1
|
Yildiz A. Mechanism and regulation of kinesin motors. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00780-6. [PMID: 39394463 DOI: 10.1038/s41580-024-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
Kinesins are a diverse superfamily of microtubule-based motors that perform fundamental roles in intracellular transport, cytoskeletal dynamics and cell division. These motors share a characteristic motor domain that powers unidirectional motility and force generation along microtubules, and they possess unique tail domains that recruit accessory proteins and facilitate oligomerization, regulation and cargo recognition. The location, direction and timing of kinesin-driven processes are tightly regulated by various cofactors, adaptors, microtubule tracks and microtubule-associated proteins. This Review focuses on recent structural and functional studies that reveal how members of the kinesin superfamily use the energy of ATP hydrolysis to transport cargoes, depolymerize microtubules and regulate microtubule dynamics. I also survey how accessory proteins and post-translational modifications regulate the autoinhibition, cargo binding and motility of some of the best-studied kinesins. Despite much progress, the mechanism and regulation of kinesins are still emerging, and unresolved questions can now be tackled using newly developed approaches in biophysics and structural biology.
Collapse
Affiliation(s)
- Ahmet Yildiz
- Physics Department, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
2
|
Vogt K, Kulkarni A, Pandey R, Dehnad M, Konopka G, Greene R. Sleep need driven oscillation of glutamate synaptic phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578985. [PMID: 38370691 PMCID: PMC10871195 DOI: 10.1101/2024.02.05.578985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Sleep loss increases AMPA-synaptic strength and number in the neocortex. However, this is only part of the synaptic sleep loss response. We report increased AMPA/NMDA EPSC ratio in frontal-cortical pyramidal neurons of layers 2-3. Silent synapses are absent, decreasing the plastic potential to convert silent NMDA to active AMPA synapses. These sleep loss changes are recovered by sleep. Sleep genes are enriched for synaptic shaping cellular components controlling glutamate synapse phenotype, overlap with autism risk genes and are primarily observed in excitatory pyramidal neurons projecting intra-telencephalically. These genes are enriched with genes controlled by the transcription factor, MEF2c and its repressor, HDAC4. Sleep genes can thus provide a framework within which motor learning and training occurs mediated by sleep-dependent oscillation of glutamate-synaptic phenotypes.
Collapse
Affiliation(s)
- K.E. Vogt
- International Institute of Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - A. Kulkarni
- Department of Neuroscience, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - R. Pandey
- Department of Psychiatry, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - M. Dehnad
- Department of Psychiatry, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - G. Konopka
- Department of Neuroscience, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - R.W. Greene
- International Institute of Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Neuroscience, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
- Department of Psychiatry, Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
3
|
Gong R, Qin L, Chen L, Wang N, Bao Y, Lu W. Myosin Va-dependent Transport of NMDA Receptors in Hippocampal Neurons. Neurosci Bull 2024; 40:1053-1075. [PMID: 38291290 PMCID: PMC11306496 DOI: 10.1007/s12264-023-01174-y] [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: 07/18/2023] [Accepted: 11/03/2023] [Indexed: 02/01/2024] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) trafficking is a key process in the regulation of synaptic efficacy and brain function. However, the molecular mechanism underlying the surface transport of NMDARs is largely unknown. Here we identified myosin Va (MyoVa) as the specific motor protein that traffics NMDARs in hippocampal neurons. We found that MyoVa associates with NMDARs through its cargo binding domain. This association was increased during NMDAR surface transport. Knockdown of MyoVa suppressed NMDAR transport. We further demonstrated that Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates NMDAR transport through its direct interaction with MyoVa. Furthermore, MyoVa employed Rab11 family-interacting protein 3 (Rab11/FIP3) as the adaptor proteins to couple themselves with NMDARs during their transport. Accordingly, the knockdown of FIP3 impairs hippocampal memory. Together, we conclude that in hippocampal neurons, MyoVa conducts active transport of NMDARs in a CaMKII-dependent manner.
Collapse
Affiliation(s)
- Ru Gong
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linwei Qin
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linlin Chen
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Ning Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Yifei Bao
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
4
|
Bouchenafa R, Johnson de Sousa Brito FM, Piróg KA. Involvement of kinesins in skeletal dysplasia: a review. Am J Physiol Cell Physiol 2024; 327:C278-C290. [PMID: 38646780 PMCID: PMC11293425 DOI: 10.1152/ajpcell.00613.2023] [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: 11/12/2023] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024]
Abstract
Skeletal dysplasias are group of rare genetic diseases resulting from mutations in genes encoding structural proteins of the cartilage extracellular matrix (ECM), signaling molecules, transcription factors, epigenetic modifiers, and several intracellular proteins. Cell division, organelle maintenance, and intracellular transport are all orchestrated by the cytoskeleton-associated proteins, and intracellular processes affected through microtubule-associated movement are important for the function of skeletal cells. Among microtubule-associated motor proteins, kinesins in particular have been shown to play a key role in cell cycle dynamics, including chromosome segregation, mitotic spindle formation, and ciliogenesis, in addition to cargo trafficking, receptor recycling, and endocytosis. Recent studies highlight the fundamental role of kinesins in embryonic development and morphogenesis and have shown that mutations in kinesin genes lead to several skeletal dysplasias. However, many questions concerning the specific functions of kinesins and their adaptor molecules as well as specific molecular mechanisms in which the kinesin proteins are involved during skeletal development remain unanswered. Here we present a review of the skeletal dysplasias resulting from defects in kinesins and discuss the involvement of kinesin proteins in the molecular mechanisms that are active during skeletal development.
