1
|
Qu Y, Lim JJY, An O, Yang H, Toh YC, Chua JJE. FEZ1 participates in human embryonic brain development by modulating neuronal progenitor subpopulation specification and migrations. iScience 2023; 26:108497. [PMID: 38213789 PMCID: PMC10783620 DOI: 10.1016/j.isci.2023.108497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 01/13/2024] Open
Abstract
Mutations in the human fasciculation and elongation protein zeta 1 (FEZ1) gene are found in schizophrenia and Jacobsen syndrome patients. Here, using human cerebral organoids (hCOs), we show that FEZ1 expression is turned on early during brain development and is detectable in both neuroprogenitor subtypes and immature neurons. FEZ1 deletion disrupts expression of neuronal and synaptic development genes. Using single-cell RNA sequencing, we detected abnormal expansion of homeodomain-only protein homeobox (HOPX)- outer radial glia (oRG), concurrent with a reduction of HOPX+ oRG, in FEZ1-null hCOs. HOPX- oRGs show higher cell mobility as compared to HOPX+ oRGs. Ectopic localization of neuroprogenitors to the outer layer is seen in FEZ1-null hCOs. Anomalous encroachment of TBR2+ intermediate progenitors into CTIP2+ deep layer neurons further indicated abnormalities in cortical layer formation these hCOs. Collectively, our findings highlight the involvement of FEZ1 in early cortical brain development and how it contributes to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yinghua Qu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Jonathan Jun-Yong Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Omer An
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
- Institute for Molecular and Cell Biology, A∗STAR, Singapore 138473, Singapore
| |
Collapse
|
2
|
Glomb O, Swaim G, Munoz LLancao P, Lovejoy C, Sutradhar S, Park J, Wu Y, Cason SE, Holzbaur ELF, Hammarlund M, Howard J, Ferguson SM, Gramlich MW, Yogev S. A kinesin-1 adaptor complex controls bimodal slow axonal transport of spectrin in Caenorhabditis elegans. Dev Cell 2023; 58:1847-1863.e12. [PMID: 37751746 PMCID: PMC10574138 DOI: 10.1016/j.devcel.2023.08.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
An actin-spectrin lattice, the membrane periodic skeleton (MPS), protects axons from breakage. MPS integrity relies on spectrin delivery via slow axonal transport, a process that remains poorly understood. We designed a probe to visualize endogenous spectrin dynamics at single-axon resolution in vivo. Surprisingly, spectrin transport is bimodal, comprising fast runs and movements that are 100-fold slower than previously reported. Modeling and genetic analysis suggest that the two rates are independent, yet both require kinesin-1 and the coiled-coil proteins UNC-76/FEZ1 and UNC-69/SCOC, which we identify as spectrin-kinesin adaptors. Knockdown of either protein led to disrupted spectrin motility and reduced distal MPS, and UNC-76 overexpression instructed excessive transport of spectrin. Artificially linking spectrin to kinesin-1 drove robust motility but inefficient MPS assembly, whereas impairing MPS assembly led to excessive spectrin transport, suggesting a balance between transport and assembly. These results provide insight into slow axonal transport and MPS integrity.
Collapse
Affiliation(s)
- Oliver Glomb
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Grace Swaim
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Pablo Munoz LLancao
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christopher Lovejoy
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Junhyun Park
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Youjun Wu
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sydney E Cason
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc Hammarlund
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Quantitative Biology Institute, Yale University, New Haven, CT 06510, USA
| | - Shawn M Ferguson
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Shaul Yogev
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
3
|
Petzoldt AG. Presynaptic Precursor Vesicles-Cargo, Biogenesis, and Kinesin-Based Transport across Species. Cells 2023; 12:2248. [PMID: 37759474 PMCID: PMC10527734 DOI: 10.3390/cells12182248] [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: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The faithful formation and, consequently, function of a synapse requires continuous and tightly controlled delivery of synaptic material. At the presynapse, a variety of proteins with unequal molecular properties are indispensable to compose and control the molecular machinery concerting neurotransmitter release through synaptic vesicle fusion with the presynaptic membrane. As presynaptic proteins are produced mainly in the neuronal soma, they are obliged to traffic along microtubules through the axon to reach the consuming presynapse. This anterograde transport is performed by highly specialised and diverse presynaptic precursor vesicles, membranous organelles able to transport as different proteins such as synaptic vesicle membrane and membrane-associated proteins, cytosolic active zone proteins, ion-channels, and presynaptic membrane proteins, coordinating synaptic vesicle exo- and endocytosis. This review aims to summarise and categorise the diverse and numerous findings describing presynaptic precursor cargo, mode of trafficking, kinesin-based axonal transport and the molecular mechanisms of presynaptic precursor vesicles biogenesis in both vertebrate and invertebrate model systems.
Collapse
Affiliation(s)
- Astrid G Petzoldt
- Institute for Biology and Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| |
Collapse
|
4
|
Razar RBBA, Qu Y, Gunaseelan S, Chua JJE. The importance of fasciculation and elongation protein zeta-1 in neural circuit establishment and neurological disorders. Neural Regen Res 2022; 17:1165-1171. [PMID: 34782550 PMCID: PMC8643053 DOI: 10.4103/1673-5374.327327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 06/20/2021] [Indexed: 11/04/2022] Open
Abstract
The human brain contains an estimated 100 billion neurons that must be systematically organized into functional neural circuits for it to function properly. These circuits range from short-range local signaling networks between neighboring neurons to long-range networks formed between various brain regions. Compelling converging evidence indicates that alterations in neural circuits arising from abnormalities during early neuronal development or neurodegeneration contribute significantly to the etiology of neurological disorders. Supporting this notion, efforts to identify genetic causes of these disorders have uncovered an over-representation of genes encoding proteins involved in the processes of neuronal differentiation, maturation, synaptogenesis and synaptic function. Fasciculation and elongation protein zeta-1, a Kinesin-1 adapter, has emerged as a key central player involved in many of these processes. Fasciculation and elongation protein zeta-1-dependent transport of synaptic cargoes and mitochondria is essential for neuronal development and synapse establishment. Furthermore, it acts downstream of guidance cue pathways to regulate axo-dendritic development. Significantly, perturbing its function causes abnormalities in neuronal development and synapse formation both in the brain as well as the peripheral nervous system. Mutations and deletions of the fasciculation and elongation protein zeta-1 gene are linked to neurodevelopmental disorders. Moreover, altered phosphorylation of the protein contributes to neurodegenerative disorders. Together, these findings strongly implicate the importance of fasciculation and elongation protein zeta-1 in the establishment of neuronal circuits and its maintenance.
Collapse
Affiliation(s)
- Rafhanah Banu Bte Abdul Razar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
| | - Yinghua Qu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Saravanan Gunaseelan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
5
|
Malikov V, Meade N, Simons LM, Hultquist JF, Naghavi MH. FEZ1 phosphorylation regulates HSPA8 localization and interferon-stimulated gene expression. Cell Rep 2022; 38:110396. [PMID: 35172151 PMCID: PMC8900055 DOI: 10.1016/j.celrep.2022.110396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/16/2021] [Accepted: 01/25/2022] [Indexed: 01/06/2023] Open
Abstract
Fasciculation and elongation protein zeta-1 (FEZ1) is a multifunctional kinesin adaptor involved in processes ranging from neurodegeneration to retrovirus and polyomavirus infection. Here, we show that, although modulating FEZ1 expression also impacts infection by large DNA viruses in human microglia, macrophages, and fibroblasts, this broad antiviral phenotype is associated with the pre-induction of interferon-stimulated genes (ISGs) in a STING-independent manner. We further reveal that S58, a key phosphorylation site in FEZ1's kinesin regulatory domain, controls both binding to, and the nuclear-cytoplasmic localization of, heat shock protein 8 (HSPA8), as well as ISG expression. FEZ1- and HSPA8-induced changes in ISG expression further involved changes in DNA-dependent protein kinase (DNA-PK) accumulation in the nucleus. Moreover, phosphorylation of endogenous FEZ1 at S58 was reduced and HSPA8 and DNA-PK translocated to the nucleus in cells stimulated with DNA, suggesting that FEZ1 is a regulatory component of the recently identified HSPA8/DNA-PK innate immune pathway.
Collapse
Affiliation(s)
- Viacheslav Malikov
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nathan Meade
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lacy M Simons
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Judd F Hultquist
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mojgan H Naghavi
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
6
|
Yang C, Wang X, Qiu C, Zheng Z, Lin K, Tu M, Zhang K, Jiang K, Gao W. Identification of FEZ2 as a potential oncogene in pancreatic ductal adenocarcinoma. PeerJ 2022; 9:e12736. [PMID: 35036176 PMCID: PMC8742541 DOI: 10.7717/peerj.12736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/12/2021] [Indexed: 12/02/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the common malignant tumors with high lethal rate and poor prognosis. Dysregulation of many genes have been reported to be involved in the occurrence and development of PDAC. However, as a highly conserved gene in eukaryotes, the role of Fasciculation and Elongation protein Zeta 2 (FEZ2) in pancreatic cancer progression is not clear. In this study, we identified the oncogenic effect of FEZ2 on PDAC. By mining of The Cancer Genome Atlas (TCGA) database, we found that FEZ2 was upregulated in PDAC tissues and FEZ2 expression was negatively regulated by its methylation. Moreover, high expression and low methylation of FEZ2 correlated with poor prognosis in PDAC patients. Besides, we found that FEZ2 could promote PDAC cells proliferation, migration and 5-FU resistance in vitro. Furthermore, Gene pathway enrichment analysis demonstrated a positive correlation between Wnt signaling activation and FEZ2 expression in PDAC patients. Western blot showed that FEZ2 knockdown significantly suppressed β-catenin expression. Collectively, our finding revealed that FEZ2 functioned as a potential oncogene on PDAC progression and migration, and the expression of FEZ2 had guidance value for the treatment and chemotherapy program of PDAC patients.
