin-1, a type-1 membrane spanning protein that acts as a direct ligand for kinesin-1 light chains (KLCs). Thus, loss of calsyntenin-1 disrupts APP axonal transport and promotes amyloidogenic processing of APP. Phosphorylation of KLC1 on serine-460 has been shown to reduce anterograde axonal transport of calsyntenin-1 by inhibiting the KLC1-calsyntenin-1 interaction. Here we demonstrate that in Alzheimer’s disease frontal cortex, KLC1 levels are reduced and the relative levels of KLC1 serine-460 phosphorylation are increased; these changes occur relatively early in the disease process. We also show that a KLC1 serine-460 phosphomimetic mutant inhibits axonal transport of APP in both mammalian neurons in culture and in Drosophila neurons in vivo. Finally, we demonstrate that expression of the KLC1 serine-460 phosphomimetic mutant promotes amyloidogenic processing of APP. Together, these results suggest that increased KLC1 serine-460 phosphorylation contributes to Alzheimer’s disease.
Collapse
|
35
|
Medina CS, Uselman TW, Barto DR, Cháves F, Jacobs RE, Bearer EL. Decoupling the Effects of the Amyloid Precursor Protein From Amyloid-β Plaques on Axonal Transport Dynamics in the Living Brain. Front Cell Neurosci 2019; 13:501. [PMID: 31849608 PMCID: PMC6901799 DOI: 10.3389/fncel.2019.00501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/22/2019] [Indexed: 11/16/2022] Open
Abstract
Amyloid precursor protein (APP) is the precursor to Aβ plaques. The cytoplasmic domain of APP mediates attachment of vesicles to molecular motors for axonal transport. In APP-KO mice, transport of Mn2+ is decreased. In old transgenic mice expressing mutated human (APPSwInd) linked to Familial Alzheimer's Disease, with both expression of APPSwInd and plaques, the rate and destination of Mn2+ axonal transport is altered, as detected by time-lapse manganese-enhanced magnetic resonance imaging (MEMRI) of the brain in living mice. To determine the relative contribution of expression of APPSwInd versus plaque on transport dynamics, we developed a Tet-off system to decouple expression of APPSwInd from plaque, and then studied hippocampal to forebrain transport by MEMRI. Three groups of mice were compared to wild-type (WT): Mice with plaque and APPSwInd expression; mice with plaque but suppression of APPSwInd expression; and mice with APPSwInd suppressed from mating until 2 weeks before imaging with no plaque. MR images were captured before at successive time points after stereotactic injection of Mn2+ (3-5 nL) into CA3 of the hippocampus. Mice were returned to their home cage between imaging sessions so that transport would occur in the awake freely moving animal. Images of multiple mice from the three groups (suppressed or expressed) together with C57/B6J WT were aligned and processed with our automated computational pipeline, and voxel-wise statistical parametric mapping (SPM) performed. At the conclusion of MR imaging, brains were harvested for biochemistry or histopathology. Paired T-tests within-group between time points (p = 0.01 FDR corrected) support the impression that both plaque alone and APPSwInd expression alone alter transport rates and destination of Mn2+ accumulation. Expression of APPSwInd in the absence of plaque or detectable Aβ also resulted in transport defects as well as pathology of hippocampus and medial septum, suggesting two sources of pathology occur in familial Alzheimer's disease, from toxic mutant protein as well as plaque. Alternatively mice with plaque without APPSwInd expression resemble the human condition of sporadic Alzheimer's, and had better transport. Thus, these mice with APPSwInd expression suppressed after plaque formation will be most useful in preclinical trials.
