1
|
Nakagawa N. The neuronal Golgi in neural circuit formation and reorganization. Front Neural Circuits 2024; 18:1504422. [PMID: 39703196 PMCID: PMC11655203 DOI: 10.3389/fncir.2024.1504422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The Golgi apparatus is a central hub in the intracellular secretory pathway. By positioning in the specific intracellular region and transporting materials to spatially restricted compartments, the Golgi apparatus contributes to the cell polarity establishment and morphological specification in diverse cell types. In neurons, the Golgi apparatus mediates several essential steps of initial neural circuit formation during early brain development, such as axon-dendrite polarization, neuronal migration, primary dendrite specification, and dendritic arbor elaboration. Moreover, neuronal activity-dependent remodeling of the Golgi structure enables morphological changes in neurons, which provides the cellular basis of circuit reorganization during postnatal critical period. In this review, I summarize recent findings illustrating the unique Golgi positioning and its developmental dynamics in various types of neurons. I also discuss the upstream regulators for the Golgi positioning in neurons, and functional roles of the Golgi in neural circuit formation and reorganization. Elucidating how Golgi apparatus sculpts neuronal connectivity would deepen our understanding of the cellular/molecular basis of neural circuit development and plasticity.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Japan
| |
Collapse
|
2
|
Sousa SC, Aroso M, Bessa R, Veríssimo E, Ferreira da Silva T, Lopes CDF, Brites P, Vieira J, Vieira CP, Aguiar PC, Sousa MM. Stretch triggers microtubule stabilization and MARCKS-dependent membrane incorporation in the shaft of embryonic axons. Curr Biol 2024; 34:4577-4588.e8. [PMID: 39265571 DOI: 10.1016/j.cub.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/28/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Neurons have a unique polarized nature that must adapt to environmental changes throughout their lifespan. During embryonic development, axon elongation is led by the growth cone,1 culminating in the formation of a presynaptic terminal. After synapses are formed, axons elongate in a growth cone-independent manner to accompany body growth while maintaining their ultrastructure and function.2,3,4,5,6 To further understand mechanical strains on the axon shaft, we developed a computer-controlled stretchable microfluidic platform compatible with multi-omics and live imaging. Our data show that sensory embryonic dorsal root ganglia (DRGs) neurons have high plasticity, with axon shaft microtubules decreasing polymerization rates, aligning with the direction of tension, and undergoing stabilization. Moreover, in embryonic DRGs, stretch triggers yes-associated protein (YAP) nuclear translocation, supporting its participation in the regulatory network that enables tension-driven axon growth. Other than cytoskeleton remodeling, stretch prompted MARCKS-dependent formation of plasmalemmal precursor vesicles (PPVs), resulting in new membrane incorporation throughout the axon shaft. In contrast, adolescent DRGs showed a less robust adaptation, with axonal microtubules being less responsive to stretch. Also, while adolescent DRGs were still amenable to strain-induced PPV formation at higher stretch rates, new membrane incorporation in the axon shaft failed to occur. In summary, we developed a new resource to study the biology of axon stretch growth. By unraveling cytoskeleton adaptation and membrane remodeling in the axon shaft of stretched neurons, we are moving forward in understanding axon growth.
Collapse
Affiliation(s)
- Sara C Sousa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; Graduate Program in Molecular and Cell Biology, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Miguel Aroso
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Rita Bessa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Eduardo Veríssimo
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; Graduate Program in Molecular and Cell Biology, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Tiago Ferreira da Silva
- Neurolipid Biology Group, IBMC-Instituto de Biologia Celular e Molecular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Cátia D F Lopes
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Pedro Brites
- Neurolipid Biology Group, IBMC-Instituto de Biologia Celular e Molecular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Jorge Vieira
- Phenotypic Evolution Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Cristina P Vieira
- Phenotypic Evolution Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Paulo C Aguiar
- Neuroengineering and Computational Neuroscience Group, i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Monica M Sousa
- Nerve Regeneration Group, IBMC-Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
3
|
Li Y, Wen Y, Li Y, Tan X, Gao S, Fan P, Tian W, Wong CC, Chen Y. Rab10-CAV1 mediated intraluminal vesicle transport to migrasomes. Proc Natl Acad Sci U S A 2024; 121:e2319267121. [PMID: 39008679 PMCID: PMC11287133 DOI: 10.1073/pnas.2319267121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 06/12/2024] [Indexed: 07/17/2024] Open
Abstract
Migrasomes, vesicular organelles generated on the retraction fibers of migrating cells, play a crucial role in migracytosis, mediating intercellular communication. The cargoes determine the functional specificity of migrasomes. Migrasomes harbor numerous intraluminal vesicles, a pivotal component of their cargoes. The mechanism underlying the transportation of these intraluminal vesicles to the migrasomes remains enigmatic. In this study, we identified that Rab10 and Caveolin-1 (CAV1) mark the intraluminal vesicles in migrasomes. Transport of Rab10-CAV1 vesicles to migrasomes required the motor protein Myosin Va and adaptor proteins RILPL2. Notably, the phosphorylation of Rab10 by the kinase LRRK2 regulated this process. Moreover, CSF-1 can be transported to migrasomes through this mechanism, subsequently fostering monocyte-macrophage differentiation in skin wound healing, which served as a proof of the physiological importance of this transporting mechanism.
Collapse
Affiliation(s)
- Yong Li
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100084, China
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Yiling Wen
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Ying Li
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Xinyi Tan
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Shuaixin Gao
- Department of Human Sciences & James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Peiyao Fan
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Wenmin Tian
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| | - Catherine C.L. Wong
- Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100084, China
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing100730, China
| | - Yang Chen
- Center for Precision Medicine Multi-Omics Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing100191, China
| |
Collapse
|
4
|
Singh V, Menard MA, Serrano GE, Beach TG, Zhao HT, Riley-DiPaolo A, Subrahmanian N, LaVoie MJ, Volpicelli-Daley LA. Cellular and subcellular localization of Rab10 and phospho-T73 Rab10 in the mouse and human brain. Acta Neuropathol Commun 2023; 11:201. [PMID: 38110990 PMCID: PMC10726543 DOI: 10.1186/s40478-023-01704-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/20/2023] Open
Abstract
Autosomal dominant pathogenic mutations in Leucine-rich repeat kinase 2 (LRRK2) cause Parkinson's disease (PD). The most common mutation, G2019S-LRRK2, increases the kinase activity of LRRK2 causing hyper-phosphorylation of its substrates. One of these substrates, Rab10, is phosphorylated at a conserved Thr73 residue (pRab10), and is one of the most abundant LRRK2 Rab GTPases expressed in various tissues. The involvement of Rab10 in neurodegenerative disease, including both PD and Alzheimer's disease makes pinpointing the cellular and subcellular localization of Rab10 and pRab10 in the brain an important step in understanding its functional role, and how post-translational modifications could impact function. To establish the specificity of antibodies to the phosphorylated form of Rab10 (pRab10), Rab10 specific antisense oligonucleotides were intraventricularly injected into the brains of mice. Further, Rab10 knock out induced neurons, differentiated from human induced pluripotent stem cells were used to test the pRab10 antibody specificity. To amplify the weak immunofluorescence signal of pRab10, tyramide signal amplification was utilized. Rab10 and pRab10 were expressed in the cortex, striatum and the substantia nigra pars compacta. Immunofluorescence for pRab10 was increased in G2019S-LRRK2 knockin mice. Neurons, astrocytes, microglia and oligodendrocytes all showed Rab10 and pRab10 expression. While Rab10 colocalized with endoplasmic reticulum, lysosome and trans-Golgi network markers, pRab10 did not localize to these organelles. However, pRab10, did overlap with markers of the presynaptic terminal in both mouse and human cortex, including α-synuclein. Results from this study suggest Rab10 and pRab10 are expressed in all brain areas and cell types tested in this study, but pRab10 is enriched at the presynaptic terminal. As Rab10 is a LRRK2 kinase substrate, increased kinase activity of G2019S-LRRK2 in PD may affect Rab10 mediated membrane trafficking at the presynaptic terminal in neurons in disease.
Collapse
Affiliation(s)
- Vijay Singh
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Marissa A Menard
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Geidy E Serrano
- Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Thomas G Beach
- Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Hien T Zhao
- Ionis Pharmaceuticals Inc, Carlsbad, CA, 92010, USA
| | - Alexis Riley-DiPaolo
- Department of Neuroscience at the University of Florida, Gainesville, FL, 32611, USA
| | - Nitya Subrahmanian
- Department of Neurology, Center for Translational Research in Neurodegenerative Disease, Fixel Institute for Neurologic Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Matthew J LaVoie
- Department of Neurology, Center for Translational Research in Neurodegenerative Disease, Fixel Institute for Neurologic Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
5
|
Cason SE, Holzbaur EL. Axonal transport of autophagosomes is regulated by dynein activators JIP3/JIP4 and ARF/RAB GTPases. J Cell Biol 2023; 222:e202301084. [PMID: 37909920 PMCID: PMC10620608 DOI: 10.1083/jcb.202301084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/28/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Neuronal autophagosomes form and engulf cargos at presynaptic sites in the axon and are then transported to the soma to recycle their cargo. Autophagic vacuoles (AVs) mature en route via fusion with lysosomes to become degradatively competent organelles; transport is driven by the microtubule motor protein cytoplasmic dynein, with motor activity regulated by a sequential series of adaptors. Using lysate-based single-molecule motility assays and live-cell imaging in primary neurons, we show that JNK-interacting proteins 3 (JIP3) and 4 (JIP4) are activating adaptors for dynein that are regulated on autophagosomes and lysosomes by the small GTPases ARF6 and RAB10. GTP-bound ARF6 promotes formation of the JIP3/4-dynein-dynactin complex. Either knockdown or overexpression of RAB10 stalls transport, suggesting that this GTPase is also required to coordinate the opposing activities of bound dynein and kinesin motors. These findings highlight the complex coordination of motor regulation during organelle transport in neurons.
