1
|
Markus SM. Microtubule motors of opposite polarity cooperate rather than compete in cargo transport. Nat Struct Mol Biol 2025; 32:595-597. [PMID: 40133460 PMCID: PMC12061008 DOI: 10.1038/s41594-025-01524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Microtubule-based cargo transport relies on the actions of dynein and kinesins, motors that walk in opposite directions yet act together to ensure appropriate distribution of cargos in cells. Research now provides mechanistic insights into how these seemingly antagonistic motors collaborate, rather than compete, to promote each other’s activities.
Collapse
Affiliation(s)
- Steven M Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
2
|
Wang L, Bu T, Wu X, Gao S, Yun D, Mao B, Li H, Silvestrini B, Li L, Sun F, Cheng CY. Microtubule-Associated Proteins (MAPs) Are Multifunctional Cytoskeletal Proteins in the Testis That Regulate Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:411-431. [PMID: 40301267 DOI: 10.1007/978-3-031-82990-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Microtubule-associated proteins (MAPs) refer to a large superfamily of proteins that bind to microtubules (MTs) structurally, modulating the rapid transition of MTs from a stable state (polymerized) to shrinkage (or catastrophe) via depolymerization through a meta-stable state. Changes of MTs from an assembled structure as linear protofilaments that are a packed/bundled ultrastructure to disassembled subunits of heterodimers of α-/ß-tubulins (or oligomers) can take place in milliseconds within a living cell. These heterodimers can also be rapidly phosphorylated, becoming GTP-bound, or rapidly polymerized into linear protofilaments of MT again. It is such rapid cyclic changes of MTs that support cellular development, growth, and changes in cell shape in response to changes in development or other physiological phenomena, such as the series of cellular events during spermatogenesis, cell divisions, and in response to environmental toxicants to protect cellular life. In this review, we seek to give a concise update and discussion on MAPs. Particularly, we focus on a specific member of the structural MAPs, namely MAP1a, and its interaction with the microtubule affinity regulatory kinases (MARKs, including MARK1, 2, 3, and 4, all are Ser/Thr protein kinases) in particular MARK4, and how these two MAPs work together to regulate MT dynamics in Sertoli cells to support germ cell development. This information should be helpful to investigators who seek to better understand the role of MAPs in testis biology.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical University Zhanjiang City, Guangdong Province, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Gao
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Damin Yun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baiping Mao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Cai XY, Ma SY, Tang MH, Hu L, Wu KD, Zhang Z, Zhang YQ, Lin Y, Patel N, Yang ZC, Mo XM. Atoh1 mediated disturbance of neuronal maturation by perinatal hypoxia induces cognitive deficits. Commun Biol 2024; 7:1121. [PMID: 39261625 PMCID: PMC11390922 DOI: 10.1038/s42003-024-06846-7] [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] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Neurodevelopmental disorders are currently one of the major complications faced by patients with congenital heart disease (CHD). Chronic hypoxia in the prenatal and postnatal preoperative brain may be associated with neurological damage and impaired long-term cognitive function, but the exact mechanisms are unknown. In this study, we find that delayed neuronal migration and impaired synaptic development are attributed to altered Atoh1 under chronic hypoxia. This is due to the fact that excessive Atoh1 facilitates expression of Kif21b, which causes excess in free-state α-tubulin, leading to disrupted microtubule dynamic stability. Furthermore, the delay in neonatal brain maturation induces cognitive disabilities in adult mice. Then, by down-regulating Atoh1 we alleviate the impairment of cell migration and synaptic development, improving the cognitive behavior of mice to some extent. Taken together, our work unveil that Atoh1 may be one of the targets to ameliorate hypoxia-induced neurodevelopmental disabilities and cognitive impairment in CHD.
Collapse
Affiliation(s)
- Xin-Yu Cai
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Si-Yu Ma
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Ming-Hui Tang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Liang Hu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ke-de Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhen Zhang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ya-Qi Zhang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ye Lin
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhao-Cong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xu-Ming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
4
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
5
|
Puri D, Barry BJ, Engle EC. TUBB3 and KIF21A in neurodevelopment and disease. Front Neurosci 2023; 17:1226181. [PMID: 37600020 PMCID: PMC10436312 DOI: 10.3389/fnins.2023.1226181] [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: 05/23/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Neuronal migration and axon growth and guidance require precise control of microtubule dynamics and microtubule-based cargo transport. TUBB3 encodes the neuronal-specific β-tubulin isotype III, TUBB3, a component of neuronal microtubules expressed throughout the life of central and peripheral neurons. Human pathogenic TUBB3 missense variants result in altered TUBB3 function and cause errors either in the growth and guidance of cranial and, to a lesser extent, central axons, or in cortical neuronal migration and organization, and rarely in both. Moreover, human pathogenic missense variants in KIF21A, which encodes an anterograde kinesin motor protein that interacts directly with microtubules, alter KIF21A function and cause errors in cranial axon growth and guidance that can phenocopy TUBB3 variants. Here, we review reported TUBB3 and KIF21A variants, resulting phenotypes, and corresponding functional studies of both wildtype and mutant proteins. We summarize the evidence that, in vitro and in mouse models, loss-of-function and missense variants can alter microtubule dynamics and microtubule-kinesin interactions. Lastly, we highlight additional studies that might contribute to our understanding of the relationship between specific tubulin isotypes and specific kinesin motor proteins in health and disease.
Collapse
Affiliation(s)
- Dharmendra Puri
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Brenda J. Barry
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Elizabeth C. Engle
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Rivera Alvarez J, Asselin L, Tilly P, Benoit R, Batisse C, Richert L, Batisse J, Morlet B, Levet F, Schwaller N, Mély Y, Ruff M, Reymann AC, Godin JD. The kinesin Kif21b regulates radial migration of cortical projection neurons through a non-canonical function on actin cytoskeleton. Cell Rep 2023; 42:112744. [PMID: 37418324 DOI: 10.1016/j.celrep.2023.112744] [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: 12/13/2022] [Revised: 05/18/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023] Open
Abstract
Completion of neuronal migration is critical for brain development. Kif21b is a plus-end-directed kinesin motor protein that promotes intracellular transport and controls microtubule dynamics in neurons. Here we report a physiological function of Kif21b during radial migration of projection neurons in the mouse developing cortex. In vivo analysis in mouse and live imaging on cultured slices demonstrate that Kif21b regulates the radial glia-guided locomotion of newborn neurons independently of its motility on microtubules. We show that Kif21b directly binds and regulates the actin cytoskeleton both in vitro and in vivo in migratory neurons. We establish that Kif21b-mediated regulation of actin cytoskeleton dynamics influences branching and nucleokinesis during neuronal locomotion. Altogether, our results reveal atypical roles of Kif21b on the actin cytoskeleton during migration of cortical projection neurons.
Collapse
Affiliation(s)
- José Rivera Alvarez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Laure Asselin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Roxane Benoit
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Claire Batisse
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Ludovic Richert
- Université de Strasbourg, 67000 Strasbourg, France; Laboratoire de Bioimagerie et Pathologies, Centre National de la Recherche Scientifique, UMR 7021, 67404 Illkirch, France
| | - Julien Batisse
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Bastien Morlet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Florian Levet
- University of Bordeaux, CNRS, UMR 5297, Interdisciplinary Institute for Neuroscience, IINS, 33000 Bordeaux, France; University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, 33600 Pessac, France
| | - Noémie Schwaller
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Yves Mély
- Université de Strasbourg, 67000 Strasbourg, France; Laboratoire de Bioimagerie et Pathologies, Centre National de la Recherche Scientifique, UMR 7021, 67404 Illkirch, France
| | - Marc Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Anne-Cécile Reymann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Juliette D Godin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, 67404 Illkirch, France; Centre National de la Recherche Scientifique, CNRS, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France.
| |
Collapse
|
7
|
Gromova KV, Thies E, Janiesch PC, Lützenkirchen FP, Zhu Y, Stajano D, Dürst CD, Schweizer M, Konietzny A, Mikhaylova M, Gee CE, Kneussel M. The kinesin Kif21b binds myosin Va and mediates changes in actin dynamics underlying homeostatic synaptic downscaling. Cell Rep 2023; 42:112743. [PMID: 37418322 DOI: 10.1016/j.celrep.2023.112743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023] Open
Abstract
Homeostatic synaptic plasticity adjusts the strength of synapses to restrain neuronal activity within a physiological range. Postsynaptic guanylate kinase-associated protein (GKAP) controls the bidirectional synaptic scaling of AMPA receptors (AMPARs); however, mechanisms by which chronic activity triggers cytoskeletal remodeling to downscale synaptic transmission are barely understood. Here, we report that the microtubule-dependent kinesin motor Kif21b binds GKAP and likewise is located in dendritic spines in a myosin Va- and neuronal-activity-dependent manner. Kif21b depletion unexpectedly alters actin dynamics in spines, and adaptation of actin turnover following chronic activity is lost in Kif21b-knockout neurons. Consistent with a role of the kinesin in regulating actin dynamics, Kif21b overexpression promotes actin polymerization. Moreover, Kif21b controls GKAP removal from spines and the decrease of GluA2-containing AMPARs from the neuronal surface, thereby inducing homeostatic synaptic downscaling. Our data highlight a critical role of Kif21b at the synaptic actin cytoskeleton underlying homeostatic scaling of neuronal firing.