Collapse
Affiliation(s)
- Roufaida Bouchenafa
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Katarzyna Anna Piróg
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
5
|
Pan Y, Wang J, Gao X, Du C, Hou C, Tang D, Zhu J. Expression Dynamics Indicate Potential Roles of KIF17 for Nuclear Reshaping and Tail Formation during Spermiogenesis in Phascolosoma esculenta. Int J Mol Sci 2023; 25:128. [PMID: 38203305 PMCID: PMC10779256 DOI: 10.3390/ijms25010128] [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: 10/25/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Kinesin family member17 (KIF17), a homologous dimer of the kinesin-2 protein family, has important microtubule-dependent and -independent roles in spermiogenesis. Little is known about KIF17 in the mollusk, Phascolosoma esculenta, a newly developed mariculture species in China. Here, we cloned the open reading frame of Pe-kif17 and its related gene, Pe-act, and performed bioinformatics analysis on both. Pe-KIF17 and Pe-ACT are structurally conserved, indicating that they may be functionally conserved. The expression pattern of kif17/act mRNA performed during spermiogenesis revealed their expression in diverse tissues, with the highest expression level in the coelomic fluid of P. esculenta. The expressions of Pe-kif17 and Pe-act mRNA were relatively high during the breeding season (July-September), suggesting that Pe-KIF17/ACT may be involved in spermatogenesis, particularly during spermiogenesis. Further analysis of Pe-kif17 mRNA via fluorescence in situ hybridization revealed the continuous expression of this mRNA during spermiogenesis, suggesting potential functions in this process. Immunofluorescence showed that Pe-KIF17 co-localized with α-tubulin and migrated from the perinuclear cytoplasm to one side of the spermatid, forming the sperm tail. Pe-KIF17 and Pe-ACT also colocalized. KIF17 may participate in spermiogenesis of P. esculenta, particularly in nuclear reshaping and tail formation by interacting with microtubule structures similar to the manchette. Moreover, Pe-KIF17 with Pe-ACT is also involved in nuclear reshaping and tail formation in the absence of microtubules. This study provides evidence for the role of KIF17 during spermiogenesis and provides theoretical data for studies of the reproductive biology of P. esculenta. These findings are important for spermatogenesis in mollusks.
Collapse
Affiliation(s)
- Yue Pan
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jingqian Wang
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xinming Gao
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chen Du
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Congcong Hou
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Daojun Tang
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Junquan Zhu
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; (Y.P.); (J.W.); (X.G.); (C.D.); (C.H.)
- Key Laboratory of Marine Biotechnology of Zhejiang Province, College of Marine Sciences, Ningbo University, Ningbo 315211, China
| |
Collapse
|
6
|
Terada N, Saitoh Y, Saito M, Yamada T, Kamijo A, Yoshizawa T, Sakamoto T. Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families. Genes (Basel) 2023; 14:1942. [PMID: 37895291 PMCID: PMC10606877 DOI: 10.3390/genes14101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The protein 4.1 and membrane palmitoylated protein (MPP) families were originally found as components in the erythrocyte membrane skeletal protein complex, which helps maintain the stability of erythrocyte membranes by linking intramembranous proteins and meshwork structures composed of actin and spectrin under the membranes. Recently, it has been recognized that cells and tissues ubiquitously use this membrane skeletal system. Various intramembranous proteins, including adhesion molecules, ion channels, and receptors, have been shown to interact with the 4.1 and MPP families, regulating cellular and tissue dynamics by binding to intracellular signal transduction proteins. In this review, we focus on our previous studies regarding genetically modified animal models, especially on 4.1G, MPP6, and MPP2, to describe their functional roles in the peripheral nervous system, the central nervous system, the testis, and bone formation. As the membrane skeletal proteins are located at sites that receive signals from outside the cell and transduce signals inside the cell, it is necessary to elucidate their molecular interrelationships, which may broaden the understanding of cell and tissue functions.