Collapse
Affiliation(s)
- Sydney E. Cason
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Erika L.F. Holzbaur
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Sasazawa Y, Hattori N, Saiki S. JNK-interacting protein 4 is a central molecule for lysosomal retrograde trafficking. Bioessays 2023; 45:e2300052. [PMID: 37559169 DOI: 10.1002/bies.202300052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
Lysosomal positioning is an important factor in regulating cellular responses, including autophagy. Because proteins encoded by disease-responsible genes are involved in lysosomal trafficking, proper intracellular lysosomal trafficking is thought to be essential for cellular homeostasis. In the past few years, the mechanisms of lysosomal trafficking have been elucidated with a focus on adapter proteins linking motor proteins to lysosomes. Here, we outline recent findings on the mechanisms of lysosomal trafficking by focusing on adapter protein c-Jun NH2 -terminal kinase-interacting protein (JIP) 4, which plays a central role in this process, and other JIP4 functions and JIP family proteins. Additionally, we discuss neuronal diseases associated with aberrance in the JIP family protein. Accumulating evidence suggests that chemical manipulation of lysosomal positioning may be a therapeutic approach for these neuronal diseases.
Collapse
Affiliation(s)
- Yukiko Sasazawa
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Neurology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Neurology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
7
|
Drozd CJ, Quinn CC. UNC-116 and UNC-16 function with the NEKL-3 kinase to promote axon targeting. Development 2023; 150:dev201654. [PMID: 37756604 PMCID: PMC10561693 DOI: 10.1242/dev.201654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
KIF5C is a kinesin-1 heavy chain that has been associated with neurodevelopmental disorders. Although the roles of kinesin-1 in axon transport are well known, little is known about how it regulates axon targeting. We report that UNC-116/KIF5C functions with the NEKL-3/NEK6/7 kinase to promote axon targeting in Caenorhabditis elegans. Loss of UNC-116 causes the axon to overshoot its target and UNC-116 gain-of-function causes premature axon termination. We find that loss of the UNC-16/JIP3 kinesin-1 cargo adaptor disrupts axon termination, but loss of kinesin-1 light chain function does not affect axon termination. Genetic analysis indicates that UNC-16 functions with the NEKL-3 kinase to promote axon termination. Consistent with this observation, imaging experiments indicate that loss of UNC-16 and UNC-116 disrupt localization of NEKL-3 in the axon. Moreover, genetic interactions suggest that NEKL-3 promotes axon termination by functioning with RPM-1, a ubiquitin ligase that regulates microtubule stability in the growth cone. These observations support a model where UNC-116 functions with UNC-16 to promote localization of NEKL-3 in the axon. NEKL-3, in turn, functions with the RPM-1 ubiquitin ligase to promote axon termination.
Collapse
Affiliation(s)
- Cody J. Drozd
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Christopher C. Quinn
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| |
Collapse
|
8
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Bunner W, Wang J, Cohen S, Bashtovyy D, Perry R, Shookster D, Landry T, Harris EM, Stackman R, Tran TD, Yasuda R, Szatmari EM. Behavioral and Transcriptome Profiling of Heterozygous Rab10 Knock-Out Mice. eNeuro 2023; 10:ENEURO.0459-22.2023. [PMID: 37156612 PMCID: PMC10208283 DOI: 10.1523/eneuro.0459-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 05/10/2023] Open
Abstract
A central question in the field of aging research is to identify the cellular and molecular basis of neuroresilience. One potential candidate is the small GTPase, Rab10. Here, we used Rab10+/- mice to investigate the molecular mechanisms underlying Rab10-mediated neuroresilience. Brain expression analysis of 880 genes involved in neurodegeneration showed that Rab10+/- mice have increased activation of pathways associated with neuronal metabolism, structural integrity, neurotransmission, and neuroplasticity compared with their Rab10+/+ littermates. Lower activation was observed for pathways involved in neuroinflammation and aging. We identified and validated several differentially expressed genes (DEGs), including Stx2, Stx1b, Vegfa, and Lrrc25 (downregulated) and Prkaa2, Syt4, and Grin2d (upregulated). Behavioral testing showed that Rab10+/- mice perform better in a hippocampal-dependent spatial task (object in place test), while their performance in a classical conditioning task (trace eyeblink classical conditioning, TECC) was significantly impaired. Therefore, our findings indicate that Rab10 differentially controls the brain circuitry of hippocampal-dependent spatial memory and higher-order behavior that requires intact cortex-hippocampal circuitry. Transcriptome and biochemical characterization of these mice suggest that glutamate ionotropic receptor NMDA type subunit 2D (GRIN2D or GluN2D) is affected by Rab10 signaling. Further work is needed to evaluate whether GRIN2D mediates the behavioral phenotypes of the Rab10+/- mice. We conclude that Rab10+/- mice described here can be a valuable tool to study the mechanisms of resilience in Alzheimer's disease (AD) model mice and to identify novel therapeutical targets to prevent cognitive decline associated with normal and pathologic aging.
Collapse
Affiliation(s)
- Wyatt Bunner
- Department of Physical Therapy, East Carolina University, Greenville, NC 27834
| | - Jie Wang
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458
| | - Sarah Cohen
- Jupiter Life Science Initiative, Florida Atlantic University, Jupiter, FL 33458
| | - Denys Bashtovyy
- Department of Physical Therapy, East Carolina University, Greenville, NC 27834
| | - Rachel Perry
- Department of Physical Therapy, East Carolina University, Greenville, NC 27834
| | | | - Taylor Landry
- Department of Kinesiology, East Carolina University, NC 27858
| | - Elizabeth M Harris
- Department of Psychology, East Carolina University, Greenville, NC 27858
| | - Robert Stackman
- Jupiter Life Science Initiative, Florida Atlantic University, Jupiter, FL 33458
| | - Tuan D Tran
- Department of Psychology, East Carolina University, Greenville, NC 27858
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458
| | - Erzsebet M Szatmari
- Department of Physical Therapy, East Carolina University, Greenville, NC 27834
| |
Collapse
|
10
|
Zhang J, Li J, You P, Jiang H, Liu Y, Han D, Liu M, Yu H, Su B. Mice with the Rab10 T73V mutation exhibit anxiety-like behavior and alteration of neuronal functions in the striatum. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166641. [PMID: 36669576 DOI: 10.1016/j.bbadis.2023.166641] [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: 09/09/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/19/2023]
Abstract
Hyperphosphorylated Rab10 has been implicated in the pathogenesis of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. However, the neurophysiological function of the evolutionarily conserved Thr73 phosphorylation of Rab10 remains poorly understood. Here, we generated a novel mouse model expressing the non-phosphorylatable T73V mutation of Rab10 and performed a comprehensive series of neurological analyses, including behavioral tests, synaptic evaluations, neuronal and glial staining, assessments of neurite arborization and spine morphogenesis. The Rab10 T73V mutantmice exhibited a characteristic anxiety-like phenotype with other behavioral modules relatively unaffected. Moreover, Rab10 T73V mutant mice displayed striatum-specific synaptic dysfunction, as indicated by aberrantly increased expression levels of synaptic proteins and impaired frequencies of miniature inhibitory postsynaptic currents. The genetic deletion of Rab10 phosphorylation enhanced neurite arborization and accelerated spine maturation in striatal medium spiny neurons. Our findings emphasize the specific role of intrinsic phospho-Rab10 in the regulation of the striatal circuitry and its related behaviors.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Li
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Pan You
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Haitian Jiang
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yanjun Liu
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Daobin Han
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Meiqi Liu
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hui Yu
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Bo Su
- Department of Cell Biology, Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
11
|
Lazo OM, Schiavo G. Rab10 regulates the sorting of internalised TrkB for retrograde axonal transport. eLife 2023; 12:81532. [PMID: 36897066 PMCID: PMC10005780 DOI: 10.7554/elife.81532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 02/15/2023] [Indexed: 03/11/2023] Open
Abstract
Neurons process real-time information from axon terminals to coordinate gene expression, growth, and plasticity. Inputs from distal axons are encoded as a stream of endocytic organelles, termed signalling endosomes, targeted to the soma. Formation of these organelles depends on target-derived molecules, such as brain-derived neurotrophic factor (BDNF), which is recognised by TrkB receptors on the plasma membrane, endocytosed, and transported to the cell body along the microtubules network. Notwithstanding its physiological and neuropathological importance, the mechanism controlling the sorting of TrkB to signalling endosomes is currently unknown. In this work, we use primary mouse neurons to uncover the small GTPase Rab10 as critical for TrkB sorting and propagation of BDNF signalling from axon terminals to the soma. Our data demonstrate that Rab10 defines a novel membrane compartment that is rapidly mobilised towards the axon terminal upon BDNF stimulation, enabling the axon to fine-tune retrograde signalling depending on BDNF availability at the synapse. These results help clarifying the neuroprotective phenotype recently associated to Rab10 polymorphisms in Alzheimer's disease and provide a new therapeutic target to halt neurodegeneration.
Collapse
Affiliation(s)
- Oscar Marcelo Lazo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| |
Collapse
|
12
|
Cason SE, Holzbaur EL. Axonal transport of autophagosomes is regulated by dynein activators JIP3/JIP4 and ARF/RAB GTPases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.526044. [PMID: 36747648 PMCID: PMC9901177 DOI: 10.1101/2023.01.28.526044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Neuronal autophagosomes, "self-eating" degradative organelles, form at presynaptic sites in the distal axon and are transported to the soma to recycle their cargo. During transit, autophagic vacuoles (AVs) mature through fusion with lysosomes to acquire the enzymes necessary to breakdown their cargo. AV transport is driven primarily by the microtubule motor cytoplasmic dynein in concert with dynactin and a series of activating adaptors that change depending on organelle maturation state. The transport of mature AVs is regulated by the scaffolding proteins JIP3 and JIP4, both of which activate dynein motility in vitro. AV transport is also regulated by ARF6 in a GTP-dependent fashion. While GTP-bound ARF6 promotes the formation of the JIP3/4-dynein-dynactin complex, RAB10 competes with the activity of this complex by increasing kinesin recruitment to axonal AVs and lysosomes. These interactions highlight the complex coordination of motors regulating organelle transport in neurons.