Collapse
Affiliation(s)
- Chaozhi Yang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xuebing Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chenjie Qiu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ziruo Zheng
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kai Lin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Min Tu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kai Zhang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wentao Gao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
7
|
FEZ1 Forms Complexes with CRMP1 and DCC to Regulate Axon and Dendrite Development. eNeuro 2021; 8:ENEURO.0193-20.2021. [PMID: 33771901 PMCID: PMC8174033 DOI: 10.1523/eneuro.0193-20.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 02/10/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
Elaboration of neuronal processes is an early step in neuronal development. Guidance cues must work closely with intracellular trafficking pathways to direct expanding axons and dendrites to their target neurons during the formation of neuronal networks. However, how such coordination is achieved remains incompletely understood. Here, we characterize an interaction between fasciculation and elongation protein zeta 1 (FEZ1), an adapter involved in synaptic protein transport, and collapsin response mediator protein (CRMP)1, a protein that functions in growth cone guidance, at neuronal growth cones. We show that similar to CRMP1 loss-of-function mutants, FEZ1 deficiency in rat hippocampal neurons causes growth cone collapse and impairs axonal development. Strikingly, FEZ1-deficient neurons also exhibited a reduction in dendritic complexity stronger than that observed in CRMP1-deficient neurons, suggesting that the former could partake in additional developmental signaling pathways. Supporting this, FEZ1 colocalizes with VAMP2 in developing hippocampal neurons and forms a separate complex with deleted in colorectal cancer (DCC) and Syntaxin-1 (Stx1), components of the Netrin-1 signaling pathway that are also involved in regulating axon and dendrite development. Significantly, developing axons and dendrites of FEZ1-deficient neurons fail to respond to Netrin-1 or Netrin-1 and Sema3A treatment, respectively. Taken together, these findings highlight the importance of FEZ1 as a common effector to integrate guidance signaling pathways with intracellular trafficking to mediate axo-dendrite development during neuronal network formation.
Collapse
|
8
|
Gunaseelan S, Wang Z, Tong VKJ, Ming SWS, Razar RBBA, Srimasorn S, Ong WY, Lim KL, Chua JJE. Loss of FEZ1, a gene deleted in Jacobsen syndrome, causes locomotion defects and early mortality by impairing motor neuron development. Hum Mol Genet 2021; 30:5-20. [PMID: 33395696 DOI: 10.1093/hmg/ddaa281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/10/2020] [Accepted: 12/23/2020] [Indexed: 01/05/2023] Open
Abstract
FEZ1-mediated axonal transport plays important roles in central nervous system development but its involvement in the peripheral nervous system is not well-characterized. FEZ1 is deleted in Jacobsen syndrome (JS), an 11q terminal deletion developmental disorder. JS patients display impaired psychomotor skills, including gross and fine motor delay, suggesting that FEZ1 deletion may be responsible for these phenotypes, given its association with the development of motor-related circuits. Supporting this hypothesis, our data show that FEZ1 is selectively expressed in the rat brain and spinal cord. Its levels progressively increase over the developmental course of human motor neurons (MN) derived from embryonic stem cells. Deletion of FEZ1 strongly impaired axon and dendrite development, and significantly delayed the transport of synaptic proteins into developing neurites. Concurring with these observations, Drosophila unc-76 mutants showed severe locomotion impairments, accompanied by a strong reduction of synaptic boutons at neuromuscular junctions. These abnormalities were ameliorated by pharmacological activation of UNC-51/ATG1, a FEZ1-activating kinase, with rapamycin and metformin. Collectively, the results highlight a role for FEZ1 in MN development and implicate its deletion as an underlying cause of motor impairments in JS patients.
Collapse
Affiliation(s)
- Saravanan Gunaseelan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ziyin Wang
- National Neuroscience Institute, Singapore, Singapore
| | - Venetia Kok Jing Tong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National Neuroscience Institute, Singapore, Singapore
| | - Sylvester Wong Shu Ming
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Sumitra Srimasorn
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kah-Leong Lim
- National Neuroscience Institute, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - John Jia En Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,LSI Neurobiology Programme, National University of Singapore, Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
9
|
Li W, Zhang S, Yang G. Dynamic organization of intracellular organelle networks. WIREs Mech Dis 2020; 13:e1505. [PMID: 32865347 DOI: 10.1002/wsbm.1505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/06/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Intracellular organelles are membrane-bound and biochemically distinct compartments constructed to serve specialized functions in eukaryotic cells. Through extensive interactions, they form networks to coordinate and integrate their specialized functions for cell physiology. A fundamental property of these organelle networks is that they constantly undergo dynamic organization via membrane fusion and fission to remodel their internal connections and to mediate direct material exchange between compartments. The dynamic organization not only enables them to serve critical physiological functions adaptively but also differentiates them from many other biological networks such as gene regulatory networks and cell signaling networks. This review examines this fundamental property of the organelle networks from a systems point of view. The focus is exclusively on homotypic networks formed by mitochondria, lysosomes, endosomes, and the endoplasmic reticulum, respectively. First, key mechanisms that drive the dynamic organization of these networks are summarized. Then, several distinct organizational properties of these networks are highlighted. Next, spatial properties of the dynamic organization of these networks are emphasized, and their functional implications are examined. Finally, some representative molecular machineries that mediate the dynamic organization of these networks are surveyed. Overall, the dynamic organization of intracellular organelle networks is emerging as a fundamental and unifying paradigm in the internal organization of eukaryotic cells. This article is categorized under: Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Shuhao Zhang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
A Large Scale Systemic RNAi Screen in the Red Flour Beetle Tribolium castaneum Identifies Novel Genes Involved in Insect Muscle Development. G3-GENES GENOMES GENETICS 2019; 9:1009-1026. [PMID: 30733381 PMCID: PMC6469426 DOI: 10.1534/g3.118.200995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although muscle development has been widely studied in Drosophila melanogaster there are still many gaps in our knowledge, and it is not known to which extent this knowledge can be transferred to other insects. To help in closing these gaps we participated in a large-scale RNAi screen that used the red flour beetle, Tribolium castaneum, as a screening platform. The effects of systemic RNAi were screened upon double-stranded RNA injections into appropriate muscle-EGFP tester strains. Injections into pupae were followed by the analysis of the late embryonic/early larval muscle patterns, and injections into larvae by the analysis of the adult thoracic muscle patterns. Herein we describe the results of the first-pass screens with pupal and larval injections, which covered ∼8,500 and ∼5,000 genes, respectively, of a total of ∼16,500 genes of the Tribolium genome. Apart from many genes known from Drosophila as regulators of muscle development, a collection of genes previously unconnected to muscle development yielded phenotypes in larval body wall and leg muscles as well as in indirect flight muscles. We then present the main candidates from the pupal injection screen that remained after being processed through a series of verification and selection steps. Further, we discuss why distinct though overlapping sets of genes are revealed by the Drosophila and Tribolium screening approaches.
Collapse
|
11
|
Teixeira MB, Alborghetti MR, Kobarg J. Fasciculation and elongation zeta proteins 1 and 2: From structural flexibility to functional diversity. World J Biol Chem 2019; 10:28-43. [PMID: 30815230 PMCID: PMC6388297 DOI: 10.4331/wjbc.v10.i2.28] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/02/2019] [Accepted: 01/28/2019] [Indexed: 02/05/2023] Open
Abstract
Fasciculation and elongation zeta/zygin (FEZ) proteins are a family of hub proteins and share many characteristics like high connectivity in interaction networks, they are involved in several cellular processes, evolve slowly and in general have intrinsically disordered regions. In 1985, unc-76 gene was firstly described and involved in axonal growth in C. elegans, and in 1997 Bloom and Horvitz enrolled also the human homologues genes, FEZ1 and FEZ2, in this process. While nematodes possess one gene (unc-76), mammalians have one more copy (FEZ1 and FEZ2). Several animal models have been used to study FEZ family functions like: C. elegans, D. melanogaster, R. novergicus and human cells. Complementation assays were performed and demonstrated the function conservation between paralogues. Human FEZ1 protein is more studied followed by UNC-76 and FEZ2 proteins, respectively. While FEZ1 and UNC-76 shared interaction partners, FEZ2 evolved and increased the number of protein-protein interactions (PPI) with cytoplasmatic partners. FEZ proteins are implicated in intracellular transport, acting as bivalent cargo transport adaptors in kinesin-mediated movement. Especially in light of this cellular function, this family of proteins has been involved in several processes like neuronal development, neurological disorders, viral infection and autophagy. However, nuclear functions of FEZ proteins have been explored as well, due to high content of PPI with nuclear proteins, correlating FEZ1 expression to Sox2 and Hoxb4 gene regulation and retinoic acid signaling. These recent findings open new avenue to study FEZ proteins functions and its involvement in already described processes. This review intends to reunite aspects of evolution, structure, interaction partners and function of FEZ proteins and correlate them to physiological and pathological processes.
Collapse
Affiliation(s)
- Mariana Bertini Teixeira
- Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
| | | | - Jörg Kobarg
- Institute of Biology, Department of Biochemistry and Tissue Biology, University of Campinas, Campinas 13083-862, Brazil
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-862, Brazil
| |
Collapse
|
12
|
Cockburn JJB, Hesketh SJ, Mulhair P, Thomsen M, O'Connell MJ, Way M. Insights into Kinesin-1 Activation from the Crystal Structure of KLC2 Bound to JIP3. Structure 2018; 26:1486-1498.e6. [PMID: 30197037 PMCID: PMC6224480 DOI: 10.1016/j.str.2018.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/03/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Kinesin-1 transports numerous cellular cargoes along microtubules. The kinesin-1 light chain (KLC) mediates cargo binding and regulates kinesin-1 motility. To investigate the molecular basis for kinesin-1 recruitment and activation by cargoes, we solved the crystal structure of the KLC2 tetratricopeptide repeat (TPR) domain bound to the cargo JIP3. This, combined with biophysical and molecular evolutionary analyses, reveals a kinesin-1 cargo binding site, located on KLC TPR1, which is conserved in homologs from sponges to humans. In the complex, JIP3 crosslinks two KLC2 TPR domains via their TPR1s. We show that TPR1 forms a dimer interface that mimics JIP3 binding in all crystal structures of the unbound KLC TPR domain. We propose that cargo-induced dimerization of the KLC TPR domains via TPR1 is a general mechanism for activating kinesin-1. We relate this to activation by tryptophan-acidic cargoes, explaining how different cargoes activate kinesin-1 through related molecular mechanisms.
Collapse
Affiliation(s)
- Joseph J B Cockburn
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Sophie J Hesketh
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Peter Mulhair
- Computational and Molecular Evolutionary Biology Research Group, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Maren Thomsen
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Mary J O'Connell
- Computational and Molecular Evolutionary Biology Research Group, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
13
|
Tropea D, Hardingham N, Millar K, Fox K. Mechanisms underlying the role of DISC1 in synaptic plasticity. J Physiol 2018; 596:2747-2771. [PMID: 30008190 PMCID: PMC6046077 DOI: 10.1113/jp274330] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Disrupted in schizophrenia 1 (DISC1) is an important hub protein, forming multimeric complexes by self-association and interacting with a large number of synaptic and cytoskeletal molecules. The synaptic location of DISC1 in the adult brain suggests a role in synaptic plasticity, and indeed, a number of studies have discovered synaptic plasticity impairments in a variety of different DISC1 mutants. This review explores the possibility that DISC1 is an important molecule for organizing proteins involved in synaptic plasticity and examines why mutations in DISC1 impair plasticity. It concentrates on DISC1's role in interacting with synaptic proteins, controlling dendritic structure and cellular trafficking of mRNA, synaptic vesicles and mitochondria. N-terminal directed mutations appear to impair synaptic plasticity through interactions with phosphodiesterase 4B (PDE4B) and hence protein kinase A (PKA)/GluA1 and PKA/cAMP response element-binding protein (CREB) signalling pathways, and affect spine structure through interactions with kalirin 7 (Kal-7) and Rac1. C-terminal directed mutations also impair plasticity possibly through altered interactions with lissencephaly protein 1 (LIS1) and nuclear distribution protein nudE-like 1 (NDEL1), thereby affecting developmental processes such as dendritic structure and spine maturation. Many of the same molecules involved in DISC1's cytoskeletal interactions are also involved in intracellular trafficking, raising the possibility that impairments in intracellular trafficking affect cytoskeletal development and vice versa. While the multiplicity of DISC1 protein interactions makes it difficult to pinpoint a single causal signalling pathway, we suggest that the immediate-term effects of N-terminal influences on GluA1, Rac1 and CREB, coupled with the developmental effects of C-terminal influences on trafficking and the cytoskeleton make up the two main branches of DISC1's effect on synaptic plasticity and dendritic spine stability.