Collapse
Affiliation(s)
- Christopher S. Medina
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Taylor W. Uselman
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Daniel R. Barto
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Frances Cháves
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Russell E. Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- California Institute of Technology, Pasadena, CA, United States
| | - Elaine L. Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
36
|
Abstract
Background Alzheimer’s disease (AD) imposes a heavy burden on society and every family. Therefore, diagnosing AD in advance and discovering new drug targets are crucial, while these could be achieved by identifying AD-related proteins. The time-consuming and money-costing biological experiment makes researchers turn to develop more advanced algorithms to identify AD-related proteins. Results Firstly, we proposed a hypothesis “similar diseases share similar related proteins”. Therefore, five similarity calculation methods are introduced to find out others diseases which are similar to AD. Then, these diseases’ related proteins could be obtained by public data set. Finally, these proteins are features of each disease and could be used to map their similarity to AD. We developed a novel method ‘LRRGD’ which combines Logistic Regression (LR) and Gradient Descent (GD) and borrows the idea of Random Forest (RF). LR is introduced to regress features to similarities. Borrowing the idea of RF, hundreds of LR models have been built by randomly selecting 40 features (proteins) each time. Here, GD is introduced to find out the optimal result. To avoid the drawback of local optimal solution, a good initial value is selected by some known AD-related proteins. Finally, 376 proteins are found to be related to AD. Conclusion Three hundred eight of three hundred seventy-six proteins are the novel proteins. Three case studies are done to prove our method’s effectiveness. These 308 proteins could give researchers a basis to do biological experiments to help treatment and diagnostic AD.
Collapse
Affiliation(s)
- Tianyi Zhao
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yang Hu
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Tianyi Zang
- School of Life Science and Technology, Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
37
|
Guo W, Stoklund Dittlau K, Van Den Bosch L. Axonal transport defects and neurodegeneration: Molecular mechanisms and therapeutic implications. Semin Cell Dev Biol 2019; 99:133-150. [PMID: 31542222 DOI: 10.1016/j.semcdb.2019.07.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/22/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
Because of the extremely polarized morphology, the proper functioning of neurons largely relies on the efficient cargo transport along the axon. Axonal transport defects have been reported in multiple neurodegenerative diseases as an early pathological feature. The discovery of mutations in human genes involved in the transport machinery provide a direct causative relationship between axonal transport defects and neurodegeneration. Here, we summarize the current genetic findings related to axonal transport in neurodegenerative diseases, and we discuss the relationship between axonal transport defects and other pathological changes observed in neurodegeneration. In addition, we summarize the therapeutic approaches targeting the axonal transport machinery in studies of neurodegenerative diseases. Finally, we review the technical advances in tracking axonal transport both in vivo and in vitro.
Collapse
Affiliation(s)
- Wenting Guo
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium; KU Leuven-Stem Cell Institute (SCIL), Leuven, Belgium
| | - Katarina Stoklund Dittlau
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
38
|
Kendrick AA, Dickey AM, Redwine WB, Tran PT, Vaites LP, Dzieciatkowska M, Harper JW, Reck-Peterson SL. Hook3 is a scaffold for the opposite-polarity microtubule-based motors cytoplasmic dynein-1 and KIF1C. J Cell Biol 2019; 218:2982-3001. [PMID: 31320392 PMCID: PMC6719453 DOI: 10.1083/jcb.201812170] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 05/06/2019] [Accepted: 06/21/2019] [Indexed: 01/06/2023] Open
Abstract
The unidirectional and opposite-polarity microtubule-based motors, dynein and kinesin, drive long-distance intracellular cargo transport. Cellular observations suggest that opposite-polarity motors may be coupled. We recently identified an interaction between the cytoplasmic dynein-1 activating adaptor Hook3 and the kinesin-3 KIF1C. Here, using in vitro reconstitutions with purified components, we show that KIF1C and dynein/dynactin can exist in a complex scaffolded by Hook3. Full-length Hook3 binds to and activates dynein/dynactin motility. Hook3 also binds to a short region in the "tail" of KIF1C, but unlike dynein/dynactin, this interaction does not activate KIF1C. Hook3 scaffolding allows dynein to transport KIF1C toward the microtubule minus end, and KIF1C to transport dynein toward the microtubule plus end. In cells, KIF1C can recruit Hook3 to the cell periphery, although the cellular role of the complex containing both motors remains unknown. We propose that Hook3's ability to scaffold dynein/dynactin and KIF1C may regulate bidirectional motility, promote motor recycling, or sequester the pool of available dynein/dynactin activating adaptors.