Collapse
Affiliation(s)
- Sydney E. Cason
- Department of Physiology, University of Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania
- Pennsylvania Muscle Institute, University of Pennsylvania
| | - Erika L.F. Holzbaur
- Department of Physiology, University of Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania
- Pennsylvania Muscle Institute, University of Pennsylvania
| |
Collapse
|
13
|
Transport-dependent maturation of organelles in neurons. Curr Opin Cell Biol 2022; 78:102121. [PMID: 36030563 DOI: 10.1016/j.ceb.2022.102121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
Some organelles show a spatial gradient of maturation along the neuronal process where more mature organelles are found closer to the cell body. This gradient is set up by progressive maturation steps that are aided by differential organelle distribution as well as transport. Autophagosomes and endosomes mature as they acquire lysosomal membrane proteins and decrease their luminal pH as they are retrogradely transported towards the cell body. The acquisition of lysosomal proteins along the neuronal processes likely occurs through fusion or membrane exchange events with Golgi-derived donor transport carriers that are transported anterogradely from the cell body. The mechanisms by which endosomes and autophagosomes mature might be applicable to other organelles that are transported along neuronal processes. Defects in axonal transport may also contribute to the accumulation of immature organelles in neurons. Such accumulations have been seen in neurons of neurodegenerative models.
Collapse
|
14
|
Zajac AL, Horne-Badovinac S. Kinesin-directed secretion of basement membrane proteins to a subdomain of the basolateral surface in Drosophila epithelial cells. Curr Biol 2022; 32:735-748.e10. [PMID: 35021047 PMCID: PMC8891071 DOI: 10.1016/j.cub.2021.12.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 11/23/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022]
Abstract
Epithelial tissues are lined with a sheet-like basement membrane (BM) extracellular matrix at their basal surfaces that plays essential roles in adhesion and signaling. BMs also provide mechanical support to guide morphogenesis. Despite their importance, we know little about how epithelial cells secrete and assemble BMs during development. BM proteins are sorted into a basolateral secretory pathway distinct from other basolateral proteins. Because BM proteins self-assemble into networks, and the BM lines only a small portion of the basolateral domain, we hypothesized that the site of BM protein secretion might be tightly controlled. Using the Drosophila follicular epithelium, we show that kinesin-3 and kinesin-1 motors work together to define this secretion site. Similar to all epithelia, the follicle cells have polarized microtubules (MTs) along their apical-basal axes. These cells collectively migrate, and they also have polarized MTs along the migratory axis at their basal surfaces. We find follicle cell MTs form one interconnected network, which allows kinesins to transport Rab10+ BM secretory vesicles both basally and to the trailing edge of each cell. This positions them near the basal surface and the basal-most region of the lateral domain for exocytosis. When kinesin transport is disrupted, the site of BM protein secretion is expanded, and ectopic BM networks form between cells that impede migration and disrupt tissue architecture. These results show how epithelial cells can define a subdomain on their basolateral surface through MT-based transport and highlight the importance of controlling the exocytic site of network-forming proteins.
Collapse
Affiliation(s)
- Allison L. Zajac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA.
| |
Collapse
|
15
|
Atkins M, Hazan J, Fassier C. In Vivo Live Imaging of Axonal Transport in Developing Zebrafish Axons. Methods Mol Biol 2022; 2431:325-350. [PMID: 35412285 DOI: 10.1007/978-1-0716-1990-2_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Axonal transport is crucial for neuronal homeostasis, survival, and development. Indeed, axonal transport needs to be precisely regulated for developing axons to swiftly and accurately respond to their complex and evolving environment in space and time. A growing number of studies have started to unravel the diversity of regulatory and adaptor proteins required to orchestrate the axonal transport machinery. Despite some discrepancies between in vitro and in vivo axonal transport studies, most analyses aiming at deciphering these regulatory complexes, as well as their mode of action, were carried out in vitro in primary cultures of neurons, and mainly focused on their impact on axon specification and elongation, but rarely on axon navigation per se. Given the clear influence of the in vivo environment on axonal transport, including chemical and physical interactions with neighboring cells, it is essential to develop in vivo models to identify and characterize the molecular complexes involved in this key process. Here, we describe an experimental system to monitor axonal transport in vivo in developing axons of live zebrafish embryos with high spatial and temporal resolution. Due to its optical transparency and easy genetic manipulation, the zebrafish embryo is ideally suited to study such cellular dynamics at a single axon scale. Using this approach, we were able to unravel the key role of Fidgetin-like 1 in the regulation of bidirectional axonal transport required for motor axon targeting. Moreover, this protocol can be easily adapted to characterize a wide range of axonal transport regulators and components in physiological conditions and may additionally be used to screen new therapeutic compounds based on their ability to recue axonal transport defects in pathological conditions.
Collapse
Affiliation(s)
- Melody Atkins
- Sorbonne Université, UPMC-Université Paris 6, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine-Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France
- INSERM, UMR-S 1270, Institut du Fer à Moulin, UMR-S 1270 Sorbonne Université, Paris, France
| | - Jamilé Hazan
- Sorbonne Université, UPMC-Université Paris 6, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine-Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France
| | - Coralie Fassier
- Sorbonne Université, UPMC-Université Paris 6, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine-Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France.
- Sorbonne Université, INSERM UMR_S 968, CNRS UMR_7210, Institut de la Vision, Paris, France.
| |
Collapse
|
16
|
Intertwined and Finely Balanced: Endoplasmic Reticulum Morphology, Dynamics, Function, and Diseases. Cells 2021; 10:cells10092341. [PMID: 34571990 PMCID: PMC8472773 DOI: 10.3390/cells10092341] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is an organelle that is responsible for many essential subcellular processes. Interconnected narrow tubules at the periphery and thicker sheet-like regions in the perinuclear region are linked to the nuclear envelope. It is becoming apparent that the complex morphology and dynamics of the ER are linked to its function. Mutations in the proteins involved in regulating ER structure and movement are implicated in many diseases including neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and amyotrophic lateral sclerosis (ALS). The ER is also hijacked by pathogens to promote their replication. Bacteria such as Legionella pneumophila and Chlamydia trachomatis, as well as the Zika virus, bind to ER morphology and dynamics-regulating proteins to exploit the functions of the ER to their advantage. This review covers our understanding of ER morphology, including the functional subdomains and membrane contact sites that the organelle forms. We also focus on ER dynamics and the current efforts to quantify ER motion and discuss the diseases related to ER morphology and dynamics.
Collapse
|
17
|
Le Guen Y, Belloy ME, Napolioni V, Eger SJ, Kennedy G, Tao R, He Z, Greicius MD. A novel age-informed approach for genetic association analysis in Alzheimer's disease. Alzheimers Res Ther 2021; 13:72. [PMID: 33794991 PMCID: PMC8017764 DOI: 10.1186/s13195-021-00808-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Many Alzheimer's disease (AD) genetic association studies disregard age or incorrectly account for it, hampering variant discovery. METHODS Using simulated data, we compared the statistical power of several models: logistic regression on AD diagnosis adjusted and not adjusted for age; linear regression on a score integrating case-control status and age; and multivariate Cox regression on age-at-onset. We applied these models to real exome-wide data of 11,127 sequenced individuals (54% cases) and replicated suggestive associations in 21,631 genotype-imputed individuals (51% cases). RESULTS Modeling variable AD risk across age results in 5-10% statistical power gain compared to logistic regression without age adjustment, while incorrect age adjustment leads to critical power loss. Applying our novel AD-age score and/or Cox regression, we discovered and replicated novel variants associated with AD on KIF21B, USH2A, RAB10, RIN3, and TAOK2 genes. CONCLUSION Our AD-age score provides a simple means for statistical power gain and is recommended for future AD studies.
Collapse
Affiliation(s)
- Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA.
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Sarah J Eger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Gabriel Kennedy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Ran Tao
- Department of Biostatistics and Vanderbilt Genetic Institute, Vanderbilt University, Nashville, TN, 37203, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94304, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| |
Collapse
|
18
|
Axonal Organelles as Molecular Platforms for Axon Growth and Regeneration after Injury. Int J Mol Sci 2021; 22:ijms22041798. [PMID: 33670312 PMCID: PMC7918155 DOI: 10.3390/ijms22041798] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Investigating the molecular mechanisms governing developmental axon growth has been a useful approach for identifying new strategies for boosting axon regeneration after injury, with the goal of treating debilitating conditions such as spinal cord injury and vision loss. The picture emerging is that various axonal organelles are important centers for organizing the molecular mechanisms and machinery required for growth cone development and axon extension, and these have recently been targeted to stimulate robust regeneration in the injured adult central nervous system (CNS). This review summarizes recent literature highlighting a central role for organelles such as recycling endosomes, the endoplasmic reticulum, mitochondria, lysosomes, autophagosomes and the proteasome in developmental axon growth, and describes how these organelles can be targeted to promote axon regeneration after injury to the adult CNS. This review also examines the connections between these organelles in developing and regenerating axons, and finally discusses the molecular mechanisms within the axon that are required for successful axon growth.
Collapse
|
19
|
Saez TMM, Fernandez Bessone I, Rodriguez MS, Alloatti M, Otero MG, Cromberg LE, Pozo Devoto VM, Oubiña G, Sosa L, Buffone MG, Gelman DM, Falzone TL. Kinesin-1-mediated axonal transport of CB1 receptors is required for cannabinoid-dependent axonal growth and guidance. Development 2020; 147:dev184069. [PMID: 32265198 PMCID: PMC7188441 DOI: 10.1242/dev.184069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/23/2020] [Indexed: 12/14/2022]
Abstract
Endocannabinoids (eCB) modulate growth cone dynamics and axonal pathfinding through the stimulation of cannabinoid type-1 receptors (CB1R), the function of which depends on their delivery and precise presentation at the growth cone surface. However, the mechanism involved in the axonal transport of CB1R and its transport role in eCB signaling remains elusive. As mutations in the kinesin-1 molecular motor have been identified in patients with abnormal cortical development and impaired white matter integrity, we studied the defects in axonal pathfinding and fasciculation in mice lacking the kinesin light chain 1 (Klc1-/-) subunit of kinesin-1. Reduced levels of CB1R were found in corticofugal projections and axonal growth cones in Klc1-/- mice. By live-cell imaging of CB1R-eGFP we characterized the axonal transport of CB1R vesicles and described the defects in transport that arise after KLC1 deletion. Cofilin activation, which is necessary for actin dynamics during growth cone remodeling, is impaired in the Klc1-/- cerebral cortex. In addition, Klc1-/- neurons showed expanded growth cones that were unresponsive to CB1R-induced axonal elongation. Together, our data reveal the relevance of kinesin-1 in CB1R axonal transport and in eCB signaling during brain wiring.