Collapse
Affiliation(s)
- Kira V Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Philipp C Janiesch
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Felix P Lützenkirchen
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yipeng Zhu
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Daniele Stajano
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Céline D Dürst
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja Konietzny
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marina Mikhaylova
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, 10099 Berlin, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Hamburg Center of Neuroscience, HCNS, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
8
|
Ganchala D, Pinto-Benito D, Baides E, Ruiz-Palmero I, Grassi D, Arevalo MA. Kif21B mediates the effect of estradiol on the morphological plasticity of mouse hippocampal neurons. Front Mol Neurosci 2023; 16:1143024. [PMID: 37078090 PMCID: PMC10106616 DOI: 10.3389/fnmol.2023.1143024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
IntroductionNeurons are polarized cells, and their ability to change their morphology has a functional implication in the development and plasticity of the nervous system in order to establish new connections. Extracellular factors strongly influence neuronal shape and connectivity. For instance, the developmental actions of estradiol on hippocampal neurons are well characterized, and we have demonstrated in previous studies that Ngn3 mediates these actions. On the other hand, Kif21B regulates microtubule dynamics and carries out retrograde transport of the TrkB/brain-derived neurotrophic factor (BDNF) complex, essential for neuronal development.MethodsIn the present study, we assessed the involvement of kinesin Kif21B in the estradiol-dependent signaling mechanisms to regulate neuritogenesis through cultured mouse hippocampal neurons.ResultsWe show that estradiol treatment increases BDNF expression, and estradiol and BDNF modify neuron morphology through TrkB signaling. Treatment with K252a, a TrkB inhibitor, decreases dendrite branching without affecting axonal length, whereas. Combined with estradiol or BDNF, it blocks their effects on axons but not dendrites. Notably, the downregulation of Kif21B abolishes the actions of estradiol and BDNF in both the axon and dendrites. In addition, Kif21B silencing also decreases Ngn3 expression, and downregulation of Ngn3 blocks the effect of BDNF on neuron morphology.DiscussionThese results suggest that Kif21B is required for the effects of estradiol and BDNF on neuronal morphology, but phosphorylation-mediated activation of TrkB is essential only for axonal growth. Our results show that the Estradiol/BDNF/TrkB/Kif21B/Ngn3 is a new and essential pathway mediating hippocampal neuron development.
Collapse
Affiliation(s)
| | - Daniel Pinto-Benito
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Isabel Ruiz-Palmero
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Proteómica, Instituto Biosanitario de Granada-IBS, Fundación Para la Investigación Biosanitaria de Andalucía Oriental—Alejandro Otero (FIBAO), Antiguo Hospital Universitario San Cecilio, Unidad de Apoyo a la Investigación (UNAI), Granada, Spain
| | - Daniela Grassi
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Madrid, Spain
| | - Maria Angeles Arevalo
- Instituto Cajal (IC), CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Maria Angeles Arevalo,
| |
Collapse
|
9
|
Verhey KJ, Ohi R. Causes, costs and consequences of kinesin motors communicating through the microtubule lattice. J Cell Sci 2023; 136:293511. [PMID: 36866642 PMCID: PMC10022682 DOI: 10.1242/jcs.260735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Microtubules are critical for a variety of important functions in eukaryotic cells. During intracellular trafficking, molecular motor proteins of the kinesin superfamily drive the transport of cellular cargoes by stepping processively along the microtubule surface. Traditionally, the microtubule has been viewed as simply a track for kinesin motility. New work is challenging this classic view by showing that kinesin-1 and kinesin-4 proteins can induce conformational changes in tubulin subunits while they are stepping. These conformational changes appear to propagate along the microtubule such that the kinesins can work allosterically through the lattice to influence other proteins on the same track. Thus, the microtubule is a plastic medium through which motors and other microtubule-associated proteins (MAPs) can communicate. Furthermore, stepping kinesin-1 can damage the microtubule lattice. Damage can be repaired by the incorporation of new tubulin subunits, but too much damage leads to microtubule breakage and disassembly. Thus, the addition and loss of tubulin subunits are not restricted to the ends of the microtubule filament but rather, the lattice itself undergoes continuous repair and remodeling. This work leads to a new understanding of how kinesin motors and their microtubule tracks engage in allosteric interactions that are critical for normal cell physiology.
Collapse
Affiliation(s)
- Kristen J. Verhey
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Author for correspondence ()
| | - Ryoma Ohi
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Sidibe DK, Kulkarni VV, Dong A, Herr JB, Vogel MC, Stempel MH, Maday S. Brain-derived neurotrophic factor stimulates the retrograde pathway for axonal autophagy. J Biol Chem 2022; 298:102673. [PMID: 36336077 PMCID: PMC9768381 DOI: 10.1016/j.jbc.2022.102673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Autophagy is a lysosomal degradation pathway important for neuronal development, function, and survival. How autophagy in axons is regulated by neurotrophins to impact neuronal viability and function is poorly understood. Here, we use live-cell imaging in primary neurons to investigate the regulation of axonal autophagy by the neurotrophin brain-derived neurotrophic factor (BDNF) and elucidate whether autophagosomes carry BDNF-mediated signaling information. We find that BDNF induces autophagic flux in primary neurons by stimulating the retrograde pathway for autophagy in axons. We observed an increase in autophagosome density and retrograde flux in axons, and a corresponding increase in autophagosome density in the soma. However, we find little evidence of autophagosomes comigrating with BDNF. In contrast, BDNF effectively engages its cognate receptor TrkB to undergo retrograde transport in the axon. These compartments, however, are distinct from LC3-positive autophagic organelles in the axon. Together, we find that BDNF stimulates autophagy in the axon, but retrograde autophagosomes do not appear to carry BDNF cargo. Thus, autophagosomes likely do not play a major role in relaying neurotrophic signaling information across the axon in the form of active BDNF/TrkB complexes. Rather, BDNF likely stimulates autophagy as a consequence of BDNF-induced processes that require canonical roles for autophagy in degradation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandra Maday
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
11
|
Hoff KJ, Neumann AJ, Moore JK. The molecular biology of tubulinopathies: Understanding the impact of variants on tubulin structure and microtubule regulation. Front Cell Neurosci 2022; 16:1023267. [PMID: 36406756 PMCID: PMC9666403 DOI: 10.3389/fncel.2022.1023267] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/30/2022] [Indexed: 11/24/2022] Open
Abstract
Heterozygous, missense mutations in both α- and β-tubulin genes have been linked to an array of neurodevelopment disorders, commonly referred to as "tubulinopathies." To date, tubulinopathy mutations have been identified in three β-tubulin isotypes and one α-tubulin isotype. These mutations occur throughout the different genetic domains and protein structures of these tubulin isotypes, and the field is working to address how this molecular-level diversity results in different cellular and tissue-level pathologies. Studies from many groups have focused on elucidating the consequences of individual mutations; however, the field lacks comprehensive models for the molecular etiology of different types of tubulinopathies, presenting a major gap in diagnosis and treatment. This review highlights recent advances in understanding tubulin structural dynamics, the roles microtubule-associated proteins (MAPs) play in microtubule regulation, and how these are inextricably linked. We emphasize the value of investigating interactions between tubulin structures, microtubules, and MAPs to understand and predict the impact of tubulinopathy mutations at the cell and tissue levels. Microtubule regulation is multifaceted and provides a complex set of controls for generating a functional cytoskeleton at the right place and right time during neurodevelopment. Understanding how tubulinopathy mutations disrupt distinct subsets of those controls, and how that ultimately disrupts neurodevelopment, will be important for establishing mechanistic themes among tubulinopathies that may lead to insights in other neurodevelopment disorders and normal neurodevelopment.
Collapse
Affiliation(s)
| | | | - Jeffrey K. Moore
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
12
|
Balabanian L, Lessard DV, Swaminathan K, Yaninska P, Sébastien M, Wang S, Stevens PW, Wiseman PW, Berger CL, Hendricks AG. Tau differentially regulates the transport of early endosomes and lysosomes. Mol Biol Cell 2022; 33:ar128. [PMID: 36129768 PMCID: PMC9634973 DOI: 10.1091/mbc.e22-01-0018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Microtubule-associated proteins (MAPs) modulate the motility of kinesin and dynein along microtubules to control the transport of vesicles and organelles. The neuronal MAP tau inhibits kinesin-dependent transport. Phosphorylation of tau at Tyr-18 by fyn kinase results in weakened inhibition of kinesin-1. We examined the motility of early endosomes and lysosomes in cells expressing wild-type (WT) tau and phosphomimetic Y18E tau. We quantified the effects on motility as a function of the tau expression level. Lysosome motility is strongly inhibited by tau. Y18E tau preferentially inhibits lysosomes in the cell periphery, while centrally located lysosomes are less affected. Early endosomes are more sensitive to tau than lysosomes and are inhibited by both WT and Y18E tau. Our results show that different cargoes have disparate responses to tau, likely governed by the types of kinesin motors driving their transport. In support of this model, kinesin-1 and -3 are strongly inhibited by tau while kinesin-2 and dynein are less affected. In contrast to kinesin-1, we find that kinesin-3 is strongly inhibited by phosphorylated tau.
Collapse
Affiliation(s)
- Linda Balabanian
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Dominique V. Lessard
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405
| | | | - Pamela Yaninska
- Chemistry and Physics, McGill University, Montreal, QC H3A 0E9, Canada
| | - Muriel Sébastien
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Samuel Wang
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Piper W. Stevens
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Paul W. Wiseman
- Chemistry and Physics, McGill University, Montreal, QC H3A 0E9, Canada
| | - Christopher L. Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405
| | - Adam G. Hendricks
- Departments of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada,*Address correspondence to: Adam G. Hendricks ()
| |
Collapse
|
13
|
Radwitz J, Hausrat TJ, Heisler FF, Janiesch PC, Pechmann Y, Rübhausen M, Kneussel M. Tubb3 expression levels are sensitive to neuronal activity changes and determine microtubule growth and kinesin-mediated transport. Cell Mol Life Sci 2022; 79:575. [DOI: 10.1007/s00018-022-04607-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
Abstract
AbstractMicrotubules are dynamic polymers of α/β-tubulin. They regulate cell structure, cell division, cell migration, and intracellular transport. However, functional contributions of individual tubulin isotypes are incompletely understood. The neuron-specific β-tubulin Tubb3 displays highest expression around early postnatal periods characterized by exuberant synaptogenesis. Although Tubb3 mutations are associated with neuronal disease, including abnormal inhibitory transmission and seizure activity in patients, molecular consequences of altered Tubb3 levels are largely unknown. Likewise, it is unclear whether neuronal activity triggers Tubb3 expression changes in neurons. In this study, we initially asked whether chemical protocols to induce long-term potentiation (cLTP) affect microtubule growth and the expression of individual tubulin isotypes. We found that growing microtubules and Tubb3 expression are sensitive to changes in neuronal activity and asked for consequences of Tubb3 downregulation in neurons. Our data revealed that reduced Tubb3 levels accelerated microtubule growth in axons and dendrites. Remarkably, Tubb3 knockdown induced a specific upregulation of Tubb4 gene expression, without changing other tubulin isotypes. We further found that Tubb3 downregulation reduces tubulin polyglutamylation, increases KIF5C motility and boosts the transport of its synaptic cargo N-Cadherin, which is known to regulate synaptogenesis and long-term potentiation. Due to the large number of tubulin isotypes, we developed and applied a computational model based on a Monte Carlo simulation to understand consequences of tubulin expression changes in silico. Together, our data suggest a feedback mechanism with neuronal activity regulating tubulin expression and consequently microtubule dynamics underlying the delivery of synaptic cargoes.