Collapse
Affiliation(s)
- Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Yurika Saitoh
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan
| | - Masaki Saito
- School of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan;
| | - Tomoki Yamada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Akio Kamijo
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Division of Basic & Clinical Medicine, Nagano College of Nursing, Komagane City, Nagano 399-4117, Japan
| | - Takahiro Yoshizawa
- Division of Animal Research, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto City, Nagano 390-8621, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| |
Collapse
|
7
|
Pascual-Alonso A, Xiol C, Smirnov D, Kopajtich R, Prokisch H, Armstrong J. Identification of molecular signatures and pathways involved in Rett syndrome using a multi-omics approach. Hum Genomics 2023; 17:85. [PMID: 37710353 PMCID: PMC10503149 DOI: 10.1186/s40246-023-00532-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Rett syndrome (RTT) is a neurodevelopmental disorder mainly caused by mutations in the methyl-CpG-binding protein 2 gene (MECP2). MeCP2 is a multi-functional protein involved in many cellular processes, but the mechanisms by which its dysfunction causes disease are not fully understood. The duplication of the MECP2 gene causes a distinct disorder called MECP2 duplication syndrome (MDS), highlighting the importance of tightly regulating its dosage for proper cellular function. Additionally, some patients with mutations in genes other than MECP2 exhibit phenotypic similarities with RTT, indicating that these genes may also play a role in similar cellular functions. The purpose of this study was to characterise the molecular alterations in patients with RTT in order to identify potential biomarkers or therapeutic targets for this disorder. METHODS We used a combination of transcriptomics (RNAseq) and proteomics (TMT mass spectrometry) to characterise the expression patterns in fibroblast cell lines from 22 patients with RTT and detected mutation in MECP2, 15 patients with MDS, 12 patients with RTT-like phenotypes and 13 healthy controls. Transcriptomics and proteomics data were used to identify differentially expressed genes at both RNA and protein levels, which were further inspected via enrichment and upstream regulator analyses and compared to find shared features in patients with RTT. RESULTS We identified molecular alterations in cellular functions and pathways that may contribute to the disease phenotype in patients with RTT, such as deregulated cytoskeletal components, vesicular transport elements, ribosomal subunits and mRNA processing machinery. We also compared RTT expression profiles with those of MDS seeking changes in opposite directions that could lead to the identification of MeCP2 direct targets. Some of the deregulated transcripts and proteins were consistently affected in patients with RTT-like phenotypes, revealing potentially relevant molecular processes in patients with overlapping traits and different genetic aetiology. CONCLUSIONS The integration of data in a multi-omics analysis has helped to interpret the molecular consequences of MECP2 dysfunction, contributing to the characterisation of the molecular landscape in patients with RTT. The comparison with MDS provides knowledge of MeCP2 direct targets, whilst the correlation with RTT-like phenotypes highlights processes potentially contributing to the pathomechanism leading these disorders.
Collapse
Affiliation(s)
- Ainhoa Pascual-Alonso
- Fundació Per La Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Clara Xiol
- Fundació Per La Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Dmitrii Smirnov
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Robert Kopajtich
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Judith Armstrong
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain.
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- Genomic Unit, Molecular and Genetic Medicine Section, Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
8
|
Ujiié Y, Ishitani Y, Nagai Y, Takaki Y, Toyofuku T, Ishii S. Unique evolution of foraminiferal calcification to survive global changes. SCIENCE ADVANCES 2023; 9:eadd3584. [PMID: 37343099 DOI: 10.1126/sciadv.add3584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 05/15/2023] [Indexed: 06/23/2023]
Abstract
Foraminifera, the most ancient known calcium carbonate-producing eukaryotes, are crucial players in global biogeochemical cycles and well-used environmental indicators in biogeosciences. However, little is known about their calcification mechanisms. This impedes understanding the organismal responses to ocean acidification, which alters marine calcium carbonate production, potentially leading to biogeochemical cycle changes. We conducted comparative single-cell transcriptomics and fluorescent microscopy and identified calcium ion (Ca2+) transport/secretion genes and α-carbonic anhydrases that control calcification in a foraminifer. They actively take up Ca2+ to boost mitochondrial adenosine triphosphate synthesis during calcification but need to pump excess intracellular Ca2+ to the calcification site to prevent cell death. Unique α-carbonic anhydrase genes induce the generation of bicarbonate and proton from multiple CO2 sources. These control mechanisms have evolved independently since the Precambrian to enable the development of large cells and calcification despite decreasing Ca2+ concentrations and pH in seawater. The present findings provide previously unknown insights into the calcification mechanisms and their subsequent function in enduring ocean acidification.