Collapse
Affiliation(s)
- Trinidad M M Saez
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - Iván Fernandez Bessone
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - María S Rodriguez
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - Matías Alloatti
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - María G Otero
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - Lucas E Cromberg
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - Victorio M Pozo Devoto
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
| | - Gonzalo Oubiña
- Instituto de Biología y Medicina Experimental, IBYME (CONICET), CP 1428 Buenos Aires, Argentina
| | - Lucas Sosa
- Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, CP 5000 Córdoba, Argentina
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental, IBYME (CONICET), CP 1428 Buenos Aires, Argentina
| | - Diego M Gelman
- Instituto de Biología y Medicina Experimental, IBYME (CONICET), CP 1428 Buenos Aires, Argentina
| | - Tomás L Falzone
- Instituto de Biología Celular y Neurociencia, IBCN (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, CP 1121 Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, IBYME (CONICET), CP 1428 Buenos Aires, Argentina
| |
Collapse
|
20
|
Lucci C, Mesquita-Ribeiro R, Rathbone A, Dajas-Bailador F. Spatiotemporal regulation of GSK3β levels by miRNA-26a controls axon development in cortical neurons. Development 2020; 147:dev.180232. [PMID: 31964775 PMCID: PMC7033742 DOI: 10.1242/dev.180232] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022]
Abstract
Both the establishment of neuronal polarity and axonal growth are crucial steps in the development of the nervous system. The local translation of mRNAs in the axon provides precise regulation of protein expression, and is now known to participate in axon development, pathfinding and synaptic formation and function. We have investigated the role of miR-26a in early stage mouse primary cortical neuron development. We show that micro-RNA-26a-5p (miR-26a) is highly expressed in neuronal cultures, and regulates both neuronal polarity and axon growth. Using compartmentalised microfluidic neuronal cultures, we identified a local role for miR-26a in the axon, where the repression of local synthesis of GSK3β controls axon development and growth. Removal of this repression in the axon triggers local translation of GSK3β protein and subsequent transport to the soma, where it can impact axonal growth. These results demonstrate how the axonal miR-26a can regulate local protein translation in the axon to facilitate retrograde communication to the soma and amplify neuronal responses, in a mechanism that influences axon development. Highlighted Article: Axonal miR-26a can regulate GSK3β translation in the axon to promote retrograde communication to the soma in a mechanism that modulates axon development.
Collapse
Affiliation(s)
- Cristiano Lucci
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH Nottingham, UK
| | - Raquel Mesquita-Ribeiro
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH Nottingham, UK
| | - Alex Rathbone
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH Nottingham, UK
| | - Federico Dajas-Bailador
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH Nottingham, UK
| |
Collapse
|
21
|
Öztürk Z, O’Kane CJ, Pérez-Moreno JJ. Axonal Endoplasmic Reticulum Dynamics and Its Roles in Neurodegeneration. Front Neurosci 2020; 14:48. [PMID: 32116502 PMCID: PMC7025499 DOI: 10.3389/fnins.2020.00048] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
The physical continuity of axons over long cellular distances poses challenges for their maintenance. One organelle that faces this challenge is endoplasmic reticulum (ER); unlike other intracellular organelles, this forms a physically continuous network throughout the cell, with a single membrane and a single lumen. In axons, ER is mainly smooth, forming a tubular network with occasional sheets or cisternae and low amounts of rough ER. It has many potential roles: lipid biosynthesis, glucose homeostasis, a Ca2+ store, protein export, and contacting and regulating other organelles. This tubular network structure is determined by ER-shaping proteins, mutations in some of which are causative for neurodegenerative disorders such as hereditary spastic paraplegia (HSP). While axonal ER shares many features with the tubular ER network in other contexts, these features must be adapted to the long and narrow dimensions of axons. ER appears to be physically continuous throughout axons, over distances that are enormous on a subcellular scale. It is therefore a potential channel for long-distance or regional communication within neurons, independent of action potentials or physical transport of cargos, but involving its physiological roles such as Ca2+ or organelle homeostasis. Despite its apparent stability, axonal ER is highly dynamic, showing features like anterograde and retrograde transport, potentially reflecting continuous fusion and breakage of the network. Here we discuss the transport processes that must contribute to this dynamic behavior of ER. We also discuss the model that these processes underpin a homeostatic process that ensures both enough ER to maintain continuity of the network and repair breaks in it, but not too much ER that might disrupt local cellular physiology. Finally, we discuss how failure of ER organization in axons could lead to axon degenerative diseases, and how a requirement for ER continuity could make distal axons most susceptible to degeneration in conditions that disrupt ER continuity.
Collapse
Affiliation(s)
| | - Cahir J. O’Kane
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
22
|
Liu W, Li X, Chen X, Zhang J, Luo L, Hu Q, Zhou J, Yan J, Lin S, Ye J. JIP1 Deficiency Protects Retinal Ganglion Cells From Apoptosis in a Rotenone-Induced Injury Model. Front Cell Dev Biol 2019; 7:225. [PMID: 31681759 PMCID: PMC6804425 DOI: 10.3389/fcell.2019.00225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/24/2019] [Indexed: 11/13/2022] Open
Abstract
Retinal ganglion cells (RGCs) undergo apoptosis after injury. c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP1) is a scaffold protein that is relevant to JNK activation and a key molecule known to regulate neuronal apoptosis. However, the specific role of JIP1 in the apoptosis of RGCs is currently undefined. Here, we used JIP1 gene knockout (KO) mice to investigate the importance of JIP1-JNK signaling in the apoptosis of RGCs in a rotenone-induced injury model. In adult JIP1 KO mice, the number and electrophysiological functions of RGCs were not different from those of wild-type (WT) mice. Ablation of JIP1 attenuated the activation of JNK and the cleavage of caspase-3 in the retina after rotenone injury and contributed to a lower number of TUNEL-positive RGCs, a greater percentage of surviving RGCs, and a significant reduction in the electrophysiological functional loss of RGCs when compared to those in WT controls. We also found that JIP1 was located in the neurites of primary RGCs, but accumulated in soma in response to rotenone treatment. Moreover, the number of TUNEL-positive RGCs, the level of activation of JNK and the rate of cleavage of caspase-3 were reduced in primary JIP1-deficient RGCs after rotenone injury than in WT controls. Together, our results demonstrate that the JIP1-mediated activation of JNK contributes to the apoptosis of RGCs in a rotenone-induced injury model in vitro and in vivo, suggesting that JIP1 may be a potential therapeutic target for RGC degeneration.
Collapse
Affiliation(s)
- Wenyi Liu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xue Li
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jieqiong Zhang
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Linlin Luo
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Qiumei Hu
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jiaxing Zhou
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jun Yan
- Department 1, Research Institute of Surgery & Daping Hospital, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Atkins M, Gasmi L, Bercier V, Revenu C, Del Bene F, Hazan J, Fassier C. FIGNL1 associates with KIF1Bβ and BICD1 to restrict dynein transport velocity during axon navigation. J Cell Biol 2019; 218:3290-3306. [PMID: 31541015 PMCID: PMC6781435 DOI: 10.1083/jcb.201805128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 05/30/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
Atkins et al. identify a new role for Fidgetin-like 1 in motor axon navigation via its regulation of bidirectional axonal transport. They show that Fidgetin-like 1 binds Kif1bβ and the opposed polarity-directed motor dynein/dynactin in a molecular complex and controls circuit wiring by reducing dynein velocity in developing motor axons. Neuronal connectivity relies on molecular motor-based axonal transport of diverse cargoes. Yet the precise players and regulatory mechanisms orchestrating such trafficking events remain largely unknown. We here report the ATPase Fignl1 as a novel regulator of bidirectional transport during axon navigation. Using a yeast two-hybrid screen and coimmunoprecipitation assays, we showed that Fignl1 binds the kinesin Kif1bβ and the dynein/dynactin adaptor Bicaudal D-1 (Bicd1) in a molecular complex including the dynactin subunit dynactin 1. Fignl1 colocalized with Kif1bβ and showed bidirectional mobility in zebrafish axons. Notably, Kif1bβ and Fignl1 loss of function similarly altered zebrafish motor axon pathfinding and increased dynein-based transport velocity of Rab3 vesicles in these navigating axons, pinpointing Fignl1/Kif1bβ as a dynein speed limiter complex. Accordingly, disrupting dynein/dynactin activity or Bicd1/Fignl1 interaction induced motor axon pathfinding defects characteristic of Fignl1 gain or loss of function, respectively. Finally, pharmacological inhibition of dynein activity partially rescued the axon pathfinding defects of Fignl1-depleted larvae. Together, our results identify Fignl1 as a key dynein regulator required for motor circuit wiring.
Collapse
Affiliation(s)
- Melody Atkins
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Laïla Gasmi
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Valérie Bercier
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Céline Revenu
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Filippo Del Bene
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Jamilé Hazan
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Coralie Fassier
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| |
Collapse
|
24
|
Rab GTPases: Switching to Human Diseases. Cells 2019; 8:cells8080909. [PMID: 31426400 PMCID: PMC6721686 DOI: 10.3390/cells8080909] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
Rab proteins compose the largest family of small GTPases and control the different steps of intracellular membrane traffic. More recently, they have been shown to also regulate cell signaling, division, survival, and migration. The regulation of these processes generally occurs through recruitment of effectors and regulatory proteins, which control the association of Rab proteins to membranes and their activation state. Alterations in Rab proteins and their effectors are associated with multiple human diseases, including neurodegeneration, cancer, and infections. This review provides an overview of how the dysregulation of Rab-mediated functions and membrane trafficking contributes to these disorders. Understanding the altered dynamics of Rabs and intracellular transport defects might thus shed new light on potential therapeutic strategies.