Collapse
Affiliation(s)
- Agnieszka A Kendrick
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Andrea M Dickey
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - William B Redwine
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Phuoc Tien Tran
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | | | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Samara L Reck-Peterson
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
39
|
Marzo MG, Griswold JM, Ruff KM, Buchmeier RE, Fees CP, Markus SM. Molecular basis for dyneinopathies reveals insight into dynein regulation and dysfunction. eLife 2019; 8:47246. [PMID: 31364990 PMCID: PMC6733598 DOI: 10.7554/elife.47246] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022] Open
Abstract
Cytoplasmic dynein plays critical roles within the developing and mature nervous systems, including effecting nuclear migration, and retrograde transport of various cargos. Unsurprisingly, mutations in dynein are causative of various developmental neuropathies and motor neuron diseases. These ‘dyneinopathies’ define a broad spectrum of diseases with no known correlation between mutation identity and disease state. To circumvent complications associated with dynein studies in human cells, we employed budding yeast as a screening platform to characterize the motility properties of seventeen disease-correlated dynein mutants. Using this system, we determined the molecular basis for several classes of etiologically related diseases. Moreover, by engineering compensatory mutations, we alleviated the mutant phenotypes in two of these cases, one of which we confirmed with recombinant human dynein. In addition to revealing molecular insight into dynein regulation, our data provide additional evidence that the type of disease may in fact be dictated by the degree of dynein dysfunction. Motor proteins maintain order by transporting biomolecules and various structures within living cells. Dynein is one such motor that moves many types of cargoes along tracks called microtubules, which are spread across the cell’s interior. This motor is particularly important in nerve cells, which can be very long and thus depend heavily on motor proteins to ensure cargoes end up where they are needed. This becomes especially apparent in human diseases that arise as a consequence of mutations in the genes that produce components of the dynein motor. It is assumed that these genetic changes simply prevent dynein from working properly, which ultimately affects the health and survival of cells. However, it is currently unknown what specific effect these mutations have on dynein’s role within the cell, and how these changes lead to particular diseases. Marzo et al. have now used dynein from a budding yeast to closely examine 17 mutations in the dynein gene that are associated with developmental and/or motor neuron diseases in humans. For each mutation, various aspects of how dynein moves (e.g. average speed, distance travelled) were measured and quantitatively compared. The results show that the severity of the effect of each mutation can be directly correlated with the type of disease caused by the mutation. In particular, mutations that lead to less severe defects are found in patients that suffer from various motor neuron diseases, while more severe dynein mutations are found in patients with developmental brain disorders. Marzo et al. confirmed the likely structural changes that caused the defects in dynein’s activity in two of the 17 cases, by engineering additional, restorative mutations that lessened the effects of the primary mutation. These findings reveal links between the molecular impact of defects in the dynein gene and human health. They also confirm that budding yeast is a powerful tool for investigating newly discovered dynein mutations that correlate with disease. This study provides a potential system that could be used to screen drugs that might lessen the effects of specific dynein mutations. However, further work is needed to determine how effective this system will be for drug discovery.