Collapse
|
14
|
Narayanan DL, Rivera Alvarez J, Tilly P, do Rosario MC, Bhat V, Godin JD, Shukla A. Further delineation of KIF21B-related neurodevelopmental disorders. J Hum Genet 2022; 67:729-733. [PMID: 36198761 DOI: 10.1038/s10038-022-01087-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022]
Abstract
Kinesin Family Member 21B (KIF21B) encoded by KIF21B (MIM*608322), belongs to the Kinesin superfamily proteins, which play a key role in microtubule organisation in neuronal dendrites and axons. Recently, heterozygous variants in KIF21B were implicated as the cause of intellectual disability and brain malformations in four unrelated individuals. We report a 9-year-old male with delayed speech, hyperactivity, poor social interaction, and autistic features. A parent-offspring trio exome sequencing identified a novel de novo rare heterozygous variant, NM_001252102.2: c.1513A>C, p.(Ser505Arg) in exon 11 of KIF21B. In vivo functional analysis using in utero electroporation in mouse embryonic cortex revealed that the expression of Ser505Arg KIF21B protein in the cerebral cortex impaired the radial migration of projection neurons, thus confirming the pathogenicity of the variant. Our report further validates pathogenic variants in KIF21B as a cause of neurodevelopmental disorder.
Collapse
Affiliation(s)
- Dhanya Lakshmi Narayanan
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.,DBT/Wellcome Trust India Alliance Early Career Fellow, Manipal, India
| | - José Rivera Alvarez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, France.,Université de Strasbourg, Strasbourg, France
| | - Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, France.,Université de Strasbourg, Strasbourg, France
| | - Michelle C do Rosario
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Vivekananda Bhat
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Juliette D Godin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France. .,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France. .,Institut National de la Santé et de la Recherche Médicale, INSERM, U1258, Illkirch, France. .,Université de Strasbourg, Strasbourg, France.
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
15
|
Yap CC, Winckler B. Spatial regulation of endosomes in growing dendrites. Dev Biol 2022; 486:5-14. [PMID: 35306006 PMCID: PMC10646839 DOI: 10.1016/j.ydbio.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/21/2022] [Accepted: 03/13/2022] [Indexed: 01/19/2023]
Abstract
Many membrane proteins are highly enriched in either dendrites or axons. This non-uniform distribution is a critical feature of neuronal polarity and underlies neuronal function. The molecular mechanisms responsible for polarized distribution of membrane proteins has been studied for some time and many answers have emerged. A less well studied feature of neurons is that organelles are also frequently non-uniformly distributed. For instance, EEA1-positive early endosomes are somatodendritic whereas synaptic vesicles are axonal. In addition, some organelles are present in both axons and dendrites, but not distributed uniformly along the processes. One well known example are lysosomes which are abundant in the soma and proximal dendrite, but sparse in the distal dendrite and the distal axon. The mechanisms that determine the spatial distribution of organelles along dendrites are only starting to be studied. In this review, we will discuss the cell biological mechanisms of how the distribution of diverse sets of endosomes along the proximal-distal axis of dendrites might be regulated. In particular, we will focus on the regulation of bulk homeostatic mechanisms as opposed to local regulation. We posit that immature dendrites regulate organelle motility differently from mature dendrites in order to spatially organize dendrite growth, branching and sculpting.
Collapse
|
16
|
Yap CC, Digilio L, McMahon LP, Wang T, Winckler B. Dynein Is Required for Rab7-Dependent Endosome Maturation, Retrograde Dendritic Transport, and Degradation. J Neurosci 2022; 42:4415-4434. [PMID: 35474277 PMCID: PMC9172292 DOI: 10.1523/jneurosci.2530-21.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/30/2022] [Accepted: 04/15/2022] [Indexed: 11/21/2022] Open
Abstract
In all cell types, endocytosed cargo is transported along a set of endosomal compartments, which are linked maturationally from early endosomes (EEs) via late endosomes (LEs) to lysosomes. Lysosomes are critical for degradation of proteins that enter through endocytic as well as autophagic pathways. Rab7 is the master regulator of early-to-late endosome maturation, motility, and fusion with lysosomes. We previously showed that most degradative lysosomes are localized in the soma and in the first 25 µm of the dendrite and that bulk degradation of dendritic membrane proteins occurs in/near the soma. Dendritic late endosomes therefore move retrogradely in a Rab7-dependent manner for fusion with somatic lysosomes. We now used cultured E18 rat hippocampal neurons of both sexes to determine which microtubule motor is responsible for degradative flux of late endosomes. Based on multiple approaches (inhibiting dynein/dynactin itself or inhibiting dynein recruitment to endosomes by expressing the C-terminus of the Rab7 effector, RILP), we now demonstrate that net retrograde flux of late endosomes in dendrites is supported by dynein. Inhibition of dynein also delays maturation of somatic endosomes, as evidenced by excessive accumulation of Rab7. In addition, degradation of dendritic cargos is inhibited. Our results also suggest that GDP-GTP cycling of Rab7 appears necessary not only for endosomal maturation but also for fusion with lysosomes subsequent to arrival in the soma. In conclusion, Rab7-dependent dynein/dynactin recruitment to dendritic endosomes plays multifaceted roles in dendritic endosome maturation as well as retrograde transport of late endosomes to sustain normal degradative flux.SIGNIFICANCE STATEMENT Lysosomes are critical for degradation of membrane and extracellular proteins that enter through endocytosis. Lysosomes are also the endpoint of autophagy and thus responsible for protein and organelle homeostasis. Endosomal-lysosomal dysfunction is linked to neurodegeneration and aging. We identify roles in dendrites for two proteins with links to human diseases, Rab7 and dynein. Our previous work identified a process that requires directional retrograde transport in dendrites, namely, efficient degradation of short-lived membrane proteins. Based on multiple approaches, we demonstrate that Rab7-dependent recruitment of dynein motors supports net retrograde transport to lysosomes and is needed for endosome maturation. Our data also suggest that GDP-GTP cycling of Rab7 is required for fusion with lysosomes and degradation, subsequent to arrival in the soma.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Lloyd P McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| | - Tuanlao Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian Province 361005, China
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
17
|
Selective motor activation in organelle transport along axons. Nat Rev Mol Cell Biol 2022; 23:699-714. [DOI: 10.1038/s41580-022-00491-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
|
18
|
Fan R, Lai KO. Understanding how kinesin motor proteins regulate postsynaptic function in neuron. FEBS J 2021; 289:2128-2144. [PMID: 34796656 DOI: 10.1111/febs.16285] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023]
Abstract
The Kinesin superfamily proteins (KIFs) are major molecular motors that transport diverse set of cargoes along microtubules to both the axon and dendrite of a neuron. Much of our knowledge about kinesin function is obtained from studies on axonal transport. Emerging evidence reveals how specific kinesin motor proteins carry cargoes to dendrites, including proteins, mRNAs and organelles that are crucial for synapse development and plasticity. In this review, we will summarize the major kinesin motors and their associated cargoes that have been characterized to regulate postsynaptic function in neuron. We will also discuss how specific kinesins are selectively involved in the development of excitatory and inhibitory postsynaptic compartments, their regulation by post-translational modifications (PTM), as well as their roles beyond conventional transport carrier.
Collapse
Affiliation(s)
- Ruolin Fan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Masucci EM, Relich PK, Lakadamyali M, Ostap EM, Holzbaur ELF. Microtubule dynamics influence the retrograde biased motility of kinesin-4 motor teams in neuronal dendrites. Mol Biol Cell 2021; 33:ar52. [PMID: 34705476 PMCID: PMC9265162 DOI: 10.1091/mbc.e21-10-0480] [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] [Indexed: 11/12/2022] Open
Abstract
Microtubules establish the directionality of intracellular transport by kinesins and dynein through polarized assembly, but it remains unclear how directed transport occurs along microtubules organized with mixed polarity. We investigated the ability of the plus end–directed kinesin-4 motor KIF21B to navigate mixed polarity microtubules in mammalian dendrites. Reconstitution assays with recombinant KIF21B and engineered microtubule bundles or extracted neuronal cytoskeletons indicate that nucleotide-independent microtubule-binding regions of KIF21B modulate microtubule dynamics and promote directional switching on antiparallel microtubules. Optogenetic recruitment of KIF21B to organelles in live neurons induces unidirectional transport in axons but bidirectional transport with a net retrograde bias in dendrites. Removal of the secondary microtubule-binding regions of KIF21B or dampening of microtubule dynamics with low concentrations of nocodazole eliminates retrograde bias in live dendrites. Further exploration of the contribution of microtubule dynamics in dendrites to directionality revealed plus end–out microtubules to be more dynamic than plus end–in microtubules, with nocodazole preferentially stabilizing the plus end–out population. We propose a model in which both nucleotide-sensitive and -insensitive microtubule-binding sites of KIF21B motors contribute to the search and selection of stable plus end–in microtubules within the mixed polarity microtubule arrays characteristic of mammalian dendrites to achieve net retrograde movement of KIF21B-bound cargoes.