Collapse
|
25
|
Tao T, Sun J, Peng Y, Li Y, Wang P, Chen X, Zhao W, Zheng YY, Wei L, Wang W, Zhou Y, Liu J, Shi YS, Zhu MS. Golgi-resident TRIO regulates membrane trafficking during neurite outgrowth. J Biol Chem 2019; 294:10954-10968. [PMID: 31152060 PMCID: PMC6635450 DOI: 10.1074/jbc.ra118.007318] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/21/2019] [Indexed: 11/06/2022] Open
Abstract
Neurite outgrowth requires coordinated cytoskeletal rearrangements in the growth cone and directional membrane delivery from the neuronal soma. As an essential Rho guanine nucleotide exchange factor (GEF), TRIO is necessary for cytoskeletal dynamics during neurite outgrowth, but its participation in the membrane delivery is unclear. Using co-localization studies, live-cell imaging, and fluorescence recovery after photobleaching analysis, along with neurite outgrowth assay and various biochemical approaches, we here report that in mouse cerebellar granule neurons, TRIO protein pools at the Golgi and regulates membrane trafficking by controlling the directional maintenance of both RAB8 (member RAS oncogene family 8)- and RAB10-positive membrane vesicles. We found that the spectrin repeats in Golgi-resident TRIO confer RAB8 and RAB10 activation by interacting with and activating the RAB GEF RABIN8. Constitutively active RAB8 or RAB10 could partially restore the neurite outgrowth of TRIO-deficient cerebellar granule neurons, suggesting that TRIO-regulated membrane trafficking has an important functional role in neurite outgrowth. Our results also suggest cross-talk between Rho GEF and Rab GEF in controlling both cytoskeletal dynamics and membrane trafficking during neuronal development. They further highlight how protein pools localized to specific organelles regulate crucial cellular activities and functions. In conclusion, our findings indicate that TRIO regulates membrane trafficking during neurite outgrowth in coordination with its GEF-dependent function in controlling cytoskeletal dynamics via Rho GTPases.
Collapse
Affiliation(s)
- Tao Tao
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Jie Sun
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yajing Peng
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Yeqiong Li
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Pei Wang
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Xin Chen
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Wei Zhao
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yan-Yan Zheng
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Lisha Wei
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Wei Wang
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yuwei Zhou
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Jianghuai Liu
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Yun Stone Shi
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and
| | - Min-Sheng Zhu
- Model Animal Research Center, State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology of the Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210061, China and.
| |
Collapse
|
26
|
Etoh K, Fukuda M. Rab10 regulates tubular endosome formation through KIF13A and KIF13B motors. J Cell Sci 2019; 132:jcs.226977. [PMID: 30700496 DOI: 10.1242/jcs.226977] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/17/2019] [Indexed: 01/02/2023] Open
Abstract
Recycling endosomes are stations that sort endocytic cargoes to their appropriate destinations. Tubular endosomes have been characterized as a recycling endosomal compartment for clathrin-independent cargoes. However, the molecular mechanism by which tubular endosome formation is regulated is poorly understood. In this study, we identified Rab10 as a novel protein localized at tubular endosomes by using a comprehensive localization screen of EGFP-tagged Rab small GTPases. Knockout of Rab10 completely abolished tubular endosomal structures in HeLaM cells. We also identified kinesin motors KIF13A and KIF13B as novel Rab10-interacting proteins by means of in silico screening. The results of this study demonstrated that both the Rab10-binding homology domain and the motor domain of KIF13A are required for Rab10-positive tubular endosome formation. Our findings provide insight into the mechanism by which the Rab10-KIF13A (or KIF13B) complex regulates tubular endosome formation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kan Etoh
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
27
|
Tavana JP, Rosene M, Jensen NO, Ridge PG, Kauwe JS, Karch CM. RAB10: an Alzheimer's disease resilience locus and potential drug target. Clin Interv Aging 2018; 14:73-79. [PMID: 30643396 PMCID: PMC6312396 DOI: 10.2147/cia.s159148] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is mainly a late-onset neurodegenerative disorder. Substantial efforts have been made to solve the complex genetic architecture of AD as a means to identify therapeutic targets. Unfortunately, to date, no disease-altering therapeutics have been developed. As therapeutics are likely to be most effective in the early stages of disease (ie, before the onset of symptoms), a recent focus of AD research has been the identification of protective factors that prevent disease. One example is the discovery of a rare variant in the 3'-UTR of RAB10 that is protective for AD. Here, we review the possible genetic, molecular, and functional role of RAB10 in AD and potential therapeutic approaches to target RAB10.
Collapse
Affiliation(s)
- Justina P Tavana
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Matthew Rosene
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,
| | - Nick O Jensen
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,
| | - Perry G Ridge
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - John Sk Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
- Department of Neuroscience, Brigham Young University, Provo, UT 84602, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA,
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, USA,
| |
Collapse
|
28
|
Shima T, Morikawa M, Kaneshiro J, Kambara T, Kamimura S, Yagi T, Iwamoto H, Uemura S, Shigematsu H, Shirouzu M, Ichimura T, Watanabe TM, Nitta R, Okada Y, Hirokawa N. Kinesin-binding-triggered conformation switching of microtubules contributes to polarized transport. J Cell Biol 2018; 217:4164-4183. [PMID: 30297389 PMCID: PMC6279379 DOI: 10.1083/jcb.201711178] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 06/13/2018] [Accepted: 09/14/2018] [Indexed: 01/15/2023] Open
Abstract
Kinesin-1, the founding member of the kinesin superfamily of proteins, is known to use only a subset of microtubules for transport in living cells. This biased use of microtubules is proposed as the guidance cue for polarized transport in neurons, but the underlying mechanisms are still poorly understood. Here, we report that kinesin-1 binding changes the microtubule lattice and promotes further kinesin-1 binding. This high-affinity state requires the binding of kinesin-1 in the nucleotide-free state. Microtubules return to the initial low-affinity state by washing out the binding kinesin-1 or by the binding of non-hydrolyzable ATP analogue AMPPNP to kinesin-1. X-ray fiber diffraction, fluorescence speckle microscopy, and second-harmonic generation microscopy, as well as cryo-EM, collectively demonstrated that the binding of nucleotide-free kinesin-1 to GDP microtubules changes the conformation of the GDP microtubule to a conformation resembling the GTP microtubule.
Collapse
Affiliation(s)
- Tomohiro Shima
- Laboratory for Cell Polarity Regulation, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Manatsu Morikawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Kaneshiro
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Taketoshi Kambara
- Laboratory for Cell Polarity Regulation, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Shinji Kamimura
- Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Toshiki Yagi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Hiroyuki Iwamoto
- Life and Environmental Division, SPring-8, Japan Synchrotron Radiation Research Institute, Hyogo, Japan
| | - Sotaro Uemura
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hideki Shigematsu
- Structural Biology Group, RIKEN Center for Biosystems Dynamics Research, Kanagawa, Japan
| | - Mikako Shirouzu
- Structural Biology Group, RIKEN Center for Biosystems Dynamics Research, Kanagawa, Japan
| | - Taro Ichimura
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Tomonobu M Watanabe
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Ryo Nitta
- Structural Biology Group, RIKEN Center for Biosystems Dynamics Research, Kanagawa, Japan
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Yasushi Okada
- Laboratory for Cell Polarity Regulation, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
- Department of Physics, Universal Biology Institute and the International Research Center for Neurointelligence, The University of Tokyo, Tokyo, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Chua CEL, Tang BL. Rab 10-a traffic controller in multiple cellular pathways and locations. J Cell Physiol 2018; 233:6483-6494. [PMID: 29377137 DOI: 10.1002/jcp.26503] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/24/2018] [Indexed: 12/29/2022]
Abstract
Rab GTPases are key regulators of eukaryotic membrane traffic, and their functions and activities are limited to particular intracellular transport steps and their membrane localization is by and large restricted. Some Rabs do participate in more than one transport steps, but broadly speaking, there is a clear demarcation between exocytic and endocytic Rabs. One Rab protein, Rab10, however, appears to be anomalous in this regard and has a diverse array of functions and subcellular localizations. Rab10 has been implicated in a myriad of activities ranging from polarized exocytosis and endosomal sorting in polarized cells, insulin-dependent Glut4 transport in adipocytes, axonal growth in neurons, and endo-phagocytic processes in macrophages. It's reported subcellular localizations include the endoplasmic reticulum (ER), Golgi/TGN, the endosomes/phagosomes and the primary cilia. In this review, we summarize and discuss the multitude of known roles of Rab10 in cellular membrane transport and the molecular players and mechanisms associated with these roles.
Collapse
Affiliation(s)
- Christelle En Lin Chua
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
30
|
Miro-Working beyond Mitochondria and Microtubules. Cells 2018; 7:cells7030018. [PMID: 29510535 PMCID: PMC5870350 DOI: 10.3390/cells7030018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/02/2018] [Accepted: 03/03/2018] [Indexed: 12/19/2022] Open
Abstract
The small GTPase Miro is best known for its regulation of mitochondrial movement by engaging with the microtubule-based motor proteins kinesin and dynein. Very recent findings have now showed that Miro also targets peroxisomes and regulates microtubule-dependent peroxisome motility. Moreover, Miro recruits and stabilizes the myosin motor Myo19 at the mitochondria to enable actin-based mitochondria movement, which is important for mitochondrial segregation during mitosis. Miro thus has much broader functions that previously known, and these new findings may have important implications on disease pathology.
Collapse
|
31
|
Kreitzer G, Myat MM. Microtubule Motors in Establishment of Epithelial Cell Polarity. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027896. [PMID: 28264820 DOI: 10.1101/cshperspect.a027896] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epithelial cells play a key role in insuring physiological homeostasis by acting as a barrier between the outside environment and internal organs. They are also responsible for the vectorial transport of ions and fluid essential to the function of many organs. To accomplish these tasks, epithelial cells must generate an asymmetrically organized plasma membrane comprised of structurally and functionally distinct apical and basolateral membranes. Adherent and occluding junctions, respectively, anchor cells within a layer and prevent lateral diffusion of proteins in the outer leaflet of the plasma membrane and restrict passage of proteins and solutes through intercellular spaces. At a fundamental level, the establishment and maintenance of epithelial polarity requires that signals initiated at cell-substratum and cell-cell adhesions are transmitted appropriately and dynamically to the cytoskeleton, to the membrane-trafficking machinery, and to the regulation of occluding and adherent junctions. Rigorous descriptive and mechanistic studies published over the last 50 years have provided great detail to our understanding of epithelial polarization. Yet still, critical early steps in morphogenesis are not yet fully appreciated. In this review, we discuss how cytoskeletal motor proteins, primarily kinesins, contribute to coordinated modification of microtubule and actin arrays, formation and remodeling of cell adhesions to targeted membrane trafficking, and to initiating the formation and expansion of an apical lumen.