Collapse
Affiliation(s)
- Yurika Ujiié
- Marine Core Research Institute, Kochi University, Kōchi, Japan
| | - Yoshiyuki Ishitani
- Institute for Extra-cutting-edge Science and Technology Avant-garde Research (X-star), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
| | - Yukiko Nagai
- Institute for Extra-cutting-edge Science and Technology Avant-garde Research (X-star), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
- National Museum of Nature and Science, Tokyo, Japan
| | - Yoshihiro Takaki
- Institute for Extra-cutting-edge Science and Technology Avant-garde Research (X-star), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
| | - Takashi Toyofuku
- Institute for Extra-cutting-edge Science and Technology Avant-garde Research (X-star), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
- Tokyo University of Marine Science and Technology (TUMSAT), Tokyo, Japan
| | - Shun'ichi Ishii
- Institute for Extra-cutting-edge Science and Technology Avant-garde Research (X-star), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Japan
| |
Collapse
|
9
|
van der Veer BK, Chen L, Custers C, Athanasouli P, Schroiff M, Cornelis R, Chui JSH, Finnell R, Lluis F, Koh K. Dual functions of TET1 in germ layer lineage bifurcation distinguished by genomic context and dependence on 5-methylcytosine oxidation. Nucleic Acids Res 2023; 51:5469-5498. [PMID: 37021585 PMCID: PMC10287924 DOI: 10.1093/nar/gkad231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/12/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Gastrulation begins when the epiblast forms the primitive streak or becomes definitive ectoderm. During this lineage bifurcation, the DNA dioxygenase TET1 has bipartite functions in transcriptional activation and repression, but the mechanisms remain unclear. By converting mouse embryonic stem cells (ESCs) into neuroprogenitors, we defined how Tet1-/- cells switch from neuroectoderm fate to form mesoderm and endoderm. We identified the Wnt repressor Tcf7l1 as a TET1 target that suppresses Wnt/β-catenin and Nodal signalling. ESCs expressing catalytic dead TET1 retain neural potential but activate Nodal and subsequently Wnt/β-catenin pathways to generate also mesoderm and endoderm. At CpG-poor distal enhancers, TET1 maintains accessible chromatin at neuroectodermal loci independently of DNA demethylation. At CpG-rich promoters, DNA demethylation by TET1 affects the expression of bivalent genes. In ESCs, a non-catalytic TET1 cooperation with Polycomb represses primitive streak genes; post-lineage priming, the interaction becomes antagonistic at neuronal genes, when TET1's catalytic activity is further involved by repressing Wnt signalling. The convergence of repressive DNA and histone methylation does not inhibit neural induction in Tet1-deficient cells, but some DNA hypermethylated loci persist at genes with brain-specific functions. Our results reveal versatile switching of non-catalytic and catalytic TET1 activities based on genomic context, lineage and developmental stage.
Collapse
Affiliation(s)
- Bernard K van der Veer
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
| | - Lehua Chen
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
| | - Colin Custers
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
| | - Paraskevi Athanasouli
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell Signaling, B-3000 Leuven, Belgium
| | - Mariana Schroiff
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
| | - Riet Cornelis
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
| | - Jonathan Sai-Hong Chui
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell Signaling, B-3000 Leuven, Belgium
| | - Richard H Finnell
- Baylor College of Medicine, Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Houston, TX 77030, USA
- Baylor College of Medicine, Department of Molecular and Human Genetics, Department of Medicine, Houston, TX 77030, USA
| | - Frederic Lluis
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell Signaling, B-3000 Leuven, Belgium
| | - Kian Peng Koh
- KU Leuven, Department of Development and Regeneration, Laboratory of Stem Cell and Developmental Epigenetics, B-3000 Leuven, Belgium
- Baylor College of Medicine, Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Houston, TX 77030, USA
| |
Collapse
|
10
|
Bahouth SW, Nooh MM, Mancarella S. Involvement of SAP97 anchored multiprotein complexes in regulating cardiorenal signaling and trafficking networks. Biochem Pharmacol 2023; 208:115406. [PMID: 36596415 DOI: 10.1016/j.bcp.2022.115406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
SAP97 is a member of the MAGUK family of proteins, but unlike other MAGUK proteins that are selectively expressed in the CNS, SAP97 is also expressed in peripheral organs, like the heart and kidneys. SAP97 has several protein binding cassettes, and this review will describe their involvement in creating SAP97-anchored multiprotein networks. SAP97-anchored networks localized at the inner leaflet of the cell membrane play a major role in trafficking and targeting of membrane G protein-coupled receptors (GPCR), channels, and structural proteins. SAP97 plays a major role in compartmentalizing voltage gated sodium and potassium channels to specific cellular compartments of heart cells. SAP97 undergoes extensive alternative splicing. These splice variants give rise to different SAP97 isoforms that alter its cellular localization, networking, signaling and trafficking effects. Regarding GPCR, SAP97 binds to the β1-adrenergic receptor and recruits AKAP5/PKA and PDE4D8 to create a multiprotein complex that regulates trafficking and signaling of cardiac β1-AR. In the kidneys, SAP97 anchored networks played a role in trafficking of aquaporin-2 water channels. Cardiac specific ablation of SAP97 (SAP97-cKO) resulted in cardiac hypertrophy and failure in aging mice. Similarly, instituting transverse aortic constriction (TAC) in young SAP97 c-KO mice exacerbated TAC-induced cardiac remodeling and dysfunction. These findings highlight a critical role for SAP97 in the pathophysiology of a number of cardiac and renal diseases, suggesting that SAP97 is a relevant target for drug discovery.
Collapse
Affiliation(s)
- Suleiman W Bahouth
- Department of Pharmacology, Addiction Science and Toxicology, The University of Tennessee-Health Sciences Center, Memphis, TN, United States.