Collapse
Affiliation(s)
- Erin M Masucci
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Peter K Relich
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Melike Lakadamyali
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - E Michael Ostap
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Erika L F Holzbaur
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
20
|
Motz CT, Kabat V, Saxena T, Bellamkonda RV, Zhu C. Neuromechanobiology: An Expanding Field Driven by the Force of Greater Focus. Adv Healthc Mater 2021; 10:e2100102. [PMID: 34342167 PMCID: PMC8497434 DOI: 10.1002/adhm.202100102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The brain processes information by transmitting signals through highly connected and dynamic networks of neurons. Neurons use specific cellular structures, including axons, dendrites and synapses, and specific molecules, including cell adhesion molecules, ion channels and chemical receptors to form, maintain and communicate among cells in the networks. These cellular and molecular processes take place in environments rich of mechanical cues, thus offering ample opportunities for mechanical regulation of neural development and function. Recent studies have suggested the importance of mechanical cues and their potential regulatory roles in the development and maintenance of these neuronal structures. Also suggested are the importance of mechanical cues and their potential regulatory roles in the interaction and function of molecules mediating the interneuronal communications. In this review, the current understanding is integrated and promising future directions of neuromechanobiology are suggested at the cellular and molecular levels. Several neuronal processes where mechanics likely plays a role are examined and how forces affect ligand binding, conformational change, and signal induction of molecules key to these neuronal processes are indicated, especially at the synapse. The disease relevance of neuromechanobiology as well as therapies and engineering solutions to neurological disorders stemmed from this emergent field of study are also discussed.
Collapse
Affiliation(s)
- Cara T Motz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Victoria Kabat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, NC, 27709, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
21
|
Bodakuntla S, Yuan X, Genova M, Gadadhar S, Leboucher S, Birling M, Klein D, Martini R, Janke C, Magiera MM. Distinct roles of α- and β-tubulin polyglutamylation in controlling axonal transport and in neurodegeneration. EMBO J 2021; 40:e108498. [PMID: 34309047 PMCID: PMC8408597 DOI: 10.15252/embj.2021108498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tubulin polyglutamylation is a post-translational modification of the microtubule cytoskeleton, which is generated by a variety of enzymes with different specificities. The "tubulin code" hypothesis predicts that modifications generated by specific enzymes selectively control microtubule functions. Our recent finding that excessive accumulation of polyglutamylation in neurons causes their degeneration and perturbs axonal transport provides an opportunity for testing this hypothesis. By developing novel mouse models and a new glutamylation-specific antibody, we demonstrate here that the glutamylases TTLL1 and TTLL7 generate unique and distinct glutamylation patterns on neuronal microtubules. We find that under physiological conditions, TTLL1 polyglutamylates α-tubulin, while TTLL7 modifies β-tubulin. TTLL1, but not TTLL7, catalyses the excessive hyperglutamylation found in mice lacking the deglutamylase CCP1. Consequently, deletion of TTLL1, but not of TTLL7, prevents degeneration of Purkinje cells and of myelinated axons in peripheral nerves in these mice. Moreover, loss of TTLL1 leads to increased mitochondria motility in neurons, while loss of TTLL7 has no such effect. By revealing how specific patterns of tubulin glutamylation, generated by distinct enzymes, translate into specific physiological and pathological readouts, we demonstrate the relevance of the tubulin code for homeostasis.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
- Present address:
National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Xidi Yuan
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Mariya Genova
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Sudarshan Gadadhar
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Sophie Leboucher
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Marie‐Christine Birling
- CELPHEDIA, PHENOMINInstitut Clinique de la Souris (ICS), CNRS, INSERMUniversity of StrasbourgIllkirchFrance
| | - Dennis Klein
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Rudolf Martini
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Carsten Janke
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Maria M Magiera
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| |
Collapse
|
22
|
Bourke AM, Schwartz SL, Bowen AB, Kleinjan MS, Winborn CS, Kareemo DJ, Gutnick A, Schwarz TL, Kennedy MJ. zapERtrap: A light-regulated ER release system reveals unexpected neuronal trafficking pathways. J Cell Biol 2021; 220:212461. [PMID: 34241635 PMCID: PMC8276314 DOI: 10.1083/jcb.202103186] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/31/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
Here we introduce zapalog-mediated endoplasmic reticulum trap (zapERtrap), which allows one to use light to precisely trigger forward trafficking of diverse integral membrane proteins from internal secretory organelles to the cell surface with single cell and subcellular spatial resolution. To demonstrate its utility, we use zapERtrap in neurons to dissect where synaptic proteins emerge at the cell surface when processed through central (cell body) or remote (dendrites) secretory pathways. We reveal rapid and direct long-range trafficking of centrally processed proteins deep into the dendritic arbor to synaptic sites. Select proteins were also trafficked to the plasma membrane of the axon initial segment, revealing a novel surface trafficking hotspot. Proteins locally processed through dendritic secretory networks were widely dispersed before surface insertion, challenging assumptions for precise trafficking at remote sites. These experiments provide new insights into compartmentalized secretory trafficking and showcase the tunability and spatiotemporal control of zapERtrap, which will have broad applications for regulating cell signaling and function.
Collapse
Affiliation(s)
- Ashley M Bourke
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Samantha L Schwartz
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Aaron B Bowen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Mason S Kleinjan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Christina S Winborn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| | - Amos Gutnick
- Department of Neurobiology, Harvard Medical School, Boston, MA
| | - Thomas L Schwarz
- Department of Neurobiology, Harvard Medical School, Boston, MA.,F.M. Kirby Neurobiology Center, Children's Hospital, Boston, MA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
23
|
Blasius TL, Yue Y, Prasad R, Liu X, Gennerich A, Verhey KJ. Sequences in the stalk domain regulate auto-inhibition and ciliary tip localization of the immotile kinesin-4 KIF7. J Cell Sci 2021; 134:jcs258464. [PMID: 34114033 PMCID: PMC8277141 DOI: 10.1242/jcs.258464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/27/2021] [Indexed: 12/31/2022] Open
Abstract
The kinesin-4 member KIF7 plays critical roles in Hedgehog signaling in vertebrate cells. KIF7 is an atypical kinesin as it binds to microtubules but is immotile. We demonstrate that, like conventional kinesins, KIF7 is regulated by auto-inhibition, as the full-length protein is inactive for microtubule binding in cells. We identify a segment, the inhibitory coiled coil (inhCC), that is required for auto-inhibition of KIF7, whereas the adjacent regulatory coiled coil (rCC) that contributes to auto-inhibition of the motile kinesin-4s KIF21A and KIF21B is not sufficient for KIF7 auto-inhibition. Disease-associated mutations in the inhCC relieve auto-inhibition and result in strong microtubule binding. Surprisingly, uninhibited KIF7 proteins did not bind preferentially to or track the plus ends of growing microtubules in cells, as suggested by previous in vitro work, but rather bound along cytosolic and axonemal microtubules. Localization to the tip of the primary cilium also required the inhCC, and could be increased by disease-associated mutations regardless of the auto-inhibition state of the protein. These findings suggest that loss of KIF7 auto-inhibition and/or altered cilium tip localization can contribute to the pathogenesis of human disease.
Collapse
Affiliation(s)
- T. Lynne Blasius
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yang Yue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - RaghuRam Prasad
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xinglei Liu
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Arne Gennerich
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Kristen J. Verhey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
24
|
Wang X, Guo G, Zhang J, Aebez N, Liu Z, Liu CF, Ross CA, Smith WW. Mutant-TMEM230-induced neurodegeneration and impaired axonal mitochondrial transport. Hum Mol Genet 2021; 30:1535-1542. [PMID: 34002226 DOI: 10.1093/hmg/ddab128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with movement disorders including resting tremor, rigidity, bradykinesia and postural instability. Recent studies have identified a new PD associated gene, TMEM230 (transmembrane protein 230). However, the pathological roles of TMEM230 and its variants are not fully understood. TMEM230 gene encodes two protein isoforms. Isoform2 is the major protein form (~95%) in human. In this study, we overexpress isoform2 TMEM230 variants (WT or PD-linked *184Wext*5 mutant) or knockdown endogenous protein in cultured SH-5Y5Y cells and mouse primary hippocampus neurons to study their pathological roles. We found that overexpression of WT and mutant TMEM230 or knockdown of endogenous TMEM230-induced neurodegeneration and impaired mitochondria transport at the retrograde direction in axons. Mutant TMEM230 caused more severe neurotoxicity and mitochondrial transport impairment than WT-TMEM230 did. Our results demonstrate that maintaining TMEM230 protein levels is critical for neuron survival and axon transport. These findings suggest that mutant-TMEM230-induced mitochondrial transport impairment could be the early event leading to neurite injury and neurodegeneration in PD development.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China
| | - Gongbo Guo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jinru Zhang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Nicolas Aebez
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhaohui Liu
- Department of Human Anatomy and Cytoneurobiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chun-Feng Liu
- Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanli W Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
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: 32] [Impact Index Per Article: 8.0] [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
|
Hooikaas PJ, Damstra HG, Gros OJ, van Riel WE, Martin M, Smits YT, van Loosdregt J, Kapitein LC, Berger F, Akhmanova A. Kinesin-4 KIF21B limits microtubule growth to allow rapid centrosome polarization in T cells. eLife 2020; 9:62876. [PMID: 33346730 PMCID: PMC7817182 DOI: 10.7554/elife.62876] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022] Open
Abstract
When a T cell and an antigen-presenting cell form an immunological synapse, rapid dynein-driven translocation of the centrosome toward the contact site leads to reorganization of microtubules and associated organelles. Currently, little is known about how the regulation of microtubule dynamics contributes to this process. Here, we show that the knockout of KIF21B, a kinesin-4 linked to autoimmune disorders, causes microtubule overgrowth and perturbs centrosome translocation. KIF21B restricts microtubule length by inducing microtubule pausing typically followed by catastrophe. Catastrophe induction with vinblastine prevented microtubule overgrowth and was sufficient to rescue centrosome polarization in KIF21B-knockout cells. Biophysical simulations showed that a relatively small number of KIF21B molecules can restrict mirotubule length and promote an imbalance of dynein-mediated pulling forces that allows the centrosome to translocate past the nucleus. We conclude that proper control of microtubule length is important for allowing rapid remodeling of the cytoskeleton and efficient T cell polarization. The immune system is composed of many types of cells that can recognize foreign molecules and pathogens so they can eliminate them. When cells in the body become infected with a pathogen, they can process the pathogen’s proteins and present them on their own surface. Specialized immune cells can then recognize infected cells and interact with them, forming an ‘immunological synapse’. These synapses play an important role in immune response: they activate the immune system and allow it to kill harmful cells. To form an immunological synapse, an immune cell must reorganize its internal contents, including an aster-shaped scaffold made of tiny protein tubes called microtubules. The center of this scaffold moves towards the immunological synapse as it forms. This re-orientation of the microtubules towards the immunological synapse is known as 'polarization' and it happens very rapidly, but it is not yet clear how it works. One molecule involved in the polarization process is called KIF21B, a protein that can walk along microtubules, building up at the ends and affecting their growth. Whether KIF21B makes microtubules grow more quickly, or more slowly, is a matter of debate, and the impact microtubule length has on immunological synapse formation is unknown. Here, Hooikaas, Damstra et al. deleted the gene for KIF21B from human immune cells called T cells to find out how it affected their ability to form an immunological synapse. Without KIF21B, the T cells grew microtubules that were longer than normal, and had trouble forming immunological synapses. When the T cells were treated with a drug that stops microtubule growth, their ability to form immunological synapses was restored, suggesting a role for KIF21B. To explore this further, Hooikaas, Damstra et al. replaced the missing KIF21B gene with a gene that coded for a version of the protein that could be seen using microscopy. This revealed that, when KIF21B reaches the ends of microtubules, it stops their growth and triggers their disassembly. Computational modelling showed that cells find it hard to reorient their microtubule scaffolding when the individual tubes are too long. It only takes a small number of KIF21B molecules to shorten the microtubules enough to allow the center of the scaffold to move. Research has linked the KIF21B gene to autoimmune conditions like multiple sclerosis. Microtubules also play an important role in cell division, a critical process driving all types of cancer. Drugs that affect microtubule growth are already available, and a deeper understanding of KIF21B and microtubule regulation in immune cells could help to improve treatments in the future.