Collapse
Affiliation(s)
- Geri Kreitzer
- Department of Pathobiology, Sophie Davis School of Biomedical Education, City College of New York, The City University of New York School of Medicine, New York, New York 10031
| | - Monn Monn Myat
- Department of Biology, Medgar Evers College, Brooklyn, New York 11225.,The Graduate Center, The City University of New York, New York, New York 10016
| |
Collapse
|
32
|
Sun T, Li Y, Li T, Ma H, Guo Y, Jiang X, Hou M, Huang S, Chen Z. JIP1 and JIP3 cooperate to mediate TrkB anterograde axonal transport by activating kinesin-1. Cell Mol Life Sci 2017; 74:4027-4044. [PMID: 28638935 PMCID: PMC11107601 DOI: 10.1007/s00018-017-2568-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 11/28/2022]
Abstract
Long-range anterograde axonal transport of TrkB is important for neurons to exert appropriate BDNF responses. TrkB anterograde axonal delivery is mediated by kinesin-1, which associates with TrkB via the adaptor protein JIP3 or the Slp1/Rab27B/CRMP-2 protein complex. However, little is known about the activation mechanisms of TrkB-loaded kinesin-1. Here, we show that JIP1 mediates TrkB anterograde axonal transport using JIP1 knockout mice, sciatic nerve ligation analysis and live imaging. Next, we proved that JIP1 and JIP3 cooperate to mediate TrkB anterograde axonal transport. Finally, microtubule-binding and microfluidic chamber assays revealed that JIP1 and JIP3 cooperate to relieve kinesin-1 autoinhibition, which depends on the binding of JIP1 to kinesin-1 heavy chain (KHC) and light chain (KLC) and the binding of JIP3 to KLC and is essential for TrkB anterograde axonal transport and BDNF-induced TrkB retrograde signal. These findings could deepen our understanding of the regulation mechanism underlying TrkB anterograde axonal transport and provide a novel kinesin-1 autoinhibition-relieving model.
Collapse
Affiliation(s)
- Tao Sun
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, People's Republic of China
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yuan Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Ting Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Huixian Ma
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yunyun Guo
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Xingyu Jiang
- Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
| | - Ming Hou
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Shuhong Huang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Zheyu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Collaborative Innovation Center for Brain Science, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
33
|
Palmisano NJ, Rosario N, Wysocki M, Hong M, Grant B, Meléndez A. The recycling endosome protein RAB-10 promotes autophagic flux and localization of the transmembrane protein ATG-9. Autophagy 2017; 13:1742-1753. [PMID: 28872980 DOI: 10.1080/15548627.2017.1356976] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy/autophagy involves the formation of an autophagosome, a double-membrane vesicle that delivers sequestered cytoplasmic cargo to lysosomes for degradation and recycling. Closely related, endocytosis mediates the sorting and transport of cargo throughout the cell, and both processes are important for cellular homeostasis. However, how endocytic proteins functionally intersect with autophagy is not clear. Mutations in the DAF-2/insulin-like IGF-1 (INSR) receptor at the permissive temperature result in a small increase in GFP::LGG-1 foci, i.e. autophagosomes, but a large increase at the nonpermissive temperature, allowing us to control the level of autophagy. In a RNAi screen for endocytic genes that alter the expression of GFP::LGG-1 in daf-2 mutants, we identified RAB-10, a small GTPase that regulates basolateral endocytosis. Loss of rab-10 in daf-2 mutants results in more GFP::LGG-1-positive foci at the permissive, but less GFP::LGG-1 or SQST-1::GFP foci at the nonpermissive temperature. As previously reported, loss of rab-10 alone resulted in an increase of GFP:LGG-1 foci. Exposure of rab-10 mutant animals to chloroquine, a known inhibitor of autophagic flux, failed to increase the number of GFP::LGG-1 foci. Moreover, colocalization between LMP-1::tagRFP and GFP::LGG-1 (the lysosome and autophagosome reporters) was decreased in daf-2; rab-10 dauers at the nonpermissive temperature. Intriguingly, RAB-10 was required to maintain the normal size of GFP::ATG-9-positive structures in daf-2 mutants at both the permissive and nonpermissive temperature. Finally, we found that RAB-10 GTPase cycling was required to control the size of GFP::ATG-9 foci. Collectively, our data support a model where rab-10 controls autophagic flux by regulating autophagosome formation and maturation.
Collapse
Affiliation(s)
- N J Palmisano
- a Biology Department, Queens College, CUNY , Flushing , NY , USA.,b Biology and Biochemistry Ph.D. Programs , The Graduate Center of the City University of New York , NY , USA
| | - N Rosario
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - M Wysocki
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - M Hong
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - B Grant
- c Department of Molecular Biology and Biochemistry , Rutgers University , Piscataway , NJ , USA
| | - A Meléndez
- a Biology Department, Queens College, CUNY , Flushing , NY , USA.,b Biology and Biochemistry Ph.D. Programs , The Graduate Center of the City University of New York , NY , USA
| |
Collapse
|
34
|
Abstract
Synaptic connections in the brain are continuously weakened or strengthened in response to changes in neuronal activity. This process, known as synaptic plasticity, is the cellular basis for learning and memory, and is thought to be altered in several neuronal disorders. An important aspect of synaptic plasticity is the tightly controlled trafficking and synaptic targeting of the AMPA-type glutamate receptors, which are the major mediators of fast excitatory transmission in the brain. This review addresses the role of Rab GTPases in AMPA receptor trafficking in neurons under basal conditions and during activity-induced synaptic plasticity, especially during long-term potentiation (LTP) and long-term depression (LTD). We highlight the importance of the tight spatio-temporal control of Rab activity and suggest that this is critical for proper neuronal functions. We also discuss how abnormal AMPA receptor trafficking and malfunctioning of Rabs can lead to neurologic disorders or memory problems.
Collapse
Affiliation(s)
- Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart , Stuttgart , Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart , Stuttgart , Germany
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University , Budapest , Hungary.,MTA-ELTE NAP B Neuronal Cell Biology Research Group, Eötvös Loránd University , Budapest , Hungary
| |
Collapse
|
35
|
Abstract
Neurons are highly polarized cells that exhibit one of the more complex morphology and function. Neuronal intracellular trafficking plays a key role in dictating the directionality and specificity of vesicle formation, transport and fusion, allowing the transmission of information in sophisticate cellular network. Thus, the integrity of protein trafficking and spatial organization is especially important in neuronal cells. RAB proteins, small monomeric GTPases belonging to the RAS superfamily, spatially and temporally orchestrate specific vesicular trafficking steps. In this review we summarise the known roles of RAB GTPases involved in the maintenance of neuronal vesicular trafficking in the central nervous system. In particular, we discriminate the axonal pre-synaptic trafficking and dendritic post-synaptic trafficking, to better underlie how a correct orchestration of vesicle movement is necessary to maintain neuronal polarity and then, to permit an accurate architecture and functionality of synaptic activity.
Collapse
Affiliation(s)
- Maria Lidia Mignogna
- a Molecular Genetics of Intellectual Disabilities Unit, Division of Neuroscience at IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Patrizia D'Adamo
- a Molecular Genetics of Intellectual Disabilities Unit, Division of Neuroscience at IRCCS San Raffaele Scientific Institute , Milan , Italy
| |
Collapse
|
36
|
Shao G, Wang Y, Guan S, Burlingame AL, Lu F, Knox R, Ferriero DM, Jiang X. Proteomic Analysis of Mouse Cortex Postsynaptic Density following Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2017; 39:66-81. [PMID: 28315865 PMCID: PMC5519436 DOI: 10.1159/000456030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/11/2017] [Indexed: 02/02/2023] Open
Abstract
Proteomics of the synapses and postsynaptic densities (PSDs) have provided a deep understanding of protein composition and signal networks in the adult brain, which underlie neuronal plasticity and neurodegenerative or psychiatric disorders. However, there is a paucity of knowledge about the architecture and organization of PSDs in the immature brain, and how it is modified by brain injury in an early developing stage. Mass spectrometry (MS)-based proteomic analysis was performed on PSDs prepared from cortices of postnatal day 9 naïve mice or pups which had suffered hypoxic-ischemic (HI) brain injury. 512 proteins of different functional groups were identified from PSDs collected 1 h after HI injury, among which 60 have not been reported previously. Seven newly identified proteins involved in neural development were highlighted. HI injury increased the yield of PSDs at early time points upon reperfusion, and multiple proteins were recruited into PSDs following the insult. Quantitative analysis was performed using spectral counting, and proteins whose relative expression was more than 50% up- or downregulated compared to the sham animals 1 h after HI insult were reported. Validation with Western blotting demonstrated changes in expression and phosphorylation of the N-methyl-D-aspartate receptor, activation of a series of postsynaptic protein kinases and dysregulation of scaffold and adaptor proteins in response to neonatal HI insult. This work, along with other recent studies of synaptic protein profiling in the immature brain, builds a foundation for future investigation on the molecular mechanisms underlying developing plasticity. Furthermore, it provides insights into the biochemical changes of PSDs following early brain hypoxia-ischemia, which is helpful for understanding not only the injury mechanisms, but also the process of repair or replenishment of neuronal circuits during recovery from brain damage.