Collapse
Affiliation(s)
- Daniela Tropea
- Neurospychiatric GeneticsTrinity Center for Health Sciences and Trinity College Institute of Neuroscience (TCIN)Trinity College DublinDublinIreland
| | - Neil Hardingham
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| | - Kirsty Millar
- Centre for Genomic & Experimental MedicineMRC Institute of Genetics & Molecular MedicineWestern General HospitalUniversity of EdinburghCrewe RoadEdinburghUK
| | - Kevin Fox
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| |
Collapse
|
14
|
UNC-16/JIP3 and UNC-76/FEZ1 limit the density of mitochondria in C. elegans neurons by maintaining the balance of anterograde and retrograde mitochondrial transport. Sci Rep 2018; 8:8938. [PMID: 29895958 PMCID: PMC5997755 DOI: 10.1038/s41598-018-27211-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/25/2018] [Indexed: 12/23/2022] Open
Abstract
We investigate the role of axonal transport in regulating neuronal mitochondrial density. We show that the density of mitochondria in the touch receptor neuron (TRN) of adult Caenorhabditis elegans is constant. Mitochondrial density and transport are controlled both by the Kinesin heavy chain and the Dynein-Dynactin complex. However, unlike in other models, the presence of mitochondria in C. elegans TRNs depends on a Kinesin light chain as well. Mutants in the three C. elegans miro genes do not alter mitochondrial density in the TRNs. Mutants in the Kinesin-1 associated proteins, UNC-16/JIP3 and UNC-76/FEZ1, show increased mitochondrial density and also have elevated levels of both the Kinesin Heavy and Light Chains in neurons. Genetic analyses suggest that, the increased mitochondrial density at the distal end of the neuronal process in unc-16 and unc-76 depends partly on Dynein. We observe a net anterograde bias in the ratio of anterograde to retrograde mitochondrial flux in the neuronal processes of unc-16 and unc-76, likely due to both increased Kinesin-1 and decreased Dynein in the neuronal processes. Our study shows that UNC-16 and UNC-76 indirectly limit mitochondrial density in the neuronal process by maintaining a balance in anterograde and retrograde mitochondrial axonal transport.
Collapse
|
15
|
Dey S, Banker G, Ray K. Anterograde Transport of Rab4-Associated Vesicles Regulates Synapse Organization in Drosophila. Cell Rep 2017; 18:2452-2463. [PMID: 28273459 DOI: 10.1016/j.celrep.2017.02.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 12/19/2016] [Accepted: 02/09/2017] [Indexed: 11/29/2022] Open
Abstract
Local endosomal recycling at synapses is essential to maintain neurotransmission. Rab4GTPase, found on sorting endosomes, is proposed to balance the flow of vesicles among endocytic, recycling, and degradative pathways in the presynaptic compartment. Here, we report that Rab4-associated vesicles move bidirectionally in Drosophila axons but with an anterograde bias, resulting in their moderate enrichment at the synaptic region of the larval ventral ganglion. Results from FK506 binding protein (FKBP) and FKBP-Rapamycin binding domain (FRB) conjugation assays in rat embryonic fibroblasts together with genetic analyses in Drosophila indicate that an association with Kinesin-2 (mediated by the tail domain of Kinesin-2α/KIF3A/KLP64D subunit) moves Rab4-associated vesicles toward the synapse. Reduction in the anterograde traffic of Rab4 causes an expansion of the volume of the synapse-bearing region in the ventral ganglion and increases the motility of Drosophila larvae. These results suggest that Rab4-dependent vesicular traffic toward the synapse plays a vital role in maintaining synaptic balance in this neuronal network.
Collapse
Affiliation(s)
- Swagata Dey
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| | - Gary Banker
- Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR 97239, USA
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
16
|
Hapairai LK, Mysore K, Chen Y, Harper EI, Scheel MP, Lesnik AM, Sun L, Severson DW, Wei N, Duman-Scheel M. Lure-and-Kill Yeast Interfering RNA Larvicides Targeting Neural Genes in the Human Disease Vector Mosquito Aedes aegypti. Sci Rep 2017; 7:13223. [PMID: 29038510 PMCID: PMC5643370 DOI: 10.1038/s41598-017-13566-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/25/2017] [Indexed: 12/23/2022] Open
Abstract
New mosquito control strategies are vitally needed to address established arthropod-borne infectious diseases such as dengue and yellow fever and emerging diseases such as Zika and chikungunya, all of which are transmitted by the disease vector mosquito Aedes aegypti. In this investigation, Saccharomyces cerevisiae (baker’s yeast) was engineered to produce short hairpin RNAs (shRNAs) corresponding to the Aedes aegypti orthologs of fasciculation and elongation protein zeta 2 (fez2) and leukocyte receptor cluster (lrc) member, two genes identified in a recent screen for A. aegypti larval lethal genes. Feeding A. aegypti with the engineered yeasts resulted in silenced target gene expression, disrupted neural development, and highly significant larval mortality. Larvicidal activities were retained following heat inactivation and drying of the yeast into tabular formulations that induced >95% mortality and were found to attract adult females to oviposit. These ready-to-use inactivated yeast interfering RNA tablets may one day facilitate the seamless integration of this new class of lure-and-kill species-specific biorational mosquito larvicides into integrated mosquito control programs.
Collapse
Affiliation(s)
- Limb K Hapairai
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA.,The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA
| | - Keshava Mysore
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA.,The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA
| | - Yingying Chen
- The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA.,The University of Notre Dame Department of Civil and Environmental Engineering, Notre Dame, IN, USA
| | - Elizabeth I Harper
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA.,The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA
| | - Max P Scheel
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA
| | - Alexandra M Lesnik
- The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA
| | - Longhua Sun
- The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA.,The University of Notre Dame Department of Biological Sciences, Notre Dame, IN, USA
| | - David W Severson
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA.,The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA.,The University of Notre Dame Department of Biological Sciences, Notre Dame, IN, USA
| | - Na Wei
- The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA.,The University of Notre Dame Department of Civil and Environmental Engineering, Notre Dame, IN, USA
| | - Molly Duman-Scheel
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, South Bend, IN, USA. .,The University of Notre Dame Eck Institute for Global Health, Notre Dame, IN, USA. .,The University of Notre Dame Department of Biological Sciences, Notre Dame, IN, USA.
| |
Collapse
|
17
|
Barlan K, Gelfand VI. Microtubule-Based Transport and the Distribution, Tethering, and Organization of Organelles. Cold Spring Harb Perspect Biol 2017; 9:9/5/a025817. [PMID: 28461574 DOI: 10.1101/cshperspect.a025817] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SUMMARYMicrotubules provide long tracks along which a broad range of organelles and vesicles are transported by kinesin and dynein motors. Motor protein complexes also tether cargoes to cytoskeletal filaments, helping facilitate their interaction and communication. The generation of biochemically distinct microtubule subpopulations allows subsets of motors to recognize a given microtubule identity, allowing further organization within the cytoplasm. Both transport and tethering are spatiotemporally regulated through multiple modes, including acute modification of both motor-cargo and motor-track associations by various physiological signals. Strict regulation of intracellular transport is particularly important in specialized cell types such as neurons. Here, we review general mechanisms by which cargo transport is controlled and also highlight examples of transport regulated by multiple mechanisms.
Collapse
Affiliation(s)
- Kari Barlan
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
18
|
Molecular pathogenesis of peripheral neuropathies: insights from Drosophila models. Curr Opin Genet Dev 2017; 44:61-73. [PMID: 28213160 DOI: 10.1016/j.gde.2017.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/10/2017] [Accepted: 01/26/2017] [Indexed: 01/18/2023]
Abstract
Peripheral neuropathies are characterized by degeneration of peripheral motor, sensory and/or autonomic axons, leading to progressive distal muscle weakness, sensory deficits and/or autonomic dysfunction. Acquired peripheral neuropathies, e.g., as a side effect of chemotherapy, are distinguished from inherited peripheral neuropathies (IPNs). Drosophila models for chemotherapy-induced peripheral neuropathy and several IPNs have provided novel insight into the molecular mechanisms underlying axonal degeneration. Forward genetic screens have predictive value for discovery of human IPN genes, and the pathogenicity of novel mutations in known IPN genes can be evaluated in Drosophila. Future screens for genes and compounds that modify Drosophila IPN phenotypes promise to make valuable contributions to unraveling the molecular pathogenesis and identification of therapeutic targets for these incurable diseases.
Collapse
|
19
|
Role of kinesin-1-based microtubule sliding in Drosophila nervous system development. Proc Natl Acad Sci U S A 2016; 113:E4985-94. [PMID: 27512046 DOI: 10.1073/pnas.1522416113] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The plus-end microtubule (MT) motor kinesin-1 is essential for normal development, with key roles in the nervous system. Kinesin-1 drives axonal transport of membrane cargoes to fulfill the metabolic needs of neurons and maintain synapses. We have previously demonstrated that kinesin-1, in addition to its well-established role in organelle transport, can drive MT-MT sliding by transporting "cargo" MTs along "track" MTs, resulting in dramatic cell shape changes. The mechanism and physiological relevance of this MT sliding are unclear. In addition to its motor domain, kinesin-1 contains a second MT-binding site, located at the C terminus of the heavy chain. Here, we mutated this C-terminal MT-binding site such that the ability of kinesin-1 to slide MTs is significantly compromised, whereas cargo transport is unaffected. We introduced this mutation into the genomic locus of kinesin-1 heavy chain (KHC), generating the Khc(mutA) allele. Khc(mutA) neurons displayed significant MT sliding defects while maintaining normal transport of many cargoes. Using this mutant, we demonstrated that MT sliding is required for axon and dendrite outgrowth in vivo. Consistent with these results, Khc(mutA) flies displayed severe locomotion and viability defects. To test the role of MT sliding further, we engineered a chimeric motor that actively slides MTs but cannot transport organelles. Activation of MT sliding in Khc(mutA) neurons using this chimeric motor rescued axon outgrowth in cultured neurons and in vivo, firmly establishing the role of sliding in axon outgrowth. These results demonstrate that MT sliding by kinesin-1 is an essential biological phenomenon required for neuronal morphogenesis and normal nervous system development.