| | - Mohammed M Nooh
- Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt and Biochemistry Department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Salvatore Mancarella
- Department of Physiology, The University of Tennessee-Health Sciences Center, Memphis, TN, United States
| |
Collapse
|
11
|
Hosseini S, van Ham M, Erck C, Korte M, Michaelsen-Preusse K. The role of α-tubulin tyrosination in controlling the structure and function of hippocampal neurons. Front Mol Neurosci 2022; 15:931859. [PMCID: PMC9627282 DOI: 10.3389/fnmol.2022.931859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubules (MTs) are central components of the neuronal cytoskeleton and play a critical role in CNS integrity, function, and plasticity. Neuronal MTs are diverse due to extensive post-translational modifications (PTMs), particularly detyrosination/tyrosination, in which the C-terminal tyrosine of α-tubulin is cyclically removed by a carboxypeptidase and reattached by a tubulin-tyrosine ligase (TTL). The detyrosination/tyrosination cycle of MTs has been shown to be an important regulator of MT dynamics in neurons. TTL-null mice exhibit impaired neuronal organization and die immediately after birth, indicating TTL function is vital to the CNS. However, the detailed cellular role of TTL during development and in the adult brain remains elusive. Here, we demonstrate that conditional deletion of TTL in the neocortex and hippocampus during network development results in a pathophysiological phenotype defined by incomplete development of the corpus callosum and anterior commissures due to axonal growth arrest. TTL loss was also associated with a deficit in spatial learning, impaired synaptic plasticity, and reduced number of spines in hippocampal neurons, suggesting that TTL also plays a critical role in hippocampal network development. TTL deletion after postnatal development, specifically in the hippocampus and in cultured hippocampal neurons, led to a loss of spines and impaired spine structural plasticity. This indicates a novel and important function of TTL for synaptic plasticity in the adult brain. In conclusion, this study reveals the importance of α-tubulin tyrosination, which defines the dynamics of MTs, in controlling proper network formation and suggests TTL-mediated tyrosination as a new key determinant of synaptic plasticity in the adult brain.
Collapse
Affiliation(s)
- Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marco van Ham
- Research Group Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christian Erck
- Research Group Cellular Proteome Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- *Correspondence: Kristin Michaelsen-Preusse,
| |
Collapse
|
12
|
Glavač D, Mladinić M, Ban J, Mazzone GL, Sámano C, Tomljanović I, Jezernik G, Ravnik-Glavač M. The Potential Connection between Molecular Changes and Biomarkers Related to ALS and the Development and Regeneration of CNS. Int J Mol Sci 2022; 23:ijms231911360. [PMID: 36232667 PMCID: PMC9570269 DOI: 10.3390/ijms231911360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases are one of the greatest medical burdens of the modern age, being mostly incurable and with limited prognostic and diagnostic tools. Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurodegenerative disease characterized by the loss of motoneurons, with a complex etiology, combining genetic, epigenetic, and environmental causes. The neuroprotective therapeutic approaches are very limited, while the diagnostics rely on clinical examination and the exclusion of other diseases. The recent advancement in the discovery of molecular pathways and gene mutations involved in ALS has deepened the understanding of the disease pathology and opened the possibility for new treatments and diagnostic procedures. Recently, 15 risk loci with distinct genetic architectures and neuron-specific biology were identified as linked to ALS through common and rare variant association analyses. Interestingly, the quantity of related proteins to these genes has been found to change during early postnatal development in mammalian spinal cord tissue (opossum Monodelphis domestica) at the particular time when neuroregeneration stops being possible. Here, we discuss the possibility that the ALS-related genes/proteins could be connected to neuroregeneration and development. Moreover, since the regulation of gene expression in developmental checkpoints is frequently regulated by non-coding RNAs, we propose that studying the changes in the composition and quantity of non-coding RNA molecules, both in ALS patients and in the developing central nervous (CNS) system of the opossum at the time when neuroregeneration ceases, could reveal potential biomarkers useful in ALS prognosis and diagnosis.
Collapse
Affiliation(s)
- Damjan Glavač
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljublana, Slovenia
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Miranda Mladinić
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Jelena Ban
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Graciela L. Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Buenos Aires B1629AHJ, Argentina
| | - Cynthia Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana Unidad Cuajimalpa, Mexico City 05348, Mexico
| | - Ivana Tomljanović
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Gregor Jezernik
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Metka Ravnik-Glavač
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
13
|
Yamada T, Saitoh Y, Kametani K, Kamijo A, Sakamoto T, Terada N. Involvement of membrane palmitoylated protein 2 (MPP2) in the synaptic molecular complex at the mouse cerebellar glomerulus. Histochem Cell Biol 2022; 158:497-511. [PMID: 35854144 DOI: 10.1007/s00418-022-02137-6] [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] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
We previously reported that the membrane skeletal protein 4.1G in the peripheral nervous system transports membrane palmitoylated protein 6 (MPP6), which interacts with the synaptic scaffolding protein Lin7 and cell adhesion molecule 4 (CADM4) in Schwann cells that form myelin. In the present study, we investigated the localization of and proteins related to MPP2, a highly homologous family protein of MPP6, in the cerebellum of the mouse central nervous system, in which neurons are well organized. Immunostaining for MPP2 was observed at cerebellar glomeruli (CG) in the granular layer after postnatal day 14. Using the high-resolution Airyscan mode of a confocal laser-scanning microscope, MPP2 was detected as a dot pattern and colocalized with CADM1 and Lin7, recognized as small ring/line patterns, as well as with calcium/calmodulin-dependent serine protein kinase (CASK), NMDA glutamate receptor 1 (GluN1), and M-cadherin, recognized as dot patterns, indicating the localization of MPP2 in the excitatory postsynaptic region and adherens junctions of granule cells. An immunoprecipitation analysis revealed that MPP2 formed a molecular complex with CADM1, CASK, M-cadherin, and Lin7. Furthermore, the Lin7 staining pattern showed small rings surrounding mossy fibers in wild-type CG, while it changed to the dot/spot pattern inside small rings detected with CADM1 staining in MPP2-deficient CG. These results indicate that MPP2 influences the distribution of Lin7 to synaptic cell membranes at postsynaptic regions in granule cells at CG, at which electric signals enter the cerebellum.