Collapse
Affiliation(s)
- Peter Jan Hooikaas
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Hugo Gj Damstra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Oane J Gros
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Wilhelmina E van Riel
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Maud Martin
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Yesper Th Smits
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Florian Berger
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
27
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Mutations in the KIF21B kinesin gene cause neurodevelopmental disorders through imbalanced canonical motor activity. Nat Commun 2020; 11:2441. [PMID: 32415109 PMCID: PMC7229210 DOI: 10.1038/s41467-020-16294-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 04/26/2020] [Indexed: 01/08/2023] Open
Abstract
KIF21B is a kinesin protein that promotes intracellular transport and controls microtubule dynamics. We report three missense variants and one duplication in KIF21B in individuals with neurodevelopmental disorders associated with brain malformations, including corpus callosum agenesis (ACC) and microcephaly. We demonstrate, in vivo, that the expression of KIF21B missense variants specifically recapitulates patients’ neurodevelopmental abnormalities, including microcephaly and reduced intra- and inter-hemispheric connectivity. We establish that missense KIF21B variants impede neuronal migration through attenuation of kinesin autoinhibition leading to aberrant KIF21B motility activity. We also show that the ACC-related KIF21B variant independently perturbs axonal growth and ipsilateral axon branching through two distinct mechanisms, both leading to deregulation of canonical kinesin motor activity. The duplication introduces a premature termination codon leading to nonsense-mediated mRNA decay. Although we demonstrate that Kif21b haploinsufficiency leads to an impaired neuronal positioning, the duplication variant might not be pathogenic. Altogether, our data indicate that impaired KIF21B autoregulation and function play a critical role in the pathogenicity of human neurodevelopmental disorder. Kinesins regulate intracellular transport and microtubule dynamics. Here, the authors show that KIF21B variants in humans associate with corpus callosum agenesis and microcephaly. Using mice and zebrafish, they showed the cellular mechanisms altered by the missense KIF21B variants.
Collapse
|
29
|
Tempes A, Weslawski J, Brzozowska A, Jaworski J. Role of dynein-dynactin complex, kinesins, motor adaptors, and their phosphorylation in dendritogenesis. J Neurochem 2020; 155:10-28. [PMID: 32196676 DOI: 10.1111/jnc.15010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
One of the characteristic features of different classes of neurons that is vital for their proper functioning within neuronal networks is the shape of their dendritic arbors. To properly develop dendritic trees, neurons need to accurately control the intracellular transport of various cellular cargo (e.g., mRNA, proteins, and organelles). Microtubules and motor proteins (e.g., dynein and kinesins) that move along microtubule tracks play an essential role in cargo sorting and transport to the most distal ends of neurons. Equally important are motor adaptors, which may affect motor activity and specify cargo that is transported by the motor. Such transport undergoes very dynamic fine-tuning in response to changes in the extracellular environment and synaptic transmission. Such regulation is achieved by the phosphorylation of motors, motor adaptors, and cargo, among other mechanisms. This review focuses on the contribution of the dynein-dynactin complex, kinesins, their adaptors, and the phosphorylation of these proteins in the formation of dendritic trees by maturing neurons. We primarily review the effects of the motor activity of these proteins in dendrites on dendritogenesis. We also discuss less anticipated mechanisms that contribute to dendrite growth, such as dynein-driven axonal transport and non-motor functions of kinesins.
Collapse
Affiliation(s)
- Aleksandra Tempes
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jan Weslawski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agnieszka Brzozowska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
30
|
Zhao HQ, Dong BL, Zhang M, Dong XH, He Y, Chen SY, Wu B, Yang XJ. Increased KIF21B expression is a potential prognostic biomarker in hepatocellular carcinoma. World J Gastrointest Oncol 2020; 12:276-288. [PMID: 32206178 PMCID: PMC7081114 DOI: 10.4251/wjgo.v12.i3.276] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/24/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The kinesin superfamily protein member KIF21B plays an important role in regulating mitotic progression; however, the function and mechanisms of KIF21B in cancer, particularly in hepatocellular carcinoma (HCC), are unknown. AIM To explore the role of KIF21B in hepatocellular carcinoma and its effect on prognosis after hepatectomy. METHODS First, data on the differential expression of KIF21B in patients with HCC from The Cancer Genome Atlas database was analyzed. Subsequently, the expression levels of KIF21B in HCC cell lines and hepatocytes were detected by reverse transcription-polymerase chain reaction, and its biological effect on BEL-7404 cells was evaluated by KIF21B knockdown. Immunohistochemical analysis was used to validate the differential expression of KIF21B in HCC tissues and adjacent normal tissues from 186 patients with HCC after hepatectomy. The Kaplan-Meier method was used to assess prognosis significance. RESULTS KIF21B expression levels were significantly higher in HCC tissues than in corresponding adjacent normal tissues. The expression levels of KIF21B in four HCC cell lines were higher than that in normal liver cells. Functional experiments showed that KIF21B knockdown remarkably suppressed cell proliferation and induced apoptosis. Moreover, immunohistochemistry results are consistent with The Cancer Genome Atlas analysis, with KIF21B expression levels being increased in HCC tissues compared to adjacent normal tissues. Univariate and multivariate analyses revealed KIF21B as an independent risk factor for overall survival and disease-free survival in patients with HCC after hepatectomy. CONCLUSION Taken together, our results provide evidence that KIF21B plays an important role in HCC progression and may be a potential diagnostic and prognostic marker for HCC.