Collapse
|
20
|
Morfini G, Schmidt N, Weissmann C, Pigino G, Kins S. Conventional kinesin: Biochemical heterogeneity and functional implications in health and disease. Brain Res Bull 2016; 126:347-353. [PMID: 27339812 DOI: 10.1016/j.brainresbull.2016.06.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/13/2016] [Accepted: 06/18/2016] [Indexed: 11/30/2022]
Abstract
Intracellular trafficking events powered by microtubule-based molecular motors facilitate the targeted delivery of selected molecular components to specific neuronal subdomains. Within this context, we provide a brief review of mechanisms underlying the execution of axonal transport (AT) by conventional kinesin, the most abundant kinesin-related motor protein in the mature nervous system. We emphasize the biochemical heterogeneity of this multi-subunit motor protein, further discussing its significance in light of recent discoveries revealing its regulation by various protein kinases. In addition, we raise issues relevant to the mode of conventional kinesin attachment to cargoes and examine recent evidence linking alterations in conventional kinesin phosphorylation to the pathogenesis of adult-onset neurodegenerative diseases.
Collapse
Affiliation(s)
- Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Nadine Schmidt
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, 67663 Kaiserslautern, Germany
| | - Carina Weissmann
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Gustavo Pigino
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016 Córdoba, Argentina
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, 67663 Kaiserslautern, Germany.
| |
Collapse
|
21
|
Phosphorylation of FEZ1 by Microtubule Affinity Regulating Kinases regulates its function in presynaptic protein trafficking. Sci Rep 2016; 6:26965. [PMID: 27247180 PMCID: PMC4887895 DOI: 10.1038/srep26965] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/11/2016] [Indexed: 12/28/2022] Open
Abstract
Adapters bind motor proteins to cargoes and therefore play essential roles in Kinesin-1 mediated intracellular transport. The regulatory mechanisms governing adapter functions and the spectrum of cargoes recognized by individual adapters remain poorly defined. Here, we show that cargoes transported by the Kinesin-1 adapter FEZ1 are enriched for presynaptic components and identify that specific phosphorylation of FEZ1 at its serine 58 regulatory site is mediated by microtubule affinity-regulating kinases (MARK/PAR-1). Loss of MARK/PAR-1 impairs axonal transport, with adapter and cargo abnormally co-aggregating in neuronal cell bodies and axons. Presynaptic specializations are markedly reduced and distorted in FEZ1 and MARK/PAR-1 mutants. Strikingly, abnormal co-aggregates of unphosphorylated FEZ1, Kinesin-1 and its putative cargoes are present in brains of transgenic mice modelling aspects of Alzheimer's disease, a neurodegenerative disorder exhibiting impaired axonal transport and altered MARK activity. Our findings suggest that perturbed FEZ1-mediated synaptic delivery of proteins arising from abnormal signalling potentially contributes to the process of neurodegeneration.
Collapse
|
22
|
Beck K, Ehmann N, Andlauer TFM, Ljaschenko D, Strecker K, Fischer M, Kittel RJ, Raabe T. Loss of the Coffin-Lowry syndrome-associated gene RSK2 alters ERK activity, synaptic function and axonal transport in Drosophila motoneurons. Dis Model Mech 2015; 8:1389-400. [PMID: 26398944 PMCID: PMC4631788 DOI: 10.1242/dmm.021246] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/27/2015] [Indexed: 01/06/2023] Open
Abstract
Plastic changes in synaptic properties are considered as fundamental for adaptive behaviors. Extracellular-signal-regulated kinase (ERK)-mediated signaling has been implicated in regulation of synaptic plasticity. Ribosomal S6 kinase 2 (RSK2) acts as a regulator and downstream effector of ERK. In the brain, RSK2 is predominantly expressed in regions required for learning and memory. Loss-of-function mutations in human RSK2 cause Coffin-Lowry syndrome, which is characterized by severe mental retardation and low IQ scores in affected males. Knockout of RSK2 in mice or the RSK ortholog in Drosophila results in a variety of learning and memory defects. However, overall brain structure in these animals is not affected, leaving open the question of the pathophysiological consequences. Using the fly neuromuscular system as a model for excitatory glutamatergic synapses, we show that removal of RSK function causes distinct defects in motoneurons and at the neuromuscular junction. Based on histochemical and electrophysiological analyses, we conclude that RSK is required for normal synaptic morphology and function. Furthermore, loss of RSK function interferes with ERK signaling at different levels. Elevated ERK activity was evident in the somata of motoneurons, whereas decreased ERK activity was observed in axons and the presynapse. In addition, we uncovered a novel function of RSK in anterograde axonal transport. Our results emphasize the importance of fine-tuning ERK activity in neuronal processes underlying higher brain functions. In this context, RSK acts as a modulator of ERK signaling.
Collapse
Affiliation(s)
- Katherina Beck
- University of Würzburg, Institute of Medical Radiation and Cell Research, Versbacherstraße 5, Würzburg D-97078, Germany
| | - Nadine Ehmann
- University of Würzburg, Institute of Physiology, Department of Neurophysiology, Röntgenring 9, Würzburg D-97070, Germany
| | - Till F M Andlauer
- University of Würzburg, Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, Josef-Schneider-Straße 2, Würzburg D-97080, Germany Freie Universität Berlin, Institute of Biology, Takusstraße 6, Berlin D-14195, Germany Max Planck Institute of Colloidals and Interfaces, Am Mühlenberg 1, Potsdam D-14476, Germany
| | - Dmitrij Ljaschenko
- University of Würzburg, Institute of Physiology, Department of Neurophysiology, Röntgenring 9, Würzburg D-97070, Germany
| | - Katrin Strecker
- University of Würzburg, Institute of Medical Radiation and Cell Research, Versbacherstraße 5, Würzburg D-97078, Germany
| | - Matthias Fischer
- University Hospital Würzburg, Department of Psychiatry, Psychosomatics and Psychotherapy, Füchsleinstraße 15, Würzburg 97080, Germany
| | - Robert J Kittel
- University of Würzburg, Institute of Physiology, Department of Neurophysiology, Röntgenring 9, Würzburg D-97070, Germany
| | - Thomas Raabe
- University of Würzburg, Institute of Medical Radiation and Cell Research, Versbacherstraße 5, Würzburg D-97078, Germany
| |
Collapse
|
23
|
Siebert M, Böhme MA, Driller JH, Babikir H, Mampell MM, Rey U, Ramesh N, Matkovic T, Holton N, Reddy-Alla S, Göttfert F, Kamin D, Quentin C, Klinedinst S, Andlauer TF, Hell SW, Collins CA, Wahl MC, Loll B, Sigrist SJ. A high affinity RIM-binding protein/Aplip1 interaction prevents the formation of ectopic axonal active zones. eLife 2015; 4. [PMID: 26274777 PMCID: PMC4536467 DOI: 10.7554/elife.06935] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/24/2015] [Indexed: 12/17/2022] Open
Abstract
Synaptic vesicles (SVs) fuse at active zones (AZs) covered by a protein scaffold, at Drosophila synapses comprised of ELKS family member Bruchpilot (BRP) and RIM-binding protein (RBP). We here demonstrate axonal co-transport of BRP and RBP using intravital live imaging, with both proteins co-accumulating in axonal aggregates of several transport mutants. RBP, via its C-terminal Src-homology 3 (SH3) domains, binds Aplip1/JIP1, a transport adaptor involved in kinesin-dependent SV transport. We show in atomic detail that RBP C-terminal SH3 domains bind a proline-rich (PxxP) motif of Aplip1/JIP1 with submicromolar affinity. Pointmutating this PxxP motif provoked formation of ectopic AZ-like structures at axonal membranes. Direct interactions between AZ proteins and transport adaptors seem to provide complex avidity and shield synaptic interaction surfaces of pre-assembled scaffold protein transport complexes, thus, favouring physiological synaptic AZ assembly over premature assembly at axonal membranes. DOI:http://dx.doi.org/10.7554/eLife.06935.001 To pass on information, the neurons that make up the nervous system connect at structures known as synapses. Chemical messengers called neurotransmitters are released from one neuron, and travel across the synapse to trigger a response in the neighbouring cell. The formation of new synapses plays an important role in learning and memory, but many aspects of this process are not well understood. In a specific region of the synapse called the active zone, a scaffold of proteins helps to release the neurotransmitters. These proteins are made in the cell body of the neuron, and are then transported to the end of the long, thin axons that protrude from the cell body. This presents a challenge for the cell, because the components of the active zone scaffold must be correctly targeted to the synapse at the end of the axon, ensuring the active zone scaffold assembles only at its proper location. Siebert, Böhme et al. studied how some of the proteins that are found in the active zone scaffold of the fruit fly Drosophila are transported along axons. Labelling the proteins with fluorescent markers allowed their movement to be examined under a microscope in living Drosophila larvae. The results showed that two of the proteins—known as BRP and RBP—are transported along the axons together. Further investigation revealed that a transport adaptor protein called Aplip1, which binds to RBP, is required for this movement. Siebert, Böhme et al. established the structure of the part of RBP where this interaction occurs, and found that mutating this region causes premature active zone scaffold assembly in the axonal part of the neuron. The interaction between RBP and Aplip1 is very strong, and this helps to prevent the scaffold assembling before it has reached the correct part of the neuron. Exactly how the transport adaptor and active zone protein are separated once they reach their final destination (the synapse) remains to be discovered. DOI:http://dx.doi.org/10.7554/eLife.06935.002
Collapse
Affiliation(s)
- Matthias Siebert
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Mathias A Böhme
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Jan H Driller
- Institute of Chemistry and Biochemisty/Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Husam Babikir
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Malou M Mampell
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Ulises Rey
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Tanja Matkovic
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Nicole Holton
- Institute of Chemistry and Biochemisty/Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Suneel Reddy-Alla
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Fabian Göttfert
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Dirk Kamin
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christine Quentin
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Susan Klinedinst
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Till Fm Andlauer
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Stefan W Hell
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Catherine A Collins
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Markus C Wahl
- Institute of Chemistry and Biochemisty/Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Bernhard Loll
- Institute of Chemistry and Biochemisty/Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
24
|
Papandréou MJ, Vacher H, Fache MP, Klingler E, Rueda-Boroni F, Ferracci G, Debarnot C, Pipéroglou C, Garcia Del Caño G, Goutebroze L, Dargent B. CK2-regulated schwannomin-interacting protein IQCJ-SCHIP-1 association with AnkG contributes to the maintenance of the axon initial segment. J Neurochem 2015; 134:527-37. [PMID: 25950943 DOI: 10.1111/jnc.13158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/14/2015] [Accepted: 05/04/2015] [Indexed: 11/30/2022]
Abstract
The axon initial segment (AIS) plays a central role in electrogenesis and in the maintenance of neuronal polarity. Its molecular organization is dependent on the scaffolding protein ankyrin (Ank) G and is regulated by kinases. For example, the phosphorylation of voltage-gated sodium channels by the protein kinase CK2 regulates their interaction with AnkG and, consequently, their accumulation at the AIS. We previously showed that IQ motif containing J-Schwannomin-Interacting Protein 1 (IQCJ-SCHIP-1), an isoform of the SCHIP-1, accumulated at the AIS in vivo. Here, we analyzed the molecular mechanisms involved in IQCJ-SCHIP-1-specific axonal location. We showed that IQCJ-SCHIP-1 accumulation in the AIS of cultured hippocampal neurons depended on AnkG expression. Pull-down assays and surface plasmon resonance analysis demonstrated that AnkG binds to CK2-phosphorylated IQCJ-SCHIP-1 but not to the non-phosphorylated protein. Surface plasmon resonance approaches using IQCJ-SCHIP-1, SCHIP-1a, another SCHIP-1 isoform, and their C-terminus tail mutants revealed that a segment including multiple CK2-phosphorylatable sites was directly involved in the interaction with AnkG. Pharmacological inhibition of CK2 diminished both IQCJ-SCHIP-1 and AnkG accumulation in the AIS. Silencing SCHIP-1 expression reduced AnkG cluster at the AIS. Finally, over-expression of IQCJ-SCHIP-1 decreased AnkG concentration at the AIS, whereas a mutant deleted of the CK2-regulated AnkG interaction site did not. Our study reveals that CK2-regulated IQJC-SCHIP-1 association with AnkG contributes to AIS maintenance. The axon initial segment (AIS) organization depends on ankyrin (Ank) G and kinases. Here we showed that AnkG binds to CK2-phosphorylated IQCJ-SCHIP-1, in a segment including 12 CK2-phosphorylatable sites. In cultured neurons, either pharmacological inhibition of CK2 or IQCJ-SCHIP-1 silencing reduced AnkG clustering. Overexpressed IQCJ-SCHIP-1 decreased AnkG concentration at the AIS whereas a mutant deleted of the CK2-regulated AnkG interaction site did not. Thus, CK2-regulated IQJC-SCHIP-1 association with AnkG contributes to AIS maintenance.