Collapse
Affiliation(s)
- Tomoki Yamada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Yurika Saitoh
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo, Japan
| | - Kiyokazu Kametani
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Akio Kamijo
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Division of Basic and Clinical Medicine, Nagano College of Nursing, Komagane, Nagano, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
| |
Collapse
|
14
|
Liu Y, Tian X, Ke P, Gu J, Ma Y, Guo Y, Xu X, Chen Y, Yang M, Wang X, Xiao F. KIF17 Modulates Epileptic Seizures and Membrane Expression of the NMDA Receptor Subunit NR2B. Neurosci Bull 2022; 38:841-856. [PMID: 35678994 PMCID: PMC9352834 DOI: 10.1007/s12264-022-00888-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 04/01/2022] [Indexed: 10/18/2022] Open
Abstract
Epilepsy is a common and severe brain disease affecting >65 million people worldwide. Recent studies have shown that kinesin superfamily motor protein 17 (KIF17) is expressed in neurons and is involved in regulating the dendrite-targeted transport of N-methyl-D-aspartate receptor subtype 2B (NR2B). However, the effect of KIF17 on epileptic seizures remains to be explored. We found that KIF17 was mainly expressed in neurons and that its expression was increased in epileptic brain tissue. In the kainic acid (KA)-induced epilepsy mouse model, KIF17 overexpression increased the severity of epileptic activity, whereas KIF17 knockdown had the opposite effect. In electrophysiological tests, KIF17 regulated excitatory synaptic transmission, potentially due to KIF17-mediated NR2B membrane expression. In addition, this report provides the first demonstration that KIF17 is modified by SUMOylation (SUMO, small ubiquitin-like modifier), which plays a vital role in the stabilization and maintenance of KIF17 in epilepsy.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Pingyang Ke
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Juan Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yi Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Xin Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Yuanyuan Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Min Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| |
Collapse
|
15
|
Kumari D, Ray K. Phosphoregulation of Kinesins Involved in Long-Range Intracellular Transport. Front Cell Dev Biol 2022; 10:873164. [PMID: 35721476 PMCID: PMC9203973 DOI: 10.3389/fcell.2022.873164] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/29/2022] [Indexed: 12/28/2022] Open
Abstract
Kinesins, the microtubule-dependent mechanochemical enzymes, power a variety of intracellular movements. Regulation of Kinesin activity and Kinesin-Cargo interactions determine the direction, timing and flux of various intracellular transports. This review examines how phosphorylation of Kinesin subunits and adaptors influence the traffic driven by Kinesin-1, -2, and -3 family motors. Each family of Kinesins are phosphorylated by a partially overlapping set of serine/threonine kinases, and each event produces a unique outcome. For example, phosphorylation of the motor domain inhibits motility, and that of the stalk and tail domains induces cargo loading and unloading effects according to the residue and context. Also, the association of accessory subunits with cargo and adaptor proteins with the motor, respectively, is disrupted by phosphorylation. In some instances, phosphorylation by the same kinase on different Kinesins elicited opposite outcomes. We discuss how this diverse range of effects could manage the logistics of Kinesin-dependent, long-range intracellular transport.
Collapse
|
16
|
Wang J, Liu Z, Gao X, Du C, Hou C, Tang D, Lou B, Shen W, Zhu J. The potential function of KIF17 in large yellow croaker (Larimichthys crocea) spermatid remodeling: molecular characterization and expression pattern during spermiogenesis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:603-616. [PMID: 35538183 DOI: 10.1007/s10695-021-01035-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/18/2021] [Indexed: 06/14/2023]
Abstract
KIF17, which belongs to the kinesin-2 protein family, plays an indispensable role in mammalian spermiogenesis. However, the role of KIF17 in fish spermatid remodeling during spermiogenesis remains poorly understood. Therefore, we aimed to study the role of KIF17 in spermatid remodeling during Larimichthys crocea (L. crocea) spermiogenesis. The kif17 cDNA sequence, 3247 bp in length, was cloned from L. crocea testis, which consisted of a 347-bp 5'-untranslated region (UTR), 413-bp 3' -UTR, and 2487-bp open reading frame. Bioinformatic analyses revealed that KIF17 obtained from L. crocea (Lc-KIF17) exhibited a high sequence identity compared with those from other teleosts and possessed the structural features of other kinesin-2 proteins. Based on structural similarity, we speculate that the role of Lc-KIF17 may be similar to that of KIF17 in other animals. Lc-kif17 mRNA was diffusely expressed in L. crocea tissues and was highly expressed in the testis, especially at stage IV testicular development. Immunofluorescence analysis revealed that Lc-KIF17 signals colocalized with β-tubulin signals and migrated from the perinuclear cytoplasm to the side of the nucleus where the tail forms during spermiogenesis. These findings revealed that KIF17 may be involved in L. crocea spermiogenesis. In particular, KIF17 may participate in spermatid remodeling by interacting with perinuclear microtubules during L. crocea spermiogenesis. Collectively, this study contributes to an improved understanding of the mechanism underlying L. crocea spermiogenesis and provides a basis for further research on L. crocea reproduction and development.