Collapse
Affiliation(s)
- Matthew G Marzo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Jacqueline M Griswold
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Kristina M Ruff
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Rachel E Buchmeier
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Colby P Fees
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, United States
| | - Steven M Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| |
Collapse
|
40
|
Amyloid beta-mediated KIF5A deficiency disrupts anterograde axonal mitochondrial movement. Neurobiol Dis 2019; 127:410-418. [PMID: 30923004 DOI: 10.1016/j.nbd.2019.03.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are crucial organelles for neurophysiology and brain mitochondrial defects constitute a characteristic of Alzheimer's disease (AD). Impaired axonal mitochondrial traffic, especially the anterograde axonal mitochondrial transport is a pronouncing mitochondrial defect that underlies synaptic failure in AD-related conditions. However, the detailed molecular mechanisms of such axonal mitochondrial abnormality have not been fully understood. KIF5A is a key isoform of kinesin-1, which is a key molecular machinery in facilitating anterograde axonal mitochondrial transport. In this study, we have determined a downregulation of KIF5A in postmortem AD temporal lobes. Further experiments on amyloid beta (Aβ)-treated primary neuron culture and 5 × FAD mice suggest a close association of Aβ toxicity and KIF5A loss. Downregulation of KIF5A mimics Aβ-induced axonal mitochondrial transport deficits, indicating a potential role of KIF5A deficiency in AD-relevant axonal mitochondrial traffic abnormalities. Importantly, the restoration of KIF5A corrects Aβ-induced impairments in axonal mitochondrial transport, especially the anterograde traffic, with little or no impact on retrograde axonal mitochondrial motility. Our findings suggest a novel KIF5A-associated mechanism conferring Aβ toxicity to axonal mitochondrial deficits. Furthermore, the results implicate a potential therapeutic avenue by protecting KIF5A function for the treatment of AD.
Collapse
|
41
|
Cromberg LE, Saez TMM, Otero MG, Tomasella E, Alloatti M, Damianich A, Pozo Devoto V, Ferrario J, Gelman D, Rubinstein M, Falzone TL. Neuronal
KIF
5b
deletion induces
striatum
‐dependent locomotor impairments and defects in membrane presentation of dopamine D2 receptors. J Neurochem 2019; 149:362-380. [DOI: 10.1111/jnc.14665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/27/2018] [Accepted: 01/11/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Lucas E. Cromberg
- Instituto de Biología Celular y Neurociencias IBCN (CONICET‐UBA) Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Trinidad M. M. Saez
- Instituto de Biología Celular y Neurociencias IBCN (CONICET‐UBA) Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
- Instituto de Biología y Medicina Experimental IBYME (CONICET) Buenos Aires Argentina
| | - María G. Otero
- Instituto de Biología Celular y Neurociencias IBCN (CONICET‐UBA) Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Eugenia Tomasella
- Instituto de Biología y Medicina Experimental IBYME (CONICET) Buenos Aires Argentina
| | - Matías Alloatti
- Instituto de Biología Celular y Neurociencias IBCN (CONICET‐UBA) Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
| | - Ana Damianich
- Instituto de Investigaciones Farmacológicas ININFA, (CONICET‐UBA) Buenos Aires Argentina
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular INGEBI (CONICET) Buenos Aires Argentina
| | - Victorio Pozo Devoto
- Center for Translational Medicine (CTM) International Clinical Research Center St. Anne's University Hospital (ICRC‐FNUSA) Brno Czech Republic
| | - Juan Ferrario
- Instituto de Investigaciones Farmacológicas ININFA, (CONICET‐UBA) Buenos Aires Argentina
| | - Diego Gelman
- Instituto de Biología y Medicina Experimental IBYME (CONICET) Buenos Aires Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular INGEBI (CONICET) Buenos Aires Argentina
- Facultad de Ciencias Exactas y Naturales Universidad de Buenos Aires Buenos Aires Argentina
| | - Tomás L. Falzone
- Instituto de Biología Celular y Neurociencias IBCN (CONICET‐UBA) Facultad de Medicina Universidad de Buenos Aires Buenos Aires Argentina
- Instituto de Biología y Medicina Experimental IBYME (CONICET) Buenos Aires Argentina
| |
Collapse
|
42
|