Collapse
Affiliation(s)
| | - Hélène Vacher
- CRN2M-UMR7286, Aix Marseille Université, CNRS, Marseille, France
| | | | - Esther Klingler
- Institut du Fer à Moulin, Inserm, UMR-S 839, Université Pierre et Marie-Curie, Paris, France
| | | | | | - Claire Debarnot
- CRN2M-UMR7286, Aix Marseille Université, CNRS, Marseille, France
| | | | - Gontzal Garcia Del Caño
- CRN2M-UMR7286, Aix Marseille Université, CNRS, Marseille, France.,Department of Neurosciences, University of the Basque Country, Vitoria-Gasteiz, Spain
| | - Laurence Goutebroze
- Institut du Fer à Moulin, Inserm, UMR-S 839, Université Pierre et Marie-Curie, Paris, France
| | | |
Collapse
|
25
|
Malikov V, da Silva ES, Jovasevic V, Bennett G, de Souza Aranha Vieira DA, Schulte B, Diaz-Griffero F, Walsh D, Naghavi MH. HIV-1 capsids bind and exploit the kinesin-1 adaptor FEZ1 for inward movement to the nucleus. Nat Commun 2015; 6:6660. [PMID: 25818806 PMCID: PMC4380233 DOI: 10.1038/ncomms7660] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022] Open
Abstract
Intracellular transport of cargos, including many viruses, involves directed movement on microtubules mediated by motor proteins. Although a number of viruses bind motors of opposing directionality, how they associate with and control these motors to accomplish directed movement remains poorly understood. Here we show that human immunodeficiency virus type 1 (HIV-1) associates with the kinesin-1 adaptor protein, Fasiculation and Elongation Factor zeta 1 (FEZ1). RNAi-mediated FEZ1 depletion blocks early infection, with virus particles exhibiting bi-directional motility but no net movement to the nucleus. Furthermore, both dynein and kinesin-1 motors are required for HIV-1 trafficking to the nucleus. Finally, the ability of exogenously expressed FEZ1 to promote early HIV-1 infection requires binding to kinesin-1. Our findings demonstrate that opposing motors both contribute to early HIV-1 movement and identify the kinesin-1 adaptor, FEZ1 as a capsid-associated host regulator of this process usurped by HIV-1 to accomplish net inward movement towards the nucleus.
Collapse
Affiliation(s)
- Viacheslav Malikov
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| | - Eveline Santos da Silva
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Vladimir Jovasevic
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Geoffrey Bennett
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| | | | - Bianca Schulte
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Felipe Diaz-Griffero
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Derek Walsh
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Mojgan H Naghavi
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| |
Collapse
|
26
|
Serpinskaya AS, Tuphile K, Rabinow L, Gelfand VI. Protein kinase Darkener of apricot and its substrate EF1γ regulate organelle transport along microtubules. J Cell Sci 2013; 127:33-9. [PMID: 24163433 DOI: 10.1242/jcs.123885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Regulation of organelle transport along microtubules is important for proper distribution of membrane organelles and protein complexes in the cytoplasm. RNAi-mediated knockdown in cultured Drosophila S2 cells demonstrates that two microtubule-binding proteins, a unique isoform of Darkener of apricot (DOA) protein kinase, and its substrate, translational elongation factor EF1γ, negatively regulate transport of several classes of membrane organelles along microtubules. Inhibition of transport by EF1γ requires its phosphorylation by DOA on serine 294. Together, our results indicate a new role for two proteins that have not previously been implicated in regulation of the cytoskeleton. These results further suggest that the biological role of some of the proteins binding to the microtubule track is to regulate cargo transport along these tracks.
Collapse
Affiliation(s)
- Anna S Serpinskaya
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 616011, USA
| | | | | | | |
Collapse
|
27
|
Alborghetti MR, Furlan ADS, da Silva JC, Sforça ML, Honorato RV, Granato DC, dos Santos Migueleti DL, Neves JL, de Oliveira PSL, Paes-Leme AF, Zeri ACDM, de Torriani ICL, Kobarg J. Structural analysis of intermolecular interactions in the kinesin adaptor complex fasciculation and elongation protein zeta 1/ short coiled-coil protein (FEZ1/SCOCO). PLoS One 2013; 8:e76602. [PMID: 24116125 PMCID: PMC3792052 DOI: 10.1371/journal.pone.0076602] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/29/2013] [Indexed: 01/15/2023] Open
Abstract
Cytoskeleton and protein trafficking processes, including vesicle transport to synapses, are key processes in neuronal differentiation and axon outgrowth. The human protein FEZ1 (fasciculation and elongation protein zeta 1 / UNC-76, in C. elegans), SCOCO (short coiled-coil protein / UNC-69) and kinesins (e.g. kinesin heavy chain / UNC116) are involved in these processes. Exploiting the feature of FEZ1 protein as a bivalent adapter of transport mediated by kinesins and FEZ1 protein interaction with SCOCO (proteins involved in the same path of axonal growth), we investigated the structural aspects of intermolecular interactions involved in this complex formation by NMR (Nuclear Magnetic Resonance), cross-linking coupled with mass spectrometry (MS), SAXS (Small Angle X-ray Scattering) and molecular modelling. The topology of homodimerization was accessed through NMR (Nuclear Magnetic Resonance) studies of the region involved in this process, corresponding to FEZ1 (92-194). Through studies involving the protein in its monomeric configuration (reduced) and dimeric state, we propose that homodimerization occurs with FEZ1 chains oriented in an anti-parallel topology. We demonstrate that the interaction interface of FEZ1 and SCOCO defined by MS and computational modelling is in accordance with that previously demonstrated for UNC-76 and UNC-69. SAXS and literature data support a heterotetrameric complex model. These data provide details about the interaction interfaces probably involved in the transport machinery assembly and open perspectives to understand and interfere in this assembly and its involvement in neuronal differentiation and axon outgrowth.
Collapse
Affiliation(s)
- Marcos Rodrigo Alborghetti
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Ariane da Silva Furlan
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - Júlio César da Silva
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Maurício Luís Sforça
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Rodrigo Vargas Honorato
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Daniela Campos Granato
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Deivid Lucas dos Santos Migueleti
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
- Departamento de Genética, Evolução e Bioagentes, Programa de Pós-graduação em Genética e Biologia Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | - Jorge L. Neves
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Paulo Sergio Lopes de Oliveira
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Adriana Franco Paes-Leme
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
| | - Ana Carolina de Mattos Zeri
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| | | | - Jörg Kobarg
- Laboratório Nacional de Biociências-LNBio, Centro Nacional de Pesquisa em Energia e Materiais-CNPEM, Campinas, SP, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
- Departamento de Genética, Evolução e Bioagentes, Programa de Pós-graduação em Genética e Biologia Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brasil
| |
Collapse
|
28
|
Behrens C, Binotti B, Schmidt C, Robinson CV, Chua JJE, Kühnel K. Crystal structure of the human short coiled coil protein and insights into SCOC-FEZ1 complex formation. PLoS One 2013; 8:e76355. [PMID: 24098481 PMCID: PMC3788124 DOI: 10.1371/journal.pone.0076355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/23/2013] [Indexed: 01/18/2023] Open
Abstract
The short coiled coil protein (SCOC) forms a complex with fasciculation and elongation protein zeta 1 (FEZ1). This complex is involved in autophagy regulation. We determined the crystal structure of the coiled coil domain of human SCOC at 2.7 Å resolution. SCOC forms a parallel left handed coiled coil dimer. We observed two distinct dimers in the crystal structure, which shows that SCOC is conformationally flexible. This plasticity is due to the high incidence of polar and charged residues at the core a/d-heptad positions. We prepared two double mutants, where these core residues were mutated to either leucines or valines (E93V/K97L and N125L/N132V). These mutations led to a dramatic increase in stability and change of oligomerisation state. The oligomerisation state of the mutants was characterized by multi-angle laser light scattering and native mass spectrometry measurements. The E93V/K97 mutant forms a trimer and the N125L/N132V mutant is a tetramer. We further demonstrate that SCOC forms a stable homogeneous complex with the coiled coil domain of FEZ1. SCOC dimerization and the SCOC surface residue R117 are important for this interaction.