Collapse
Affiliation(s)
- Hui-Qi Zhao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Bao-Long Dong
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Min Zhang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Xiao-Hua Dong
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Yu He
- School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Shi-Yong Chen
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Gansu Province, China
| | - Biao Wu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750000, Gansu Province, China
| | - Xiao-Jun Yang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
31
|
Bodakuntla S, Schnitzler A, Villablanca C, Gonzalez-Billault C, Bieche I, Janke C, Magiera MM. Tubulin polyglutamylation is a general traffic-control mechanism in hippocampal neurons. J Cell Sci 2020; 133:jcs241802. [PMID: 31932508 DOI: 10.1242/jcs.241802] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/23/2019] [Indexed: 08/31/2023] Open
Abstract
Neurons are highly complex cells that heavily rely on intracellular transport to distribute a range of functionally essential cargoes within the cell. Post-translational modifications of tubulin are emerging as mechanisms for regulating microtubule functions, but their impact on neuronal transport is only marginally understood. Here, we have systematically studied the impact of post-translational polyglutamylation on axonal transport. In cultured hippocampal neurons, deletion of a single deglutamylase, CCP1 (also known as AGTPBP1), is sufficient to induce abnormal accumulation of polyglutamylation, i.e. hyperglutamylation. We next investigated how hyperglutamylation affects axonal transport of a range of functionally different neuronal cargoes: mitochondria, lysosomes, LAMP1 endosomes and BDNF vesicles. Strikingly, we found a reduced motility for all these cargoes, suggesting that polyglutamylation could act as a regulator of cargo transport in neurons. This, together with the recent discovery that hyperglutamylation induces neurodegeneration, makes it likely that perturbed neuronal trafficking could be one of the central molecular causes underlying this novel type of degeneration.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Anne Schnitzler
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
| | - Cristopher Villablanca
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Christian Gonzalez-Billault
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
- Department of Biology, Faculty of Sciences, University of Chile, Santiago 7800003, Chile
| | - Ivan Bieche
- Institut Curie, PSL Research University, Department of Genetics, F-75005 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, F-75005 Paris, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91405 Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, F-91405 Orsay, France
| |
Collapse
|
32
|
Bodakuntla S, Magiera MM, Janke C. Measuring the Impact of Tubulin Posttranslational Modifications on Axonal Transport. Methods Mol Biol 2020; 2101:353-370. [PMID: 31879913 DOI: 10.1007/978-1-0716-0219-5_20] [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: 12/24/2022]
Abstract
Axonal transport is a process essential for neuronal function and survival that takes place on the cellular highways-the microtubules. It requires three major components: the microtubules that serve as tracks for the transport, the motor proteins that drive the movement, and the transported cargoes with their adaptor proteins. Axonal transport could be controlled by tubulin posttranslational modifications, which by decorating specific microtubule tracks could determine the specificity of cargo delivery inside neurons. However, it appears that the effects of tubulin modifications on transport can be rather subtle, and might thus be easily overlooked depending on which parameter of the transport process is analyzed. Here we propose an analysis paradigm that allows detecting rather subtle alterations in neuronal transport, as induced for instance by accumulation of posttranslational polyglutamylation. Analyzing mitochondria movements in axons, we found that neither the average speed nor the distance traveled were affected by hyperglutamylation, but we detected an about 50% reduction of the overall motility, suggesting that polyglutamylation controls the efficiency of mitochondria transport in axons. Our protocol can readily be expanded to the analysis of the impact of other tubulin modifications on the transport of a range of different neuronal cargoes.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Centre Universitaire, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| |
Collapse
|
33
|
Morikawa M, Tanaka Y, Cho HS, Yoshihara M, Hirokawa N. The Molecular Motor KIF21B Mediates Synaptic Plasticity and Fear Extinction by Terminating Rac1 Activation. Cell Rep 2019; 23:3864-3877. [PMID: 29949770 DOI: 10.1016/j.celrep.2018.05.089] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/16/2018] [Accepted: 05/25/2018] [Indexed: 12/15/2022] Open
Abstract
Fear extinction is a component of cognitive flexibility that is relevant for important psychiatric diseases, but its molecular mechanism is still largely elusive. We established mice lacking the kinesin-4 motor KIF21B as a model for fear extinction defects. Postsynaptic NMDAR-dependent long-term depression (LTD) is specifically impaired in knockouts. NMDAR-mediated LTD-causing stimuli induce dynamic association of KIF21B with the Rac1GEF subunit engulfment and cell motility protein 1 (ELMO1), leading to ELMO1 translocation out of dendritic spines and its sequestration in endosomes. This process may essentially terminate transient activation of Rac1, shrink spines, facilitate AMPAR endocytosis, and reduce postsynaptic strength, thereby forming a mechanistic link to LTD expression. Antagonizing ELMO1/Dock Rac1GEF activity by the administration of 4-[3'-(2″-chlorophenyl)-2'-propen-1'-ylidene]-1-phenyl-3,5-pyrazolidinedione (CPYPP) significantly reverses the knockout phenotype. Therefore, we propose that KIF21B-mediated Rac1 inactivation is a key molecular event in NMDAR-dependent LTD expression underlying cognitive flexibility in fear extinction.
Collapse
Affiliation(s)
- Momo Morikawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yosuke Tanaka
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hyun-Soo Cho
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masaharu Yoshihara
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
34
|
Boecker CA, Olenick MA, Gallagher ER, Ward ME, Holzbaur ELF. ToolBox: Live Imaging of intracellular organelle transport in induced pluripotent stem cell-derived neurons. Traffic 2019; 21:138-155. [PMID: 31603614 DOI: 10.1111/tra.12701] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold promise to revolutionize studies of intracellular transport in live human neurons and to shed new light on the role of dysfunctional transport in neurodegenerative disorders. Here, we describe an approach for live imaging of axonal and dendritic transport in iPSC-derived cortical neurons. We use transfection and transient expression of genetically-encoded fluorescent markers to characterize the motility of Rab-positive vesicles, including early, late and recycling endosomes, as well as autophagosomes and mitochondria in iPSC-derived neurons. Comparing transport parameters of these organelles with data from primary rat hippocampal neurons, we uncover remarkable similarities. In addition, we generated lysosomal-associated membrane protein 1 (LAMP1)-enhanced green fluorescent protein (EGFP) knock-in iPSCs and show that knock-in neurons can be used to study the transport of endogenously labeled vesicles, as a parallel approach to the transient overexpression of fluorescently labeled organelle markers.
Collapse
Affiliation(s)
- Clemens Alexander Boecker
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mara A Olenick
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth R Gallagher
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael E Ward
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
35
|
Gromova KV, Muhia M, Rothammer N, Gee CE, Thies E, Schaefer I, Kress S, Kilimann MW, Shevchuk O, Oertner TG, Kneussel M. Neurobeachin and the Kinesin KIF21B Are Critical for Endocytic Recycling of NMDA Receptors and Regulate Social Behavior. Cell Rep 2019; 23:2705-2717. [PMID: 29847800 DOI: 10.1016/j.celrep.2018.04.112] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 02/20/2018] [Accepted: 04/25/2018] [Indexed: 11/24/2022] Open
Abstract
Autism spectrum disorders (ASDs) are associated with mutations affecting synaptic components, including GluN2B-NMDA receptors (NMDARs) and neurobeachin (NBEA). NBEA participates in biosynthetic pathways to regulate synapse receptor targeting, synaptic function, cognition, and social behavior. However, the role of NBEA-mediated transport in specific trafficking routes is unclear. Here, we highlight an additional function for NBEA in the local delivery and surface re-insertion of synaptic receptors in mouse neurons. NBEA dynamically interacts with Rab4-positive recycling endosomes, transiently enters spines in an activity-dependent manner, and regulates GluN2B-NMDAR recycling. Furthermore, we show that the microtubule growth inhibitor kinesin KIF21B constrains NBEA dynamics and is present in the NBEA-recycling endosome-NMDAR complex. Notably, Kif21b knockout decreases NMDAR surface expression and alters social behavior in mice, consistent with reported social deficits in Nbea mutants. The influence of NBEA-KIF21B interactions on GluN2B-NMDAR local recycling may be relevant to mechanisms underlying ASD etiology.
Collapse
Affiliation(s)
- Kira V Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mary Muhia
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irina Schaefer
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabrina Kress
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred W Kilimann
- Department of Molecular Neurobiology, Max-Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Olga Shevchuk
- Cellular Proteomics Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany; Leibniz Institute for Analytical Sciences, ISAS, Dortmund, Germany
| | - Thomas G Oertner
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
36
|
Yang R, Bostick Z, Garbouchian A, Luisi J, Banker G, Bentley M. A novel strategy to visualize vesicle-bound kinesins reveals the diversity of kinesin-mediated transport. Traffic 2019; 20:851-866. [PMID: 31461551 PMCID: PMC7714429 DOI: 10.1111/tra.12692] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023]
Abstract
In mammals, 15 to 20 kinesins are thought to mediate vesicle transport. Little is known about the identity of vesicles moved by each kinesin or the functional significance of such diversity. To characterize the transport mediated by different kinesins, we developed a novel strategy to visualize vesicle-bound kinesins in living cells. We applied this method to cultured neurons and systematically determined the localization and transport parameters of vesicles labeled by different members of the Kinesin-1, -2, and -3 families. We observed vesicle labeling with nearly all kinesins. Only six kinesins bound vesicles that undergo long-range transport in neurons. Of these, three had an axonal bias (KIF5B, KIF5C and KIF13B), two were unbiased (KIF1A and KIF1Bβ), and one transported only in dendrites (KIF13A). Overall, the trafficking of vesicle-bound kinesins to axons or dendrites did not correspond to their motor domain preference, suggesting that on-vesicle regulation is crucial for kinesin targeting. Surprisingly, several kinesins were associated with populations of somatodendritic vesicles that underwent little long-range transport. This assay should be broadly applicable for investigating kinesin function in many cell types.
Collapse
Affiliation(s)
- Rui Yang
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
- Department of Biochemistry, Duke University, Durham, North Carolina
| | - Zoe Bostick
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| | - Alex Garbouchian
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| | - Julie Luisi
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
| | - Gary Banker
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
| | - Marvin Bentley
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
37
|
Hasan A, Afzal M. Gene and environment interplay in cognition: Evidence from twin and molecular studies, future directions and suggestions for effective candidate gene x environment (cGxE) research. Mult Scler Relat Disord 2019; 33:121-130. [PMID: 31185373 DOI: 10.1016/j.msard.2019.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/20/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
Last decade of molecular research in the field of cognitive science has shown that no single approach can give satisfactory results as far as gene hunt is concerned. Cohesive theory of gene-environment interaction seems to be a rational idea for bridging the gap in our knowledge of disorders involving cognitive deficit. It may even be helpful to some extent in resolving issues of missing heritability. We review the current state of play in the area of cognition at genetic and environmental fronts. Evidence of apparent gene-environment (GxE) interactions from various studies has been mentioned with the aim of redirecting the focus of research community towards studying such interactions with the help of sensitive designs and molecular techniques. We re-evaluate candidate gene-environment research in order to emphasize its potential if carried out strategically.
Collapse
Affiliation(s)
- Anam Hasan
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India.
| |
Collapse
|
38
|
Kelliher MT, Saunders HA, Wildonger J. Microtubule control of functional architecture in neurons. Curr Opin Neurobiol 2019; 57:39-45. [PMID: 30738328 DOI: 10.1016/j.conb.2019.01.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/20/2023]
Abstract
Neurons are exquisitely polarized cells whose structure and function relies on microtubules. Microtubules in signal-receiving dendrites and signal-sending axons differ in their organization and microtubule-associated proteins. These differences, coupled with microtubule post-translational modifications, combine to locally regulate intracellular transport, morphology, and function. Recent discoveries provide new insight into the regulation of non-centrosomal microtubule arrays in neurons, the relationship between microtubule acetylation and mechanosensation, and the spatial patterning of microtubules that regulates motor activity and cargo delivery in axons and dendrites. Together, these new studies bring us closer to understanding how microtubule function is locally tuned to match the specialized tasks associated with signal reception and transmission.