Kinesin-1 Proteins KIF5A, -5B, and -5C Promote Anterograde Transport of Herpes Simplex Virus Enveloped Virions in Axons. J Virol 2018; 92:JVI.01269-18. [PMID: 30068641 DOI: 10.1128/jvi.01269-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 01/09/2023] Open
Abstract
Herpes simplex virus (HSV) and other alphaherpesviruses must spread from sites of viral latency in sensory ganglia to peripheral tissues, where the viruses can replicate to higher titers before spreading to other hosts. These viruses move in neuronal axons from ganglia to the periphery propelled by kinesin motors moving along microtubules. Two forms of HSV particles undergo this anterograde transport in axons: (i) unenveloped capsids that become enveloped after reaching axon tips and (ii) enveloped virions that are transported within membrane vesicles in axons. Fundamental to understanding this axonal transport is the question of which of many different axonal kinesins convey HSV particles. Knowing which kinesins promote axonal transport would provide clues to the identity of HSV proteins that tether onto kinesins. Prominent among axonal kinesins are the kinesin-1 (KIF5A, -5B, and -5C) and kinesin-3 (e.g., KIF1A and -1B) families. We characterized fluorescent forms of cellular cargo molecules to determine if enveloped HSV particles were present in the vesicles containing these cargos. Kinesin-1 cargo proteins were present in vesicles containing HSV particles, but not kinesin-3 cargos. Fluorescent kinesin-1 protein KIF5C extensively colocalized with HSV particles, while fluorescent kinesin-1 KIF1A did not. Silencing of kinesin-1 proteins KIF5A, -5B, and -5C or light chains KLC1 and KLC2 inhibited the majority of HSV anterograde transport, while silencing of KIF1A had little effect on HSV transport in axons. We concluded that kinesin-1 proteins are important in the anterograde transport of the majority of HSV enveloped virions in neuronal axons and kinesin-3 proteins are less important.IMPORTANCE Herpes simplex virus (HSV) and other alphaherpesviruses, such as varicella-zoster virus, depend upon the capacity to navigate in neuronal axons. To do this, virus particles tether onto dyneins and kinesins that motor along microtubules from axon tips to neuronal cell bodies (retrograde) or from cell bodies to axon tips (anterograde). Following reactivation from latency, alphaherpesviruses absolutely depend upon anterograde transport of virus particles in axons in order to reinfect peripheral tissues and spread to other hosts. Which of the many axonal kinesins transport HSV in axons is not clear. We characterized fluorescent cellular cargo molecules and kinesins to provide evidence that HSV enveloped particles are ferried by kinesin-1 proteins KIF5A, -5B, and -5C and their light chains, KLC1 and KLC2, in axons. Moreover, we obtained evidence that kinesin-1 proteins are functionally important in anterograde transport of HSV virions by silencing these proteins.
Collapse
|
43
|
Recent Advances by In Silico and In Vitro Studies of Amyloid-β 1-42 Fibril Depicted a S-Shape Conformation. Int J Mol Sci 2018; 19:ijms19082415. [PMID: 30115846 PMCID: PMC6121414 DOI: 10.3390/ijms19082415] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022] Open
Abstract
The amyloid-β 1-42 (Aβ1-42) peptide is produced by proteolytic cleavage of the amyloid precursor protein (APP) by sequential reactions that are catalyzed by γ and β secretases. Aβ1-42, together with the Tau protein are two principal hallmarks of Alzheimer's disease (AD) that are related to disease genesis and progression. Aβ1-42 possesses a higher aggregation propensity, and it is able to form fibrils via nucleated fibril formation. To date, there are compounds available that prevent Aβ1-42 aggregation, but none have been successful in clinical trials, possibly because the Aβ1-42 structure and aggregation mechanisms are not thoroughly understood. New molecules have been designed, employing knowledge of the Aβ1-42 structure and are based on preventing or breaking the ionic interactions that have been proposed for formation of the Aβ1-42 fibril U-shaped structure. Recently, a new Aβ1-42 fibril S-shaped structure was reported that, together with its aggregation and catalytic properties, could be helpful in the design of new inhibitor molecules. Therefore, in silico and in vitro methods have been employed to analyze the Aβ1-42 fibril S-shaped structure and its aggregation to obtain more accurate Aβ1-42 oligomerization data for the design and evaluation of new molecules that can prevent the fibrillation process.