Collapse
Affiliation(s)
- Guo Shao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Cell polarity refers to the asymmetric localization of cellular components that allows cells to carry out their specialized functions, be they epithelial barrier function, transmission of action potentials in nerve cells, or modulation of the immune response. The establishment and maintenance of cell polarity requires the directed trafficking of membrane proteins and lipids - essential processes that are mediated by Rab GTPases. Interestingly, several of the Rabs that impact polarity are present in the earliest eukaryotes, and the Rab polarity repertoire has expanded as cells have become more complex. There is a substantial conservation of Rab function across diverse cell types. Rabs act through an assortment of effector proteins that include scaffolding proteins, cytoskeletal motors, and other small GTPases. In this review we highlight the similarities and differences in Rab function for the instruction of polarity in diverse cell types.
Collapse
Affiliation(s)
- Sara S Parker
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| | - Christopher Cox
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| | - Jean M Wilson
- a Department of Cellular and Molecular Medicine , University of Arizona , Tucson , AZ , USA
| |
Collapse
|
38
|
Villarroel-Campos D, Bronfman FC, Gonzalez-Billault C. Rab GTPase signaling in neurite outgrowth and axon specification. Cytoskeleton (Hoboken) 2016; 73:498-507. [PMID: 27124121 DOI: 10.1002/cm.21303] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/30/2022]
Abstract
Neurons are highly polarized cells that contain specialized subcellular domains involved in information transmission in the nervous system. Specifically, the somatodendritic compartment receives neuronal inputs while the axons convey information through the synapse. The establishment of asymmetric domains requires a specific delivery of components, including organelles, proteins, and membrane. The Rab family of small GTPases plays an essential role in membrane trafficking. Signaling cascades triggered by extrinsic and intrinsic factors tightly regulate Rab functions in cells, with Rab protein activation depending on GDP/GTP binding to establish a binary mode of action. This review summarizes the contributions of several Rab family members involved in trans-Golgi, early/late endosomes, and recycling endosomes during neurite development and axonal outgrowth. The regulation of some Rabs by guanine exchanging factors and GTPase activating proteins will also be addressed. Finally, discussion will be provided on how specific effector-mediated Rab activation modifies several molecules essential to neuronal differentiation. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David Villarroel-Campos
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad De Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Francisca C Bronfman
- MINREB And Center for Ageing and Regeneration (CARE), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad De Chile, Santiago, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| |
Collapse
|
39
|
Homma Y, Fukuda M. Rabin8 regulates neurite outgrowth in both GEF activity-dependent and -independent manners. Mol Biol Cell 2016; 27:2107-18. [PMID: 27170183 PMCID: PMC4927283 DOI: 10.1091/mbc.e16-02-0091] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/03/2016] [Indexed: 11/13/2022] Open
Abstract
Several Rab GTPases have been implicated in neurite outgrowth, but their regulatory mechanisms are poorly understood. Rab10 is a novel substrate of a Rab8-GEF, Rabin8, and Rabin8 regulates neurite outgrowth of PC12 cells by coordinating with Rab8, Rab10, and Rab11 and by a GEF activity–independent mechanism. Many aspects of membrane-trafficking events are regulated by Rab-family small GTPases. Neurite outgrowth requires massive addition of proteins and lipids to the tips of growing neurites by membrane trafficking, and although several Rabs, including Rab8, Rab10, and Rab11, have been implicated in this process, their regulatory mechanisms during neurite outgrowth are poorly understood. Here, we show that Rabin8, a Rab8-guanine nucleotide exchange factor (GEF), regulates nerve growth factor (NGF)–induced neurite outgrowth of PC12 cells. Knockdown of Rabin8 results in inhibition of neurite outgrowth, whereas overexpression promotes it. We also find that Rab10 is a novel substrate of Rabin8 and that both Rab8 and Rab10 function during neurite outgrowth downstream of Rabin8. Surprisingly, however, a GEF activity–deficient isoform of Rabin8 also promotes neurite outgrowth, indicating the existence of a GEF activity–independent role of Rabin8. The Arf6/Rab8-positive recycling endosomes (Arf6/Rab8-REs) and Rab10/Rab11-positive REs (Rab10/Rab11-REs) in NGF-stimulated PC12 cells are differently distributed. Rabin8 localizes on both RE populations and appears to activate Rab8 and Rab10 there. These localizations and functions of Rabin8 are Rab11 dependent. Thus Rabin8 regulates neurite outgrowth both by coordinating with Rab8, Rab10, and Rab11 and by a GEF activity–independent mechanism.
Collapse
Affiliation(s)
- Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
40
|
Miller KG. Keeping Neuronal Cargoes on the Right Track: New Insights into Regulators of Axonal Transport. Neuroscientist 2016; 23:232-250. [PMID: 27154488 DOI: 10.1177/1073858416648307] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In neurons, a single motor (dynein) transports large organelles as well as synaptic and dense core vesicles toward microtubule minus ends; however, it is unclear why dynein appears more active on organelles, which are generally excluded from mature axons, than on synaptic and dense core vesicles, which are maintained at high levels. Recent studies in Zebrafish and Caenorhabditis elegans have shown that JIP3 promotes dynein-mediated retrograde transport to clear some organelles (lysosomes, early endosomes, and Golgi) from axons and prevent their potentially harmful accumulation in presynaptic regions. A JIP3 mutant suppressor screen in C. elegans revealed that JIP3 promotes the clearance of organelles from axons by blocking the action of the CSS system (Cdk5, SAD Kinase, SYD-2/Liprin). A synthesis of results in vertebrates with the new findings suggests that JIP3 blocks the CSS system from disrupting the connection between dynein and organelles. Most components of the CSS system are enriched at presynaptic active zones where they normally contribute to maintaining optimal levels of captured synaptic and dense core vesicles, in part by inhibiting dynein transport. The JIP3-CSS system model explains how neurons selectively regulate a single minus-end motor to exclude specific classes of organelles from axons, while at the same time ensuring optimal levels of synaptic and dense core vesicles.
Collapse
Affiliation(s)
- Kenneth G Miller
- 1 Genetic Models of Disease Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| |
Collapse
|
41
|
Membrane Trafficking in Neuronal Development: Ins and Outs of Neural Connectivity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:247-80. [PMID: 26940520 DOI: 10.1016/bs.ircmb.2015.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During development, neurons progress through rapid yet stereotypical shape changes to achieve proper neuronal connectivity. This morphological progression requires carefully orchestrated plasma membrane expansion, insertion of membrane components including receptors for extracellular cues into the plasma membrane and removal and trafficking of membrane materials and proteins to specific locations. This review outlines the cellular machinery of membrane trafficking that play an integral role in neuronal cell shape change and function from initial neurite formation to pathway navigation and synaptogenesis.
Collapse
|
42
|
RAB-10 Regulates Dendritic Branching by Balancing Dendritic Transport. PLoS Genet 2015; 11:e1005695. [PMID: 26633194 PMCID: PMC4669152 DOI: 10.1371/journal.pgen.1005695] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 10/31/2015] [Indexed: 11/19/2022] Open
Abstract
The construction of a large dendritic arbor requires robust growth and the precise delivery of membrane and protein cargoes to specific subcellular regions of the developing dendrite. How the microtubule-based vesicular trafficking and sorting systems are regulated to distribute these dendritic development factors throughout the dendrite is not well understood. Here we identify the small GTPase RAB-10 and the exocyst complex as critical regulators of dendrite morphogenesis and patterning in the C. elegans sensory neuron PVD. In rab-10 mutants, PVD dendritic branches are reduced in the posterior region of the cell but are excessive in the distal anterior region of the cell. We also demonstrate that the dendritic branch distribution within PVD depends on the balance between the molecular motors kinesin-1/UNC-116 and dynein, and we propose that RAB-10 regulates dendrite morphology by balancing the activity of these motors to appropriately distribute branching factors, including the transmembrane receptor DMA-1. Building a complex dendritic arbor requires tremendous cellular growth, and how membrane and protein components are transported to support a rapidly growing, polarized dendrite remains unclear. We have identified the small GTPase RAB-10 as a key regulator of this process, providing insight into both dendritic development and the control of trafficking by small GTPases.
Collapse
|
43
|
Basolateral Endocytic Recycling Requires RAB-10 and AMPH-1 Mediated Recruitment of RAB-5 GAP TBC-2 to Endosomes. PLoS Genet 2015; 11:e1005514. [PMID: 26393361 PMCID: PMC4578947 DOI: 10.1371/journal.pgen.1005514] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 08/18/2015] [Indexed: 11/27/2022] Open
Abstract
The small GTPase RAB-5/Rab5 is a master regulator of the early endosome, required for a myriad of coordinated activities, including the degradation and recycling of internalized cargo. Here we focused on the recycling function of the early endosome and the regulation of RAB-5 by GAP protein TBC-2 in the basolateral C. elegans intestine. We demonstrate that downstream basolateral recycling regulators, GTPase RAB-10/Rab10 and BAR domain protein AMPH-1/Amphiphysin, bind to TBC-2 and help to recruit it to endosomes. In the absence of RAB-10 or AMPH-1 binding to TBC-2, RAB-5 membrane association is abnormally high and recycling cargo is trapped in early endosomes. Furthermore, the loss of TBC-2 or AMPH-1 leads to abnormally high spatial overlap of RAB-5 and RAB-10. Taken together our results indicate that RAB-10 and AMPH-1 mediated down-regulation of RAB-5 is an important step in recycling, required for cargo exit from early endosomes and regulation of early endosome–recycling endosome interactions. When cargo is internalized from the cell surface by endocytosis, it enters a series of intracellular organelles called endosomes. Endosomes sort cargo, such that some cargos are sent to the lysosome for degradation, while others are recycled to the plasma membrane. Small GTPase proteins of the Rabs family are master regulators of endosomes, functioning by acting as molecular switches. As cargo moves through the endosomal system, it must pass from the domain controlled by one Rab-GTPase to the domain controlled by another. Little is known about how transitions along the recycling pathway are controlled. Here we analyze a group of protein interactions that act along the early-to-recycling pathway. Our work shows that RAB-5 deactivation mediated by TBC-2 and its recruiters RAB-10 and AMPH-1 is important for cargo recycling. This work provides mechanistic insight into how Rab proteins controlling different steps of trafficking interact during endocytic recycling.