Collapse
Affiliation(s)
- Michael T Kelliher
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Harriet Aj Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jill Wildonger
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
39
|
Moya-Alvarado G, Gonzalez A, Stuardo N, Bronfman FC. Brain-Derived Neurotrophic Factor (BDNF) Regulates Rab5-Positive Early Endosomes in Hippocampal Neurons to Induce Dendritic Branching. Front Cell Neurosci 2018; 12:493. [PMID: 30618640 PMCID: PMC6304382 DOI: 10.3389/fncel.2018.00493] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/30/2018] [Indexed: 11/24/2022] Open
Abstract
Neurotrophin receptors use endosomal pathways for signaling in neurons. However, how neurotrophins regulate the endosomal system for proper signaling is unknown. Rabs are monomeric GTPases that act as molecular switches to regulate membrane trafficking by binding a wide range of effectors. Among the Rab GTPases, Rab5 is the key GTPase regulating early endosomes and is the first sorting organelle of endocytosed receptors. The objective of our work was to study the regulation of Rab5-positive endosomes by BDNF at different levels, including dynamic, activity and protein levels in hippocampal neurons. Short-term treatment with BDNF increased the colocalization of TrkB in dendrites and cell bodies, increasing the vesiculation of Rab5-positive endosomes. Consistently, BDNF increased the number and mobility of Rab5 endosomes in dendrites. Cell body fluorescence recovery after photobleaching of Rab-EGFP-expressing neurons suggested increased movement of Rab5 endosomes from dendrites to cell bodies. These results correlated with the BDNF-induced activation of Rab5 in dendrites, followed by increased activation of Rab5 in cell bodies. Long-term treatment of hippocampal neurons with BDNF increased the protein levels of Rab5 and Rab11 in an mTOR-dependent manner. While BDNF regulation of Rab5a levels occurred at both the transcriptional and translational levels, Rab11a levels were regulated at the translational level at the time points analyzed. Finally, expression of a dominant-negative mutant of Rab5 reduced the basal arborization of nontreated neurons, and although BDNF was partially able to rescue the effect of Rab5DN at the level of primary dendrites, BDNF-induced dendritic branching was largely reduced. Our findings indicate that BDNF regulates the Rab5-Rab11 endosomal system at different levels and that these processes are likely required for BDNF-induced dendritic branching.
Collapse
Affiliation(s)
- Guillermo Moya-Alvarado
- Department of Physiology, Faculty of Biological Sciences, Center for Aging and Regeneration (CARE UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andres Gonzalez
- Department of Physiology, Faculty of Biological Sciences, Center for Aging and Regeneration (CARE UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolas Stuardo
- Department of Physiology, Faculty of Biological Sciences, Center for Aging and Regeneration (CARE UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca C Bronfman
- Department of Physiology, Faculty of Biological Sciences, Center for Aging and Regeneration (CARE UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
Kilinc D. The Emerging Role of Mechanics in Synapse Formation and Plasticity. Front Cell Neurosci 2018; 12:483. [PMID: 30574071 PMCID: PMC6291423 DOI: 10.3389/fncel.2018.00483] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 11/27/2018] [Indexed: 12/26/2022] Open
Abstract
The regulation of synaptic strength forms the basis of learning and memory, and is a key factor in understanding neuropathological processes that lead to cognitive decline and dementia. While the mechanical aspects of neuronal development, particularly during axon growth and guidance, have been extensively studied, relatively little is known about the mechanical aspects of synapse formation and plasticity. It is established that a filamentous actin network with complex spatiotemporal behavior controls the dendritic spine shape and size, which is thought to be crucial for activity-dependent synapse plasticity. Accordingly, a number of actin binding proteins have been identified as regulators of synapse plasticity. On the other hand, a number of cell adhesion molecules (CAMs) are found in synapses, some of which form transsynaptic bonds to align the presynaptic active zone (PAZ) with the postsynaptic density (PSD). Considering that these CAMs are key components of cellular mechanotransduction, two critical questions emerge: (i) are synapses mechanically regulated? and (ii) does disrupting the transsynaptic force balance lead to (or exacerbate) synaptic failure? In this mini review article, I will highlight the mechanical aspects of synaptic structures-focusing mainly on cytoskeletal dynamics and CAMs-and discuss potential mechanoregulation of synapses and its relevance to neurodegenerative diseases.
Collapse
Affiliation(s)
- Devrim Kilinc
- INSERM U1167, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
41
|
Magiera MM, Bodakuntla S, Žiak J, Lacomme S, Marques Sousa P, Leboucher S, Hausrat TJ, Bosc C, Andrieux A, Kneussel M, Landry M, Calas A, Balastik M, Janke C. Excessive tubulin polyglutamylation causes neurodegeneration and perturbs neuronal transport. EMBO J 2018; 37:e100440. [PMID: 30420556 PMCID: PMC6276888 DOI: 10.15252/embj.2018100440] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
Posttranslational modifications of tubulin are emerging regulators of microtubule functions. We have shown earlier that upregulated polyglutamylation is linked to rapid degeneration of Purkinje cells in mice with a mutation in the deglutamylating enzyme CCP1. How polyglutamylation leads to degeneration, whether it affects multiple neuron types, or which physiological processes it regulates in healthy neurons has remained unknown. Here, we demonstrate that excessive polyglutamylation induces neurodegeneration in a cell-autonomous manner and can occur in many parts of the central nervous system. Degeneration of selected neurons in CCP1-deficient mice can be fully rescued by simultaneous knockout of the counteracting polyglutamylase TTLL1. Excessive polyglutamylation reduces the efficiency of neuronal transport in cultured hippocampal neurons, suggesting that impaired cargo transport plays an important role in the observed degenerative phenotypes. We thus establish polyglutamylation as a cell-autonomous mechanism for neurodegeneration that might be therapeutically accessible through manipulation of the enzymes that control this posttranslational modification.
Collapse
Affiliation(s)
- Maria M Magiera
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Satish Bodakuntla
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Jakub Žiak
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
- Faculty of Science, Charles University, Prague 2, Czech Republic
| | - Sabrina Lacomme
- Bordeaux Imaging Center, BIC, UMS 3420, Université Bordeaux, Bordeaux, France
| | - Patricia Marques Sousa
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Sophie Leboucher
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Torben J Hausrat
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christophe Bosc
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Matthias Kneussel
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Landry
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - André Calas
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - Martin Balastik
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| |
Collapse
|
42
|
Kinesin Family of Proteins Kif11 and Kif21B Act as Inhibitory Constraints of Excitatory Synaptic Transmission Through Distinct Mechanisms. Sci Rep 2018; 8:17419. [PMID: 30479371 PMCID: PMC6258692 DOI: 10.1038/s41598-018-35634-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/09/2018] [Indexed: 02/03/2023] Open
Abstract
Despite our understanding of the functions of the kinesin family of motor proteins (Kifs) in neurons, their specific roles in neuronal communication are less understood. To address this, by carrying out RNAi-mediated loss of function studies, we assessed the necessity of 18 Kifs in excitatory synaptic transmission in mouse primary hippocampal neurons prepared from both sexes. Our measurements of excitatory post-synaptic currents (EPSCs) have identified 7 Kifs that were found to be not critical and 11 Kifs that are essential for synaptic transmission by impacting either frequency or amplitude or both components of EPSCs. Intriguingly we found that knockdown of mitotic Kif4A and Kif11 and post-mitotic Kif21B resulted in an increase in EPSCs suggesting that they function as inhibitory constraints on synaptic transmission. Furthermore, Kifs (11, 21B, 13B) with distinct effects on synaptic transmission are expressed in the same hippocampal neuron. Mechanistically, unlike Kif21B, Kif11 requires the activity of pre-synaptic NMDARs. In addition, we find that Kif11 knockdown enhanced dendritic arborization, synapse number, expression of synaptic vesicle proteins synaptophysin and active zone protein Piccolo. Moreover, expression of Piccolo constrained Kif11 function in synaptic transmission. Together these results suggest that neurons are able to utilize specific Kifs as tools for calibrating synaptic function. These studies bring novel insights into the biology of Kifs and functioning of neural circuits.
Collapse
|
43
|
Baltussen LL, Negraes PD, Silvestre M, Claxton S, Moeskops M, Christodoulou E, Flynn HR, Snijders AP, Muotri AR, Ultanir SK. Chemical genetic identification of CDKL5 substrates reveals its role in neuronal microtubule dynamics. EMBO J 2018; 37:embj.201899763. [PMID: 30266824 PMCID: PMC6293278 DOI: 10.15252/embj.201899763] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/07/2018] [Accepted: 08/31/2018] [Indexed: 01/23/2023] Open
Abstract
Loss‐of‐function mutations in CDKL5 kinase cause severe neurodevelopmental delay and early‐onset seizures. Identification of CDKL5 substrates is key to understanding its function. Using chemical genetics, we found that CDKL5 phosphorylates three microtubule‐associated proteins: MAP1S, EB2 and ARHGEF2, and determined the phosphorylation sites. Substrate phosphorylations are greatly reduced in CDKL5 knockout mice, verifying these as physiological substrates. In CDKL5 knockout mouse neurons, dendritic microtubules have longer EB3‐labelled plus‐end growth duration and these altered dynamics are rescued by reduction of MAP1S levels through shRNA expression, indicating that CDKL5 regulates microtubule dynamics via phosphorylation of MAP1S. We show that phosphorylation by CDKL5 is required for MAP1S dissociation from microtubules. Additionally, anterograde cargo trafficking is compromised in CDKL5 knockout mouse dendrites. Finally, EB2 phosphorylation is reduced in patient‐derived human neurons. Our results reveal a novel activity‐dependent molecular pathway in dendritic microtubule regulation and suggest a pathological mechanism which may contribute to CDKL5 deficiency disorder.