Collapse
Affiliation(s)
- Jingqian Wang
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Zhao Liu
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Xinming Gao
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Chen Du
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Congcong Hou
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Daojun Tang
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China
| | - Bao Lou
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province, 310021, People's Republic of China
| | - Weiliang Shen
- Ningbo Academy of Oceanology and Fishery, Ningbo, 315012, Zhejiang, China
| | - Junquan Zhu
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province, 315211, People's Republic of China.
| |
Collapse
|
17
|
Peña-Ortega F, Robles-Gómez ÁA, Xolalpa-Cueva L. Microtubules as Regulators of Neural Network Shape and Function: Focus on Excitability, Plasticity and Memory. Cells 2022; 11:cells11060923. [PMID: 35326374 PMCID: PMC8946818 DOI: 10.3390/cells11060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
Neuronal microtubules (MTs) are complex cytoskeletal protein arrays that undergo activity-dependent changes in their structure and function as a response to physiological demands throughout the lifespan of neurons. Many factors shape the allostatic dynamics of MTs and tubulin dimers in the cytosolic microenvironment, such as protein–protein interactions and activity-dependent shifts in these interactions that are responsible for their plastic capabilities. Recently, several findings have reinforced the role of MTs in behavioral and cognitive processes in normal and pathological conditions. In this review, we summarize the bidirectional relationships between MTs dynamics, neuronal processes, and brain and behavioral states. The outcomes of manipulating the dynamicity of MTs by genetic or pharmacological approaches on neuronal morphology, intrinsic and synaptic excitability, the state of the network, and behaviors are heterogeneous. We discuss the critical position of MTs as responders and adaptative elements of basic neuronal function whose impact on brain function is not fully understood, and we highlight the dilemma of artificially modulating MT dynamics for therapeutic purposes.
Collapse
|
18
|
Zheng R, Du Y, Wang X, Liao T, Zhang Z, Wang N, Li X, Shen Y, Shi L, Luo J, Xia J, Wang Z, Xu J. KIF2C regulates synaptic plasticity and cognition in mice through dynamic microtubule depolymerization. eLife 2022; 11:72483. [PMID: 35138249 PMCID: PMC8828051 DOI: 10.7554/elife.72483] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Dynamic microtubules play a critical role in cell structure and function. In nervous system, microtubules are the major route for cargo protein trafficking and they specially extend into and out of synapses to regulate synaptic development and plasticity. However, the detailed depolymerization mechanism that regulates dynamic microtubules in synapses and dendrites is still unclear. In this study, we find that KIF2C, a dynamic microtubule depolymerization protein without known function in the nervous system, plays a pivotal role in the structural and functional plasticity of synapses and regulates cognitive function in mice. Through its microtubule depolymerization capability, KIF2C regulates microtubule dynamics in dendrites, and regulates microtubule invasion of spines in neurons in a neuronal activity-dependent manner. Using RNAi knockdown and conditional knockout approaches, we showed that KIF2C regulates spine morphology and synaptic membrane expression of AMPA receptors. Moreover, KIF2C deficiency leads to impaired excitatory transmission, long-term potentiation, and altered cognitive behaviors in mice. Collectively, our study explores a novel function of KIF2C in the nervous system and provides an important regulatory mechanism on how activity-dependent microtubule dynamic regulates synaptic plasticity and cognition behaviors.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yonglan Du
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xintai Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Tailin Liao
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Zhe Zhang
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Na Wang
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xiumao Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Shen
- Department of Physiology and Department of Neurology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guanzhou, China
| | - Jianhong Luo
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jun Xia
- Division of Life Science and The Brain and Intelligence Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ziyi Wang
- Innovative Institute of Basic Medical Sciences of Zhejiang University (Yuhang), Hangzhou, China
| | - Junyu Xu
- Department of Neurobiology and Department of Rehabilitation of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Sanyal C, Pietsch N, Ramirez Rios S, Peris L, Carrier L, Moutin MJ. The detyrosination/re-tyrosination cycle of tubulin and its role and dysfunction in neurons and cardiomyocytes. Semin Cell Dev Biol 2021; 137:46-62. [PMID: 34924330 DOI: 10.1016/j.semcdb.2021.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
Among the variety of post-translational modifications to which microtubules are subjected, the detyrosination/re-tyrosination cycle is specific to tubulin. It is conserved by evolution and characterized by the enzymatic removal and re-addition of a gene-encoded tyrosine residue at the C-terminus of α-tubulin. Detyrosinated tubulin can be further converted to Δ2-tubulin by the removal of an additional C-terminal glutamate residue. Detyrosinated and Δ2-tubulin are carried by stable microtubules whereas tyrosinated microtubules are present on dynamic polymers. The cycle regulates trafficking of many cargo transporting molecular motors and is linked to the microtubule dynamics via regulation of microtubule interactions with specific cellular effectors such as kinesin-13. Here, we give an historical overview of the general features discovered for the cycle. We highlight the recent progress toward structure and functioning of the enzymes that keep the levels of tyrosinated and detyrosinated tubulin in cells, the long-known tubulin tyrosine ligase and the recently discovered vasohibin-SVBP complexes. We further describe how the cycle controls microtubule functions in healthy neurons and cardiomyocytes and how deregulations of the cycle are involved in dysfunctions of these highly differentiated cells, leading to neurodegeneration and heart failure in humans.