Collapse
|
44
|
Zou W, Yadav S, DeVault L, Jan YN, Sherwood DR. RAB-10-Dependent Membrane Transport Is Required for Dendrite Arborization. PLoS Genet 2015; 11:e1005484. [PMID: 26394140 PMCID: PMC4578882 DOI: 10.1371/journal.pgen.1005484] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/05/2015] [Indexed: 01/07/2023] Open
Abstract
Formation of elaborately branched dendrites is necessary for the proper input and connectivity of many sensory neurons. Previous studies have revealed that dendritic growth relies heavily on ER-to-Golgi transport, Golgi outposts and endocytic recycling. How new membrane and associated cargo is delivered from the secretory and endosomal compartments to sites of active dendritic growth, however, remains unknown. Using a candidate-based genetic screen in C. elegans, we have identified the small GTPase RAB-10 as a key regulator of membrane trafficking during dendrite morphogenesis. Loss of rab-10 severely reduced proximal dendritic arborization in the multi-dendritic PVD neuron. RAB-10 acts cell-autonomously in the PVD neuron and localizes to the Golgi and early endosomes. Loss of function mutations of the exocyst complex components exoc-8 and sec-8, which regulate tethering, docking and fusion of transport vesicles at the plasma membrane, also caused proximal dendritic arborization defects and led to the accumulation of intracellular RAB-10 vesicles. In rab-10 and exoc-8 mutants, the trans-membrane proteins DMA-1 and HPO-30, which promote PVD dendrite stabilization and branching, no longer localized strongly to the proximal dendritic membranes and instead were sequestered within intracellular vesicles. Together these results suggest a crucial role for the Rab10 GTPase and the exocyst complex in controlling membrane transport from the secretory and/or endosomal compartments that is required for dendritic growth. Dendrites are cellular extensions from neurons that gather information from other neurons or cues from the external environment to convey to the nervous system of an organism. Dendrites are often extensively branched, raising the question of how neurons supply plasma membrane and dendrite specific proteins from the source of synthesis inside the cell to developing dendrites. We have examined membrane trafficking in the PVD neuron in the nematode worm C. elegans to investigate how new membrane and dendrite proteins are trafficked. The PVD neuron is easy to visualize and has remarkably long and widely branched dendrites positioned along the skin of the worm, which transmits information about harsh touch and cold temperature to the nervous system. We have discovered that a key organizer of vesicle trafficking, the RAB-10 protein, localizes to membrane vesicles and is required to traffic these vesicles that contain plasma membrane and dendrite proteins to the growing PVD dendrite. Further, our work revealed that a complex of proteins, termed the exocyst, that helps fuse membrane vesicles at the plasma membrane, localizes with RAB-10 and is required for dendrite branching. Together, our work has revealed a novel mechanism for how neurons build dendrites that could be used to help repair damaged neurons in human diseases and during aging.
Collapse
Affiliation(s)
- Wei Zou
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Smita Yadav
- Howard Hughes Medical Institute, Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Laura DeVault
- Howard Hughes Medical Institute, Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, Department of Physiology, University of California, San Francisco, San Francisco, California, United States of America
| | - David R. Sherwood
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Ishida M, Ohbayashi N, Fukuda M. Rab1A regulates anterograde melanosome transport by recruiting kinesin-1 to melanosomes through interaction with SKIP. Sci Rep 2015; 5:8238. [PMID: 25649263 PMCID: PMC4316160 DOI: 10.1038/srep08238] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/13/2015] [Indexed: 01/01/2023] Open
Abstract
Melanosomes are lysosome-related organelles in melanocytes that are transported from the perinucleus to the cell periphery by coordination between bidirectional (anterograde and retrograde) microtubule-dependent transport and unidirectional actin-dependent transport. Although the molecular machineries that mediate retrograde transport and actin-dependent transport have already been identified, little is known about the anterograde transport complex on microtubules in mammalian cells. Here we discovered that small GTPase Rab1A on melanosomes recruits SKIP/PLEKHM2 as a Rab1A-specific effector and that Rab1A, SKIP, and a kinesin-1/(Kif5b+KLC2) motor form a transport complex that mediates anterograde melanosome transport in melanocytes. Interestingly, Arl8, Arf-like small GTPase that also interacts with SKIP, is specifically localized at lysosomes and regulates their anterograde transport in melanocytes. Our findings suggest that the anterograde microtubule-dependent transport of melanosomes and lysosomes are differently regulated by independent cargo receptors, i.e., Rab1A and Arl8, respectively, but that a SKIP–kinesin-1 mechanism is responsible for the transport of both.
Collapse
Affiliation(s)
- Morié Ishida
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Norihiko Ohbayashi
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
46
|
JSAP1/JIP3 and JLP regulate kinesin-1-dependent axonal transport to prevent neuronal degeneration. Cell Death Differ 2015; 22:1260-74. [PMID: 25571974 DOI: 10.1038/cdd.2014.207] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 11/08/2022] Open
Abstract
Axonal transport is critical for neuronal development and function, and defective axonal transport has been implicated in neurodegenerative diseases. However, how axonal transport is regulated, or how defective transport leads to neuronal degeneration, remains unclear. Here, we report that c-Jun NH2-terminal kinase (JNK)/stress-activated protein kinase-associated protein 1 (JSAP1, also known as JNK-interacting protein 3 (JIP3)) and JNK-associated leucine zipper protein (JLP) are essential for postnatal brain development. Mice with a double-knockout (dKO) in Jsap1 and Jlp in the dorsal telencephalon developed progressive neuron loss. Using a primary neuron culture system with induced disruption of targeted genes, combined with gene rescue experiments, we show that JSAP1 and JLP regulate kinesin-1-dependent axonal transport with functional redundancy. We also show that the binding of JSAP1 and JLP to kinesin-1 heavy chain is crucial for interactions between kinesin-1 and microtubules. Furthermore, we describe a molecular mechanism by which defective kinesin-1-dependent axonal transport in Jsap1:Jlp dKO neurons causes axonal degeneration and subsequent neuronal death. JNK hyperactivation because of increased intra-axonal Ca(2+) in the Jsap1:Jlp dKO neurons was found to mediate both the axonal degeneration and neuronal death, in cooperation with the Ca(2+)-dependent protease calpain. Our results indicate that axonal JNK may relocate to the nucleus in a dynein-dependent manner, where it activates the transcription factor c-Jun, resulting in neuronal death. Taken together, our data establish JSAP1 and JLP as positive regulators of kinesin-1-dependent axonal transport, which prevents neuronal degeneration.
Collapse
|
47
|
The role of rab proteins in neuronal cells and in the trafficking of neurotrophin receptors. MEMBRANES 2014; 4:642-77. [PMID: 25295627 PMCID: PMC4289860 DOI: 10.3390/membranes4040642] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/27/2014] [Accepted: 09/16/2014] [Indexed: 12/11/2022]
Abstract
Neurotrophins are a family of proteins that are important for neuronal development, neuronal survival and neuronal functions. Neurotrophins exert their role by binding to their receptors, the Trk family of receptor tyrosine kinases (TrkA, TrkB, and TrkC) and p75NTR, a member of the tumor necrosis factor (TNF) receptor superfamily. Binding of neurotrophins to receptors triggers a complex series of signal transduction events, which are able to induce neuronal differentiation but are also responsible for neuronal maintenance and neuronal functions. Rab proteins are small GTPases localized to the cytosolic surface of specific intracellular compartments and are involved in controlling vesicular transport. Rab proteins, acting as master regulators of the membrane trafficking network, play a central role in both trafficking and signaling pathways of neurotrophin receptors. Axonal transport represents the Achilles' heel of neurons, due to the long-range distance that molecules, organelles and, in particular, neurotrophin-receptor complexes have to cover. Indeed, alterations of axonal transport and, specifically, of axonal trafficking of neurotrophin receptors are responsible for several human neurodegenerative diseases, such as Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis and some forms of Charcot-Marie-Tooth disease. In this review, we will discuss the link between Rab proteins and neurotrophin receptor trafficking and their influence on downstream signaling pathways.
Collapse
|
48
|
Lalli G. Regulation of neuronal polarity. Exp Cell Res 2014; 328:267-75. [PMID: 25107381 DOI: 10.1016/j.yexcr.2014.07.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/24/2014] [Accepted: 07/26/2014] [Indexed: 12/28/2022]
Abstract
The distinctive polarized morphology of neuronal cells is essential for the proper wiring of the nervous system. The rodent hippocampal neuron culture established about three decades ago has provided an amenable in vitro system to uncover the molecular mechanisms underlying neuronal polarization, a process relying on highly regulated cytoskeletal dynamics, membrane traffic and localized protein degradation. More recent research in vivo has highlighted the importance of the extracellular environment and cell-cell interactions in neuronal polarity. Here, I will review some key signaling pathways regulating neuronal polarization and provide some insights on the complexity of this process gained from in vivo studies.
Collapse
Affiliation(s)
- Giovanna Lalli
- Wolfson Centre for Age-Related Diseases, King׳s College London, Guy׳s Campus, London SE1 1UL, UK.
| |
Collapse
|
49
|
MARCKS regulates membrane targeting of Rab10 vesicles to promote axon development. Cell Res 2014; 24:576-94. [PMID: 24662485 DOI: 10.1038/cr.2014.33] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 12/31/2013] [Accepted: 02/25/2014] [Indexed: 01/19/2023] Open
Abstract
Axon development requires membrane addition from the intracellular supply, which has been shown to be mediated by Rab10-positive plasmalemmal precursor vesicles (PPVs). However, the molecular mechanisms underlying the membrane trafficking processes of PPVs remain unclear. Here, we show that myristoylated alanine-rich C-kinase substrate (MARCKS) mediates membrane targeting of Rab10-positive PPVs, and this regulation is critical for axon development. We found that the GTP-locked active form of Rab10 binds to membrane-associated MARCKS, whose affinity depends on the phosphorylation status of the MARCKS effector domain. Either genetic silencing of MARCKS or disruption of its interaction with Rab10 inhibited axon growth of cortical neurons, impaired docking and fusion of Rab10 vesicles with the plasma membrane, and consequently caused a loss of membrane insertion of axonal receptors responsive to extracellular axon growth factors. Thus, this study has identified a novel function of MARCKS in mediating membrane targeting of PPVs during axon development.
Collapse
|