Collapse
|
44
|
Tsukamoto M, Chiba K, Sobu Y, Shiraki Y, Okumura Y, Hata S, Kitamura A, Nakaya T, Uchida S, Kinjo M, Taru H, Suzuki T. The cytoplasmic region of the amyloid β-protein precursor (APP) is necessary and sufficient for the enhanced fast velocity of APP transport by kinesin-1. FEBS Lett 2018; 592:2716-2724. [PMID: 30055048 DOI: 10.1002/1873-3468.13204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 11/10/2022]
Abstract
Amyloid β-protein precursor (APP) is transported mainly by kinesin-1 and at a higher velocity than other kinesin-1 cargos, such as Alcadein α (Alcα); this is denoted by the enhanced fast velocity (EFV). Interaction of the APP cytoplasmic region with kinesin-1, which is essential for EFV transport, is mediated by JNK-interacting protein 1 (JIP1). To determine the roles of interactions between the APP luminal region and cargo components, we monitored transport of chimeric cargo receptors, Alcα (luminal)-APP (cytoplasmic) and APP (luminal)-Alcα (cytoplasmic). Alcα-APP is transported at the EFV, whereas APP-Alcα is transported at the same velocity as wild-type Alcα. Thus, the cytoplasmic region of APP is necessary and sufficient for the EFV of APP transport by kinesin-1.
Collapse
Affiliation(s)
- Maoko Tsukamoto
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Kyoko Chiba
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuzuha Shiraki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yuka Okumura
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Akira Kitamura
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Tadashi Nakaya
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Seiichi Uchida
- Human interface Laboratory, Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Hidenori Taru
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
45
|
Karasmanis EP, Phan CT, Angelis D, Kesisova IA, Hoogenraad CC, McKenney RJ, Spiliotis ET. Polarity of Neuronal Membrane Traffic Requires Sorting of Kinesin Motor Cargo during Entry into Dendrites by a Microtubule-Associated Septin. Dev Cell 2018; 46:204-218.e7. [PMID: 30016622 DOI: 10.1016/j.devcel.2018.06.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/04/2018] [Accepted: 06/15/2018] [Indexed: 01/22/2023]
Abstract
Neuronal function requires axon-dendrite membrane polarity, which depends on sorting of membrane traffic during entry into axons. Due to a microtubule network of mixed polarity, dendrites receive vesicles from the cell body without apparent capacity for directional sorting. We found that, during entry into dendrites, axonally destined cargos move with a retrograde bias toward the cell body, while dendritically destined cargos are biased in the anterograde direction. A microtubule-associated septin (SEPT9), which localizes specifically in dendrites, impedes axonal cargo of kinesin-1/KIF5 and boosts kinesin-3/KIF1 motor cargo further into dendrites. In neurons and in vitro single-molecule motility assays, SEPT9 suppresses kinesin-1/KIF5 and enhances kinesin-3/KIF1 in a manner that depends on a lysine-rich loop of the kinesin motor domain. This differential regulation impacts partitioning of neuronal membrane proteins into axons-dendrites. Thus, polarized membrane traffic requires sorting during entry into dendrites by a septin-mediated mechanism that bestows directional bias on microtubules of mixed orientation.