Collapse
Affiliation(s)
- Caroline Behrens
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Beyenech Binotti
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Carla Schmidt
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - Carol V. Robinson
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - John Jia En Chua
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| | - Karin Kühnel
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
- * E-mail:
| |
Collapse
|
29
|
Chua JJE, Jahn R, Klopfenstein DR. Managing intracellular transport. WORM 2013; 2:e21564. [PMID: 24058857 PMCID: PMC3670458 DOI: 10.4161/worm.21564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 07/18/2012] [Accepted: 07/20/2012] [Indexed: 11/19/2022]
Abstract
Formation and normal function of neuronal synapses are intimately dependent on the delivery to and removal of biological materials from synapses by the intracellular transport machinery. Indeed, defects in intracellular transport contribute to the development and aggravation of neurodegenerative disorders. Despite its importance, regulatory mechanisms underlying this machinery remain poorly defined. We recently uncovered a phosphorylation-regulated mechanism that controls FEZ1-mediated Kinesin-1-based delivery of Stx1 into neuronal axons. Using C. elegans as a model organism to investigate transport defects, we show that FEZ1 mutations resulted in abnormal Stx1 aggregation in neuronal cell bodies and axons. This phenomenon closely resembles transport defects observed in neurodegenerative disorders. Importantly, diminished transport due to mutations of FEZ1 and Kinesin-1 were concomitant with increased accumulation of autophagosomes. Here, we discuss the significance of our findings in a broader context in relation to regulation of Kinesin-mediated transport and neurodegenerative disorders.
Collapse
Affiliation(s)
- John J E Chua
- Department of Neurobiology; Max-Planck-Institute for Biophysical Chemistry; Germany
| | | | | |
Collapse
|
30
|
Mitochondrial trafficking in neuropsychiatric diseases. Neurobiol Dis 2013; 51:66-71. [DOI: 10.1016/j.nbd.2012.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/07/2012] [Accepted: 06/22/2012] [Indexed: 12/31/2022] Open
|
31
|
Seeger MA, Zhang Y, Rice SE. Kinesin tail domains are intrinsically disordered. Proteins 2012; 80:2437-46. [PMID: 22674872 DOI: 10.1002/prot.24128] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/22/2012] [Accepted: 05/25/2012] [Indexed: 12/11/2022]
Abstract
Kinesin motor proteins transport a wide variety of molecular cargoes in a spatially and temporally regulated manner. Kinesin motor domains, which hydrolyze ATP to produce a directed mechanical force along a microtubule, are well conserved throughout the entire superfamily. Outside of the motor domains, kinesin sequences diverge along with their transport functions. The nonmotor regions, particularly the tails, respond to a wide variety of structural and molecular cues that enable kinesins to carry specific cargoes in response to particular cellular signals. Here, we demonstrate that intrinsic disorder is a common structural feature of kinesins. A bioinformatics survey of the full-length sequences of all 43 human kinesins predicts that significant regions of intrinsically disordered residues are present in all kinesins. These regions are concentrated in the nonmotor domains, particularly in the tails and near sites for ligand binding or post-translational modifications. In order to experimentally verify these predictions, we expressed and purified the tail domains of kinesins representing three different families (Kif5B, Kif10, and KifC3). Circular dichroism and NMR spectroscopy experiments demonstrate that the isolated tails are disordered in vitro, yet they retain their functional microtubule-binding activity. On the basis of these results, we propose that intrinsic disorder is a common structural feature that confers functional specificity to kinesins.
Collapse
Affiliation(s)
- Mark A Seeger
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | |
Collapse
|
32
|
Phosphorylation-regulated axonal dependent transport of syntaxin 1 is mediated by a Kinesin-1 adapter. Proc Natl Acad Sci U S A 2012; 109:5862-7. [PMID: 22451907 DOI: 10.1073/pnas.1113819109] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Presynaptic nerve terminals are formed from preassembled vesicles that are delivered to the prospective synapse by kinesin-mediated axonal transport. However, precisely how the various cargoes are linked to the motor proteins remains unclear. Here, we report a transport complex linking syntaxin 1a (Stx) and Munc18, two proteins functioning in synaptic vesicle exocytosis at the presynaptic plasma membrane, to the motor protein Kinesin-1 via the kinesin adaptor FEZ1. Mutation of the FEZ1 ortholog UNC-76 in Caenorhabditis elegans causes defects in the axonal transport of Stx. We also show that binding of FEZ1 to Kinesin-1 and Munc18 is regulated by phosphorylation, with a conserved site (serine 58) being essential for binding. When expressed in C. elegans, wild-type but not phosphorylation-deficient FEZ1 (S58A) restored axonal transport of Stx. We conclude that FEZ1 operates as a kinesin adaptor for the transport of Stx, with cargo loading and unloading being regulated by protein kinases.
Collapse
|
33
|
Reis GF, Yang G, Szpankowski L, Weaver C, Shah SB, Robinson JT, Hays TS, Danuser G, Goldstein LSB. Molecular motor function in axonal transport in vivo probed by genetic and computational analysis in Drosophila. Mol Biol Cell 2012; 23:1700-14. [PMID: 22398725 PMCID: PMC3338437 DOI: 10.1091/mbc.e11-11-0938] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Amyloid precursor protein (APP) vesicle movement by kinesin-1 and cytoplasmic dynein exhibits kinesin-1–dependent velocity. Our data also suggest that kinesin-1 and cytoplasmic dynein motors assemble in stable mixtures on APP vesicles and that their direction and velocity are controlled at least in part by dynein IC. Bidirectional axonal transport driven by kinesin and dynein along microtubules is critical to neuronal viability and function. To evaluate axonal transport mechanisms, we developed a high-resolution imaging system to track the movement of amyloid precursor protein (APP) vesicles in Drosophila segmental nerve axons. Computational analyses of a large number of moving vesicles in defined genetic backgrounds with partial reduction or overexpression of motor proteins enabled us to test with high precision existing and new models of motor activity and coordination in vivo. We discovered several previously unknown features of vesicle movement, including a surprising dependence of anterograde APP vesicle movement velocity on the amount of kinesin-1. This finding is largely incompatible with the biophysical properties of kinesin-1 derived from in vitro analyses. Our data also suggest kinesin-1 and cytoplasmic dynein motors assemble in stable mixtures on APP vesicles and their direction and velocity are controlled at least in part by dynein intermediate chain.
Collapse
Affiliation(s)
- Gerald F Reis
- Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Gene products such as organelles, proteins and RNAs are actively transported to synaptic terminals for the remodeling of pre-existing neuronal connections and formation of new ones. Proteins described as molecular motors mediate this transport and utilize specialized cytoskeletal proteins that function as molecular tracks for the motor based transport of cargos. Molecular motors such as kinesins and dynein's move along microtubule tracks formed by tubulins whereas myosin motors utilize tracks formed by actin. Deficits in active transport of gene products have been implicated in a number of neurological disorders. We describe such disorders collectively as "transportopathies". Here we review current knowledge of critical components of active transport and their relevance to neurodegenerative diseases.
Collapse
|
35
|
Flores R, Hirota Y, Armstrong B, Sawa A, Tomoda T. DISC1 regulates synaptic vesicle transport via a lithium-sensitive pathway. Neurosci Res 2011; 71:71-7. [PMID: 21664390 PMCID: PMC3156137 DOI: 10.1016/j.neures.2011.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 04/26/2011] [Accepted: 05/24/2011] [Indexed: 12/11/2022]
Abstract
Disrupted-in-Schizophrenia 1 (DISC1) is a susceptibility gene for major mental illnesses, including bipolar disorder and schizophrenia. Although the roles of DISC1 in nervous system development and functions are increasingly recognized, pathophysiological mechanisms underlying a range of neuropsychiatric symptoms caused by DISC1 mutations remain unclear. Here we show that DISC1 enhances synaptic vesicle transport along microtubules. Knocking down DISC1 expression results in attenuated vesicle transport in primary cortical neurons. Likewise, expressing the dominant-negative, breakpoint mutant version of DISC1 causes defective vesicle transport, by disrupting the assembly between the kinesin-1 adaptor FEZ1 and the cargo protein Synaptotagmin-1 (Syt-1). In addition, lithium, a mood-stabilizing agent used for the treatment of bipolar disorder, can restore the assembly of FEZ1 and Syt-1, and normalizes the defective transport caused by the dominant-negative DISC1. Thus, this study addresses a new role of DISC1 in organelle transport in neurons, and suggests that this cellular pathway could be therapeutically targeted for the treatment against neuropsychiatric diseases.
Collapse
Affiliation(s)
- Rafael Flores
- Department of Neurosciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Yuki Hirota
- Department of Neurosciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Brian Armstrong
- Department of Neurosciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Department of Neuroscience, Johns Hopkins University, 600N, Wolfe St., Baltimore, MD 21287, USA
| | - Toshifumi Tomoda
- Department of Neurosciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| |
Collapse
|
36
|
Sunday Driver/JIP3 binds kinesin heavy chain directly and enhances its motility. EMBO J 2011; 30:3416-29. [PMID: 21750526 DOI: 10.1038/emboj.2011.229] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 06/21/2011] [Indexed: 01/03/2023] Open
Abstract
Neuronal development, function and repair critically depend on axonal transport of vesicles and protein complexes, which is mediated in part by the molecular motor kinesin-1. Adaptor proteins recruit kinesin-1 to vesicles via direct association with kinesin heavy chain (KHC), the force-generating component, or via the accessory light chain (KLC). Binding of adaptors to the motor is believed to engage the motor for microtubule-based transport. We report that the adaptor protein Sunday Driver (syd, also known as JIP3 or JSAP1) interacts directly with KHC, in addition to and independently of its known interaction with KLC. Using an in vitro motility assay, we show that syd activates KHC for transport and enhances its motility, increasing both KHC velocity and run length. syd binding to KHC is functional in neurons, as syd mutants that bind KHC but not KLC are transported to axons and dendrites similarly to wild-type syd. This transport does not rely on syd oligomerization with itself or other JIP family members. These results establish syd as a positive regulator of kinesin activity and motility.