Collapse
Affiliation(s)
- Lucas L Baltussen
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Priscilla D Negraes
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Margaux Silvestre
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Suzanne Claxton
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Max Moeskops
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | | | - Helen R Flynn
- Proteomics Science Technology Platform, The Francis Crick Institute, London, UK
| | | | - Alysson R Muotri
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA .,Department of Pediatrics/Cellular & Molecular Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, School of Medicine, Rady Children's Hospital San Diego, University of California San Diego, La Jolla, CA, USA
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
44
|
Neuronal network maturation differently affects secretory vesicles and mitochondria transport in axons. Sci Rep 2018; 8:13429. [PMID: 30194421 PMCID: PMC6128875 DOI: 10.1038/s41598-018-31759-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/14/2018] [Indexed: 11/09/2022] Open
Abstract
Studying intracellular dynamics in neurons is crucial to better understand how brain circuits communicate and adapt to environmental changes. In neurons, axonal secretory vesicles underlie various functions from growth during development to plasticity in the mature brain. Similarly, transport of mitochondria, the power plant of the cell, regulates both axonal development and synaptic homeostasis. However, because of their submicrometric size and rapid velocities, studying the kinetics of these organelles in projecting axons in vivo is technically challenging. In parallel, primary neuronal cultures are adapted to study axonal transport but they lack the physiological organization of neuronal networks, which in turn may bias observations. We previously developed a microfluidic platform to reconstruct a physiologically-relevant and functional corticostriatal network in vitro that is compatible with high-resolution videorecording of axonal trafficking. Here, using this system we report progressive changes in axonal transport kinetics of both dense core vesicles and mitochondria that correlate with network development and maturation. Interestingly, axonal flow of both types of organelles change in opposite directions, with rates increasing for vesicles and decreasing for mitochondria. Overall, our observations highlight the need for a better spatiotemporal control for the study of intracellular dynamics in order to avoid misinterpretations and improve reproducibility.
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
|
Yue Y, Blasius TL, Zhang S, Jariwala S, Walker B, Grant BJ, Cochran JC, Verhey KJ. Altered chemomechanical coupling causes impaired motility of the kinesin-4 motors KIF27 and KIF7. J Cell Biol 2018; 217:1319-1334. [PMID: 29351996 PMCID: PMC5881503 DOI: 10.1083/jcb.201708179] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/28/2017] [Accepted: 01/02/2018] [Indexed: 01/08/2023] Open
Abstract
Kinesin-4 motors play important roles in cell division, microtubule organization, and signaling. Understanding how motors perform their functions requires an understanding of their mechanochemical and motility properties. We demonstrate that KIF27 can influence microtubule dynamics, suggesting a conserved function in microtubule organization across the kinesin-4 family. However, kinesin-4 motors display dramatically different motility characteristics: KIF4 and KIF21 motors are fast and processive, KIF7 and its Drosophila melanogaster homologue Costal2 (Cos2) are immotile, and KIF27 is slow and processive. Neither KIF7 nor KIF27 can cooperate for fast processive transport when working in teams. The mechanistic basis of immotile KIF7 behavior arises from an inability to release adenosine diphosphate in response to microtubule binding, whereas slow processive KIF27 behavior arises from a slow adenosine triphosphatase rate and a high affinity for both adenosine triphosphate and microtubules. We suggest that evolutionarily selected sequence differences enable immotile KIF7 and Cos2 motors to function not as transporters but as microtubule-based tethers of signaling complexes.
Collapse
Affiliation(s)
- Yang Yue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - T Lynne Blasius
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Stephanie Zhang
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN
| | - Shashank Jariwala
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Benjamin Walker
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN
| | - Barry J Grant
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Jared C Cochran
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN
| | - Kristen J Verhey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
47
|
Martínez-Noël G, Luck K, Kühnle S, Desbuleux A, Szajner P, Galligan JT, Rodriguez D, Zheng L, Boyland K, Leclere F, Zhong Q, Hill DE, Vidal M, Howley PM. Network Analysis of UBE3A/E6AP-Associated Proteins Provides Connections to Several Distinct Cellular Processes. J Mol Biol 2018; 430:1024-1050. [PMID: 29426014 PMCID: PMC5866790 DOI: 10.1016/j.jmb.2018.01.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 12/18/2022]
Abstract
Perturbations in activity and dosage of the UBE3A ubiquitin-ligase have been linked to Angelman syndrome and autism spectrum disorders. UBE3A was initially identified as the cellular protein hijacked by the human papillomavirus E6 protein to mediate the ubiquitylation of p53, a function critical to the oncogenic potential of these viruses. Although a number of substrates have been identified, the normal cellular functions and pathways affected by UBE3A are largely unknown. Previously, we showed that UBE3A associates with HERC2, NEURL4, and MAPK6/ERK3 in a high-molecular-weight complex of unknown function that we refer to as the HUN complex (HERC2, UBE3A, and NEURL4). In this study, the combination of two complementary proteomic approaches with a rigorous network analysis revealed cellular functions and pathways in which UBE3A and the HUN complex are involved. In addition to finding new UBE3A-associated proteins, such as MCM6, SUGT1, EIF3C, and ASPP2, network analysis revealed that UBE3A-associated proteins are connected to several fundamental cellular processes including translation, DNA replication, intracellular trafficking, and centrosome regulation. Our analysis suggests that UBE3A could be involved in the control and/or integration of these cellular processes, in some cases as a component of the HUN complex, and also provides evidence for crosstalk between the HUN complex and CAMKII interaction networks. This study contributes to a deeper understanding of the cellular functions of UBE3A and its potential role in pathways that may be affected in Angelman syndrome, UBE3A-associated autism spectrum disorders, and human papillomavirus-associated cancers.
Collapse
Affiliation(s)
- Gustavo Martínez-Noël
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Katja Luck
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Simone Kühnle
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alice Desbuleux
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; GIGA-R, University of Liège, Liège 4000, Belgium
| | - Patricia Szajner
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey T Galligan
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Diana Rodriguez
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Leon Zheng
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Boyland
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Flavian Leclere
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Quan Zhong
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Peter M Howley
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
A small-molecule activator of kinesin-1 drives remodeling of the microtubule network. Proc Natl Acad Sci U S A 2017; 114:13738-13743. [PMID: 29229862 DOI: 10.1073/pnas.1715115115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The microtubule motor kinesin-1 interacts via its cargo-binding domain with both microtubules and organelles, and hence plays an important role in controlling organelle transport and microtubule dynamics. In the absence of cargo, kinesin-1 is found in an autoinhibited conformation. The molecular basis of how cargo engagement affects the balance between kinesin-1's active and inactive conformations and roles in microtubule dynamics and organelle transport is not well understood. Here we describe the discovery of kinesore, a small molecule that in vitro inhibits kinesin-1 interactions with short linear peptide motifs found in organelle-specific cargo adaptors, yet activates kinesin-1's function of controlling microtubule dynamics in cells, demonstrating that these functions are mechanistically coupled. We establish a proof-of-concept that a microtubule motor-cargo interface and associated autoregulatory mechanism can be manipulated using a small molecule, and define a target for the modulation of microtubule dynamics.
Collapse
|
49
|
Yap CC, Digilio L, McMahon L, Winckler B. The endosomal neuronal proteins Nsg1/NEEP21 and Nsg2/P19 are itinerant, not resident proteins of dendritic endosomes. Sci Rep 2017; 7:10481. [PMID: 28874679 PMCID: PMC5585371 DOI: 10.1038/s41598-017-07667-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/29/2017] [Indexed: 12/28/2022] Open
Abstract
Membrane traffic critically regulates most aspects of neuronal function. Neurons express many neuronal-specific proteins that regulate membrane traffic, including the poorly understood small transmembrane proteins neural-specific gene 1 and 2 (Nsg1/NEEP21 and Nsg2/P19). Nsg1 has been implicated in regulating endosomal recycling and sorting of several important neuronal receptors. Nsg2 is largely unstudied. At steady-state, Nsg1 and Nsg2 only partially co-localize with known endosomal compartments, and it was suggested that they mark a neuronal-specific endosome. Since Nsg1 localizes to and functions in the dendritic endosome, we set out to discover how Nsg1 and Nsg2 localization to endosomes is regulated in primary rat hippocampal neurons, using quadruple immunolocalization against endogenous proteins, live imaging of dendritic endosomes, and interference approaches against the endosomal regulators Rab5 and Rab7. In contrast to previous conclusions, we now show that Nsg1 and Nsg2 are not resident endosomal proteins, but traffic rapidly from the cell surface to lysosomes and have a half-life of less than two hours. Their partial co-localization with canonical endosomal markers thus reflects their rapid flux towards degradation rather than specific targeting to a singular compartment. These findings will require rethinking of how this class of endosomal proteins regulates trafficking of much longer-lived receptors.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lloyd McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
50
|
Nirschl JJ, Ghiretti AE, Holzbaur ELF. The impact of cytoskeletal organization on the local regulation of neuronal transport. Nat Rev Neurosci 2017; 18:585-597. [PMID: 28855741 DOI: 10.1038/nrn.2017.100] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurons are akin to modern cities in that both are dependent on robust transport mechanisms. Like the best mass transit systems, trafficking in neurons must be tailored to respond to local requirements. Neurons depend on both high-speed, long-distance transport and localized dynamics to correctly deliver cargoes and to tune synaptic responses. Here, we focus on the mechanisms that provide localized regulation of the transport machinery, including the cytoskeleton and molecular motors, to yield compartment-specific trafficking in the axon initial segment, axon terminal, dendrites and spines. The synthesis of these mechanisms provides a sophisticated and responsive transit system for the cell.
Collapse
Affiliation(s)
- Jeffrey J Nirschl
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 638A Clinical Research Building, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Amy E Ghiretti
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 638A Clinical Research Building, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 638A Clinical Research Building, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|