Collapse
Affiliation(s)
- Eva P Karasmanis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Cat-Thi Phan
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Dimitrios Angelis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Ilona A Kesisova
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Utrecht University, Utrecht 3584 CH, the Netherlands
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Hares K, Miners JS, Cook AJ, Rice C, Scolding N, Love S, Wilkins A. Overexpression of Kinesin Superfamily Motor Proteins in Alzheimer's Disease. J Alzheimers Dis 2018; 60:1511-1524. [PMID: 29060936 DOI: 10.3233/jad-170094] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Defects in motor protein-mediated neuronal transport mechanisms have been implicated in a number of neurodegenerative disorders but remain relatively little studied in Alzheimer's disease (AD). Our aim in the present study was to assess the expression of the anterograde kinesin superfamily motor proteins KIF5A, KIF1B, and KIF21B, and to examine their relationship to levels of hyperphosphorylated tau, amyloid-β protein precursor (AβPP), and amyloid-β (Aβ) in human brain tissue. We used a combination of qPCR, immunoblotting, and ELISA to perform these analyses in midfrontal cortex from 49 AD and 46 control brains. Expression of KIF5A, KIF1B, and KIF21B at gene and protein level was significantly increased in AD. KIF5A protein expression correlated inversely with the levels of AβPP and soluble Aβ in AD brains. Upregulation of KIFs may be an adaptive response to impaired axonal transport in AD.
Collapse
Affiliation(s)
- Kelly Hares
- MS and Stem Cell Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - James Scott Miners
- Dementia Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Amelia Jane Cook
- MS and Stem Cell Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Claire Rice
- MS and Stem Cell Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Neil Scolding
- MS and Stem Cell Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Seth Love
- Dementia Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Alastair Wilkins
- MS and Stem Cell Group, Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| |
Collapse
|
47
|
Bearer EL, Manifold-Wheeler BC, Medina CS, Gonzales AG, Chaves FL, Jacobs RE. Alterations of functional circuitry in aging brain and the impact of mutated APP expression. Neurobiol Aging 2018; 70:276-290. [PMID: 30055413 DOI: 10.1016/j.neurobiolaging.2018.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a disease of aging that results in cognitive impairment, dementia, and death. Pathognomonic features of AD are amyloid plaques composed of proteolytic fragments of the amyloid precursor protein (APP) and neurofibrillary tangles composed of hyperphosphorylated tau protein. One type of familial AD occurs when mutant forms of APP are inherited. Both APP and tau are components of the microtubule-based axonal transport system, which prompts the hypothesis that axonal transport is disrupted in AD, and that such disruption impacts cognitive function. Transgenic mice expressing mutated forms of APP provide preclinical experimental systems to study AD. Here, we perform manganese-enhanced magnetic resonance imaging to study transport from hippocampus to forebrain in four cohorts of living mice: young and old wild-type and transgenic mice expressing a mutant APP with both Swedish and Indiana mutations (APPSwInd). We find that transport is decreased in normal aging and further altered in aged APPSwInd plaque-bearing mice. These findings support the hypothesis that transport deficits are a component of AD pathology and thus may contribute to cognitive deficits.
Collapse
Affiliation(s)
- Elaine L Bearer
- University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Division of Biology, California Institute of Technology, Pasadena, CA, USA.
| | | | | | - Aaron G Gonzales
- University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Frances L Chaves
- University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Russell E Jacobs
- Division of Biology, California Institute of Technology, Pasadena, CA, USA; Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
48
|
UNC-16/JIP3 and UNC-76/FEZ1 limit the density of mitochondria in C. elegans neurons by maintaining the balance of anterograde and retrograde mitochondrial transport. Sci Rep 2018; 8:8938. [PMID: 29895958 PMCID: PMC5997755 DOI: 10.1038/s41598-018-27211-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/25/2018] [Indexed: 12/23/2022] Open
Abstract
We investigate the role of axonal transport in regulating neuronal mitochondrial density. We show that the density of mitochondria in the touch receptor neuron (TRN) of adult Caenorhabditis elegans is constant. Mitochondrial density and transport are controlled both by the Kinesin heavy chain and the Dynein-Dynactin complex. However, unlike in other models, the presence of mitochondria in C. elegans TRNs depends on a Kinesin light chain as well. Mutants in the three C. elegans miro genes do not alter mitochondrial density in the TRNs. Mutants in the Kinesin-1 associated proteins, UNC-16/JIP3 and UNC-76/FEZ1, show increased mitochondrial density and also have elevated levels of both the Kinesin Heavy and Light Chains in neurons. Genetic analyses suggest that, the increased mitochondrial density at the distal end of the neuronal process in unc-16 and unc-76 depends partly on Dynein. We observe a net anterograde bias in the ratio of anterograde to retrograde mitochondrial flux in the neuronal processes of unc-16 and unc-76, likely due to both increased Kinesin-1 and decreased Dynein in the neuronal processes. Our study shows that UNC-16 and UNC-76 indirectly limit mitochondrial density in the neuronal process by maintaining a balance in anterograde and retrograde mitochondrial axonal transport.