Collapse
|
37
|
Cai Q, Davis ML, Sheng ZH. Regulation of axonal mitochondrial transport and its impact on synaptic transmission. Neurosci Res 2011; 70:9-15. [PMID: 21352858 PMCID: PMC3086944 DOI: 10.1016/j.neures.2011.02.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/07/2011] [Accepted: 02/07/2011] [Indexed: 12/11/2022]
Abstract
Mitochondria are essential organelles for neuronal survival and play important roles in ATP generation, calcium buffering, and apoptotic signaling. Due to their extreme polarity, neurons utilize specialized mechanisms to regulate mitochondrial transport and retention along axons and near synaptic terminals where energy supply and calcium homeostasis are in high demand. Axonal mitochondria undergo saltatory and bidirectional movement and display complex mobility patterns. In cultured neurons, approximately one-third of axonal mitochondria are mobile, while the rest remain stationary. Stationary mitochondria at synapses serve as local energy stations that produce ATP to support synaptic function. In addition, axonal mitochondria maintain local Ca²+ homeostasis at presynaptic boutons. The balance between mobile and stationary mitochondria is dynamic and responds quickly to changes in axonal and synaptic physiology. The coordination of mitochondrial mobility and synaptic activity is crucial for neuronal function synaptic plasticity. In this update article, we introduce recent advances in our understanding of the motor-adaptor complexes and docking machinery that mediate mitochondrial transport and axonal distribution. We will also discuss the molecular mechanisms underlying the complex mobility patterns of axonal mitochondria and how mitochondrial mobility impacts the physiology and function of synapses.
Collapse
Affiliation(s)
- Qian Cai
- Synaptic Function Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 35, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| | - Matthew L. Davis
- Synaptic Function Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 35, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 35, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| |
Collapse
|
38
|
The Caenorhabditis elegans JIP3 protein UNC-16 functions as an adaptor to link kinesin-1 with cytoplasmic dynein. J Neurosci 2011; 31:2216-24. [PMID: 21307258 DOI: 10.1523/jneurosci.2653-10.2011] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Kinesin-1 is a microtubule plus-end-directed motor that transports various cargos along the axon. Previous studies have elucidated the physical and genetic interactions between kinesin-1 and cytoplasmic dynein, a microtubule minus-end-directed motor, in neuronal cells. However, the physiological importance of kinesin-1 in the dynein-dependent retrograde transport of cargo molecules remains obscure. Here, we show that Caenorhabditis elegans kinesin-1 forms a complex with dynein via its interaction with UNC-16, which binds to the dynein light intermediate (DLI) chain. Both kinesin-1 and UNC-16 are required for localization of DLI-1 at the plus ends of nerve process microtubules. In addition, retrograde transport of APL-1 depends on kinesin-1, UNC-16, and dynein. These results suggest that kinesin-1 mediates the anterograde transport of dynein using UNC-16 as a scaffold and that dynein in turn mediates the retrograde transport of cargo molecules in vivo. Thus, UNC-16 functions as an adaptor for kinesin-1-mediated transport of dynein.
Collapse
|
39
|
Alborghetti MR, Furlan AS, Kobarg J. FEZ2 has acquired additional protein interaction partners relative to FEZ1: functional and evolutionary implications. PLoS One 2011; 6:e17426. [PMID: 21408165 PMCID: PMC3050892 DOI: 10.1371/journal.pone.0017426] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 02/04/2011] [Indexed: 12/16/2022] Open
Abstract
Background The FEZ (fasciculation and elongation protein zeta) family designation was purposed by Bloom and Horvitz by genetic analysis of C. elegans unc-76. Similar human sequences were identified in the expressed sequence tag database as FEZ1 and FEZ2. The unc-76 function is necessary for normal axon fasciculation and is required for axon-axon interactions. Indeed, the loss of UNC-76 function results in defects in axonal transport. The human FEZ1 protein has been shown to rescue defects caused by unc-76 mutations in nematodes, indicating that both UNC-76 and FEZ1 are evolutionarily conserved in their function. Until today, little is known about FEZ2 protein function. Methodology/Principal Findings Using the yeast two-hybrid system we demonstrate here conserved evolutionary features among orthologs and non-conserved features between paralogs of the FEZ family of proteins, by comparing the interactome profiles of the C-terminals of human FEZ1, FEZ2 and UNC-76 from C. elegans. Furthermore, we correlate our data with an analysis of the molecular evolution of the FEZ protein family in the animal kingdom. Conclusions/Significance We found that FEZ2 interacted with 59 proteins and that of these only 40 interacted with FEZ1. Of the 40 FEZ1 interacting proteins, 36 (90%), also interacted with UNC-76 and none of the 19 FEZ2 specific proteins interacted with FEZ1 or UNC-76. This together with the duplication of unc-76 gene in the ancestral line of chordates suggests that FEZ2 is in the process of acquiring new additional functions. The results provide also an explanation for the dramatic difference between C. elegans and D. melanogaster unc-76 mutants on one hand, which cause serious defects in the nervous system, and the mouse FEZ1 -/- knockout mice on the other, which show no morphological and no strong behavioural phenotype. Likely, the ubiquitously expressed FEZ2 can completely compensate the lack of neuronal FEZ1, since it can interact with all FEZ1 interacting proteins and additional 19 proteins.
Collapse
Affiliation(s)
- Marcos R. Alborghetti
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil
| | - Ariane S. Furlan
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil
| | - Jörg Kobarg
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil
- Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil
- * E-mail:
| |
Collapse
|
40
|
Kinesin I transports tetramerized Kv3 channels through the axon initial segment via direct binding. J Neurosci 2010; 30:15987-6001. [PMID: 21106837 DOI: 10.1523/jneurosci.3565-10.2010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Precise targeting of various voltage-gated ion channels to proper membrane domains is crucial for their distinct roles in neuronal excitability and synaptic transmission. How each channel protein is transported within the cytoplasm is poorly understood. Here, we report that KIF5/kinesin I transports Kv3.1 voltage-gated K(+) (Kv) channels through the axon initial segment (AIS) via direct binding. First, we have identified a novel interaction between Kv3.1 and KIF5, confirmed by immunoprecipitation from mouse brain lysates and by pull-down assays with exogenously expressed proteins. The interaction is mediated by a direct binding between the Kv3.1 N-terminal T1 domain and a conserved region in KIF5 tail domains, in which proper T1 tetramerization is crucial. Overexpression of this region of KIF5B markedly reduces axonal levels of Kv3.1bHA. In mature hippocampal neurons, endogenous Kv3.1b and KIF5 colocalize. Suppressing the endogenous KIF5B level by RNA interference significantly reduces the Kv3.1b axonal level. Furthermore, mutating the Zn(2+)-binding site within T1 markedly decreases channel axonal targeting and forward trafficking, likely through disrupting T1 tetramerization and hence eliminating the binding to KIF5 tail. The mutation also alters channel activity. Interestingly, coexpression of the YFP (yellow fluorescent protein)-tagged KIF5B assists dendritic Kv3.1a and even mutants with a faulty axonal targeting motif to penetrate the AIS. Finally, fluorescently tagged Kv3.1 channels colocalize and comove with KIF5B along axons revealed by two-color time-lapse imaging. Our findings suggest that the binding to KIF5 ensures properly assembled and functioning Kv3.1 channels to be transported into axons.
Collapse
|
41
|
Loiseau P, Davies T, Williams LS, Mishima M, Palacios IM. Drosophila PAT1 is required for Kinesin-1 to transport cargo and to maximize its motility. Development 2010; 137:2763-72. [PMID: 20630947 DOI: 10.1242/dev.048108] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Kinesin heavy chain (KHC), the force-generating component of Kinesin-1, is required for the localization of oskar mRNA and the anchoring of the nucleus in the Drosophila oocyte. These events are crucial for the establishment of the anterior-posterior and dorsal-ventral axes. KHC is also essential for the localization of Dynein and for all ooplasmic flows. Interestingly, oocytes without Kinesin light chain show no major defects in these KHC-dependent processes, suggesting that KHC binds its cargoes and is activated by a novel mechanism. Here, we shed new light on the molecular mechanism of Kinesin function in the germline. Using a combination of genetic, biochemical and motor-tracking studies, we show that PAT1, an APP-binding protein, interacts with Kinesin-1, functions in the transport of oskar mRNA and Dynein and is required for the efficient motility of KHC along microtubules. This work suggests that the role of PAT1 in cargo transport in the cell is linked to PAT1 function as a positive regulator of Kinesin motility.
Collapse
|
42
|
Abstract
Intracellular transport is fundamental for cellular function, survival and morphogenesis. Kinesin superfamily proteins (also known as KIFs) are important molecular motors that directionally transport various cargos, including membranous organelles, protein complexes and mRNAs. The mechanisms by which different kinesins recognize and bind to specific cargos, as well as how kinesins unload cargo and determine the direction of transport, have now been identified. Furthermore, recent molecular genetic experiments have uncovered important and unexpected roles for kinesins in the regulation of such physiological processes as higher brain function, tumour suppression and developmental patterning. These findings open exciting new areas of kinesin research.
Collapse
|
43
|
Nieratschker V, Schubert A, Jauch M, Bock N, Bucher D, Dippacher S, Krohne G, Asan E, Buchner S, Buchner E. Bruchpilot in ribbon-like axonal agglomerates, behavioral defects, and early death in SRPK79D kinase mutants of Drosophila. PLoS Genet 2009; 5:e1000700. [PMID: 19851455 PMCID: PMC2759580 DOI: 10.1371/journal.pgen.1000700] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Accepted: 09/23/2009] [Indexed: 12/18/2022] Open
Abstract
Defining the molecular structure and function of synapses is a central theme in brain research. In Drosophila the Bruchpilot (BRP) protein is associated with T-shaped ribbons ("T-bars") at presynaptic active zones (AZs). BRP is required for intact AZ structure and normal evoked neurotransmitter release. By screening for mutations that affect the tissue distribution of Bruchpilot, we have identified a P-transposon insertion in gene CG11489 (location 79D) which shows high homology to mammalian genes for SR protein kinases (SRPKs). SRPKs phosphorylate serine-arginine rich splicing factors (SR proteins). Since proteins expressed from CG11489 cDNAs phosphorylate a peptide from a human SR protein in vitro, we name CG11489 the Drosophila Srpk79D gene. We have characterized Srpk79D transcripts and generated a null mutant. Mutation of the Srpk79D gene causes conspicuous accumulations of BRP in larval and adult nerves. At the ultrastructural level, these correspond to extensive axonal agglomerates of electron-dense ribbons surrounded by clear vesicles. Basic synaptic structure and function at larval neuromuscular junctions appears normal, whereas life expectancy and locomotor behavior of adult mutants are significantly impaired. All phenotypes of the mutant can be largely or completely rescued by panneural expression of SRPK79D isoforms. Isoform-specific antibodies recognize panneurally overexpressed GFP-tagged SRPK79D-PC isoform co-localized with BRP at presynaptic active zones while the tagged -PB isoform is found in spots within neuronal perikarya. SRPK79D concentrations in wild type apparently are too low to be revealed by these antisera. We propose that the Drosophila Srpk79D gene characterized here may be expressed at low levels throughout the nervous system to prevent the assembly of BRP containing agglomerates in axons and maintain intact brain function. The discovery of an SR protein kinase required for normal BRP distribution calls for the identification of its substrate and the detailed analysis of SRPK function for the maintenance of nervous system integrity.