Collapse
|
49
|
Fong LK, Yang MM, Dos Santos Chaves R, Reyna SM, Langness VF, Woodruff G, Roberts EA, Young JE, Goldstein LSB. Full-length amyloid precursor protein regulates lipoprotein metabolism and amyloid-β clearance in human astrocytes. J Biol Chem 2018; 293:11341-11357. [PMID: 29858247 DOI: 10.1074/jbc.ra117.000441] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 05/11/2018] [Indexed: 02/05/2023] Open
Abstract
Mounting evidence suggests that alterations in cholesterol homeostasis are involved in Alzheimer's disease (AD) pathogenesis. Amyloid precursor protein (APP) or multiple fragments generated by proteolytic processing of APP have previously been implicated in the regulation of cholesterol metabolism. However, the physiological function of APP in regulating lipoprotein homeostasis in astrocytes, which are responsible for de novo cholesterol biosynthesis and regulation in the brain, remains unclear. To address this, here we used CRISPR/Cas9 genome editing to generate isogenic APP-knockout (KO) human induced pluripotent stem cells (hiPSCs) and differentiated them into human astrocytes. We found that APP-KO astrocytes have reduced cholesterol and elevated levels of sterol regulatory element-binding protein (SREBP) target gene transcripts and proteins, which were both downstream consequences of reduced lipoprotein endocytosis. To elucidate which APP fragments regulate cholesterol homeostasis and to examine whether familial AD mutations in APP affect lipoprotein metabolism, we analyzed an isogenic allelic series harboring the APP Swedish and APP V717F variants. Only astrocytes homozygous for the APP Swedish (APPSwe/Swe) mutation, which had reduced full-length APP (FL APP) due to increased β-secretase cleavage, recapitulated the APP-KO phenotypes. Astrocytic internalization of β-amyloid (Aβ), another ligand for low-density lipoprotein (LDL) receptors, was also impaired in APP-KO and APPSwe/Swe astrocytes. Finally, impairing cleavage of FL APP through β-secretase inhibition in APPSwe/Swe astrocytes reversed the LDL and Aβ endocytosis defects. In conclusion, FL APP is involved in the endocytosis of LDL receptor ligands and is required for proper cholesterol homeostasis and Aβ clearance in human astrocytes.
Collapse
Affiliation(s)
- Lauren K Fong
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Max M Yang
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Rodrigo Dos Santos Chaves
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Sol M Reyna
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Vanessa F Langness
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Grace Woodruff
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Elizabeth A Roberts
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093
| | - Jessica E Young
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Department of Pathology and Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093; Sanford Consortium for Regenerative Medicine, La Jolla, California 92093; Department of Neurosciences, University of California at San Diego, La Jolla, California 92093.
| |
Collapse
|
50
|
Matsuda S, Senda T. BRI2 as an anti-Alzheimer gene. Med Mol Morphol 2018; 52:1-7. [DOI: 10.1007/s00795-018-0191-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 04/18/2018] [Indexed: 12/16/2022]
|