Collapse
Affiliation(s)
- Vanessa Nieratschker
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Alice Schubert
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Mandy Jauch
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Nicole Bock
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Daniel Bucher
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Sonja Dippacher
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University, Würzburg, Germany
| | - Georg Krohne
- Department of Electron Microscopy, Julius-Maximilians-University, Würzburg, Germany
| | - Esther Asan
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University, Würzburg, Germany
| | - Sigrid Buchner
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Erich Buchner
- Department of Genetics and Neurobiology, Julius-Maximilians-University, Würzburg, Germany
| |
Collapse
|
44
|
The brain-specific factor FEZ1 is a determinant of neuronal susceptibility to HIV-1 infection. Proc Natl Acad Sci U S A 2009; 106:14040-5. [PMID: 19667186 DOI: 10.1073/pnas.0900502106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neurons are one of the few cell types in the human body that do not support HIV type-1 (HIV-1) replication. Although the lack of key receptors is a major obstacle to infection, studies suggest that additional functions inhibit virus replication to explain the exquisite resistance of neurons to HIV-1. However, specific neuronal factors that may explain this resistance remain to be discovered. In a screen for antiviral factors using a fibroblast line chemically mutagenized and selected for resistance to retroviral infection, we recently identified induction of rat FEZ1 (fasciculation and elongation protein zeta-1), a brain-specific protein, as the cause of this resistance. When exogenously expressed in nonneuronal cell lines rat FEZ1 blocked nuclear entry of retroviral DNA. Here, we demonstrate that among human brain cells, neurons naturally express high levels of FEZ1 compared to astrocytes or microglia cells and are correspondingly less susceptible to infection with pseudotyped HIV-1 that bypasses receptor-mediated viral entry. Demonstrating that endogenous FEZ1 was functionally important in the resistance of neurons to HIV-1 infection, siRNA-mediated knockdown of endogenous FEZ1 increased the infectivity of neurons while sensitive brain cell types like microglia became more resistant upon FEZ1 overexpression. In addition, FEZ1 expression was not induced in response to IFN treatment. As such, in contrast to other widely expressed, IFN-inducible antiviral factors, FEZ1 appears to represent a unique neuron-specific determinant of cellular susceptibility to infection in a cell type that is naturally resistant to HIV-1.
Collapse
|
45
|
Sann S, Wang Z, Brown H, Jin Y. Roles of endosomal trafficking in neurite outgrowth and guidance. Trends Cell Biol 2009; 19:317-24. [PMID: 19540123 DOI: 10.1016/j.tcb.2009.05.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 05/04/2009] [Accepted: 05/07/2009] [Indexed: 02/06/2023]
Abstract
Membrane trafficking and cargo delivery are essential for axonal and dendritic growth and guidance. Neurons have numerous diverse post-Golgi vesicles and recent advances have clarified their identity and regulation. Combinatorial approaches using in vivo imaging of 'intracellular cargo address labels' and functional perturbation have provided insight into these processes. In particular, the UNC-51 kinase regulates the trafficking of early endosomes and their axon guidance molecular cargos in several types of neurons in multiple organisms. Vesicular compartments bearing features of recycling endosomes, late endosomes or lysosomes also contribute to membrane addition and protein trafficking during neurite outgrowth and extension. New work shows that ubiquitylation of cargos and Rab effectors further specifies the trafficking routes of post-Golgi vesicles. These findings have begun to provide a more detailed view of the molecular mechanisms involved in neurite outgrowth and guidance. Additionally, high-resolution light microscopy imaging promises greater temporal and spatial understanding of vesicular exchange and maturation in neurons in the near future.
Collapse
Affiliation(s)
- Sharon Sann
- Division of Biological Sciences, University of California, San Diego, CA 92093, USA.
| | | | | | | |
Collapse
|
46
|
Hackney DD, Baek N, Snyder AC. Half-site inhibition of dimeric kinesin head domains by monomeric tail domains. Biochemistry 2009; 48:3448-56. [PMID: 19320433 DOI: 10.1021/bi8022575] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The two heavy chains of kinesin-1 are dimerized through extensive coiled coil regions and fold into an inactive conformation through interaction of the C-terminal tail domains with the N-terminal motor (head) domains. Although this potentially allows a dimer of tail domains to interact symmetrically with a dimer of head domains, we report here that only one of the two available monomeric tail peptides is sufficient for tight binding and inhibition of a dimer of head domains. With a dimeric tail construct, the other tail peptide does not make tight contact with the head dimer and can bind a second head dimer to form a complex containing one tail dimer and two head dimers. The IAK domain and neighboring positively charged region of the tail is sufficient for tight half-site interaction with a dimer of heads. The interaction of tails with monomeric heads is weak, but a head dimer produced by the dimerization of the neck coil is not required because an artificial dimer of head domains also binds monomeric tail peptides with half-site stoichiometry in the complete absence of the native neck coil. The binding of tail peptides to head dimers is fast and readily reversible as determined by FRET between mant-ADP bound to the head dimer and a tail labeled with GFP. The association and dissociation rates are 81 microM(-1) s(-1) and 32 s(-1), respectively. This half-site interaction suggests that the second tail peptide in a folded kinesin-1 might be available to bind other molecules while kinesin-1 remained folded.
Collapse
Affiliation(s)
- David D Hackney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA.
| | | | | |
Collapse
|
47
|
Toda H, Mochizuki H, Flores R, Josowitz R, Krasieva TB, Lamorte VJ, Suzuki E, Gindhart JG, Furukubo-Tokunaga K, Tomoda T. UNC-51/ATG1 kinase regulates axonal transport by mediating motor-cargo assembly. Genes Dev 2009; 22:3292-307. [PMID: 19056884 DOI: 10.1101/gad.1734608] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Axonal transport mediated by microtubule-dependent motors is vital for neuronal function and viability. Selective sets of cargoes, including macromolecules and organelles, are transported long range along axons to specific destinations. Despite intensive studies focusing on the motor machinery, the regulatory mechanisms that control motor-cargo assembly are not well understood. Here we show that UNC-51/ATG1 kinase regulates the interaction between synaptic vesicles and motor complexes during transport in Drosophila. UNC-51 binds UNC-76, a kinesin heavy chain (KHC) adaptor protein. Loss of unc-51 or unc-76 leads to severe axonal transport defects in which synaptic vesicles are segregated from the motor complexes and accumulate along axons. Genetic studies show that unc-51 and unc-76 functionally interact in vivo to regulate axonal transport. UNC-51 phosphorylates UNC-76 on Ser(143), and the phosphorylated UNC-76 binds Synaptotagmin-1, a synaptic vesicle protein, suggesting that motor-cargo interactions are regulated in a phosphorylation-dependent manner. In addition, defective axonal transport in unc-76 mutants is rescued by a phospho-mimetic UNC-76, but not a phospho-defective UNC-76, demonstrating the essential role of UNC-76 Ser(143) phosphorylation in axonal transport. Thus, our data provide insight into axonal transport regulation that depends on the phosphorylation of adaptor proteins.
Collapse
Affiliation(s)
- Hirofumi Toda
- Division of Neurosciences, Beckman Research Institute of the City of Hope, California 91010, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Axonal transport and the delivery of pre-synaptic components. Curr Opin Neurobiol 2008; 18:495-503. [PMID: 18950710 DOI: 10.1016/j.conb.2008.10.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 09/30/2008] [Accepted: 10/05/2008] [Indexed: 12/30/2022]
Abstract
The mechanisms for delivering components to nerve terminals are diverse and highly regulated. The diversity of kinesin motors alone is insufficient to account for the specificity of delivery. Additional specificity and control are contributed by adaptor proteins and associated regulatory molecules. The interaction of cargos with these complexes can confer distinct behaviors on the transport of synaptic organelles. The rich regulatory mechanisms of transport that are only now emerging as the cargo-motor complexes are defined and subsequent local events that regulate their dynamic relationship are examined. Here we review recent studies of kinesin-related axonal transport of three crucial synaptic components, Piccolo-bassoon Transport Vesicles (PTVs), Synaptic Vesicle Precursors (SVPs), and mitochondria, and the mechanisms that modulate their transport.
Collapse
|
49
|
Sakae N, Yamasaki N, Kitaichi K, Fukuda T, Yamada M, Yoshikawa H, Hiranita T, Tatsumi Y, Kira JI, Yamamoto T, Miyakawa T, Nakayama KI. Mice lacking the schizophrenia-associated protein FEZ1 manifest hyperactivity and enhanced responsiveness to psychostimulants. Hum Mol Genet 2008; 17:3191-203. [PMID: 18647754 DOI: 10.1093/hmg/ddn215] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
FEZ1 (fasciculation and elongation protein zeta 1), a mammalian ortholog of Caenorhabditis elegans UNC-76, interacts with DISC1 (disrupted in schizophrenia 1), a schizophrenia susceptibility gene product, and polymorphisms of human FEZ1 have been associated with schizophrenia. We have now investigated the role of FEZ1 in brain development and the pathogenesis of schizophrenia by generating mice that lack Fez1. Immunofluorescence staining revealed FEZ1 to be located predominantly in gamma-aminobutyric acid-containing interneurons. The Fez1(-/-) mice showed marked hyperactivity in a variety of behavioral tests as well as enhanced behavioral responses to the psychostimulants MK-801 and methamphetamine. In vivo microdialysis revealed that the methamphetamine-induced release of dopamine in the nucleus accumbens was exaggerated in the mutant mice, suggesting that enhanced mesolimbic dopaminergic transmission contributes to their hyperactivity phenotype. These observations implicate impairment of FEZ1 function in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Nobutaka Sakae
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-2-2 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hirokawa N, Noda Y. Intracellular Transport and Kinesin Superfamily Proteins, KIFs: Structure, Function, and Dynamics. Physiol Rev 2008; 88:1089-118. [DOI: 10.1152/physrev.00023.2007] [Citation(s) in RCA: 345] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Various molecular cell biology and molecular genetic approaches have indicated significant roles for kinesin superfamily proteins (KIFs) in intracellular transport and have shown that they are critical for cellular morphogenesis, functioning, and survival. KIFs not only transport various membrane organelles, protein complexes, and mRNAs for the maintenance of basic cellular activity, but also play significant roles for various mechanisms fundamental for life, such as brain wiring, higher brain functions such as memory and learning and activity-dependent neuronal survival during brain development, and for the determination of important developmental processes such as left-right asymmetry formation and suppression of tumorigenesis. Accumulating data have revealed a molecular mechanism of cargo recognition involving scaffolding or adaptor protein complexes. Intramolecular folding and phosphorylation also regulate the binding activity of motor proteins. New techniques using molecular biophysics, cryoelectron microscopy, and X-ray crystallography have detected structural changes in motor proteins, synchronized with ATP hydrolysis cycles, leading to the development of independent models of monomer and dimer motors for processive movement along microtubules.
Collapse
|