Collapse
Affiliation(s)
- Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sacnicte Ramirez Rios
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
20
|
Fan R, Lai KO. Understanding how kinesin motor proteins regulate postsynaptic function in neuron. FEBS J 2021; 289:2128-2144. [PMID: 34796656 DOI: 10.1111/febs.16285] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023]
Abstract
The Kinesin superfamily proteins (KIFs) are major molecular motors that transport diverse set of cargoes along microtubules to both the axon and dendrite of a neuron. Much of our knowledge about kinesin function is obtained from studies on axonal transport. Emerging evidence reveals how specific kinesin motor proteins carry cargoes to dendrites, including proteins, mRNAs and organelles that are crucial for synapse development and plasticity. In this review, we will summarize the major kinesin motors and their associated cargoes that have been characterized to regulate postsynaptic function in neuron. We will also discuss how specific kinesins are selectively involved in the development of excitatory and inhibitory postsynaptic compartments, their regulation by post-translational modifications (PTM), as well as their roles beyond conventional transport carrier.
Collapse
Affiliation(s)
- Ruolin Fan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Uniyal A, Thakur V, Rani M, Tiwari V, Akhilesh, Gadepalli A, Ummadisetty O, Modi A, Tiwari V. Kinesin Nanomotors Mediated Trafficking of NMDA-Loaded Cargo as A Novel Target in Chronic Pain. ACS Chem Neurosci 2021; 12:2956-2963. [PMID: 34324307 DOI: 10.1021/acschemneuro.1c00319] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic pain is among the most prevalent burdensome disorders worldwide. The N-methyl-d-aspartate (NMDA) receptor system plays a critical role in central sensitization, a primary feature of chronic pain. Despite the proven efficacy of exogenous ligands to this receptor system in preclinical studies, evidence for the clinical efficacy of NMDA antagonists for the treatment of chronic pain is weak. Researchers are studying alternate approaches, rather than direct inhibition of the NMDA receptors in pain processing neurons. This indirect approach utilizes the modulation of molecular switches that regulates the synthesis, maturation, and transport of receptors from cellular organelles to the synaptic membrane. Kinesins are nanomotors that anterogradely transport the cargo using microtubule tracks across the neurons. Various members of the kinesin family, including KIF17, KIF11, KIF5b, and KIF21a, regulate the intracellular transport of NMDA receptors. Pharmacological targeting of these ATP-driven nanomotors could be a useful tool for manipulating the NMDAR functioning. It could provide the potential for the development of a novel strategy for the management of chronic pain.
Collapse
Affiliation(s)
- Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Vaibhav Thakur
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Mousmi Rani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Vineeta Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Ajay Modi
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology B.H.U., Varanasi 221005, India
| |
Collapse
|
22
|
Brachet A, Lario A, Fernández-Rodrigo A, Heisler FF, Gutiérrez Y, Lobo C, Kneussel M, Esteban JA. A kinesin 1-protrudin complex mediates AMPA receptor synaptic removal during long-term depression. Cell Rep 2021; 36:109499. [PMID: 34348158 DOI: 10.1016/j.celrep.2021.109499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 05/19/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022] Open
Abstract
The synaptic removal of AMPA-type glutamate receptors (AMPARs) is a core mechanism for hippocampal long-term depression (LTD). In this study, we address the role of microtubule-dependent transport of AMPARs as a driver for vesicular trafficking and sorting during LTD. Here, we show that the kinesin-1 motor KIF5A/C is strictly required for LTD expression in CA3-to-CA1 hippocampal synapses. Specifically, we find that KIF5 is required for an efficient internalization of AMPARs after NMDA receptor activation. We show that the KIF5/AMPAR complex is assembled in an activity-dependent manner and associates with microsomal membranes upon LTD induction. This interaction is facilitated by the vesicular adaptor protrudin, which is also required for LTD expression. We propose that protrudin links KIF5-dependent transport to endosomal sorting, preventing AMPAR recycling to synapses after LTD induction. Therefore, this work identifies an activity-dependent molecular motor and the vesicular adaptor protein that executes AMPAR synaptic removal during LTD.
Collapse
Affiliation(s)
- Anna Brachet
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | - Argentina Lario
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Alba Fernández-Rodrigo
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Frank F Heisler
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), D-20251 Hamburg, Germany
| | - Yolanda Gutiérrez
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Clara Lobo
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Matthias Kneussel
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), D-20251 Hamburg, Germany
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| |
Collapse
|
23
|
Hara T, Saeki K, Jinnouchi H, Kazuno S, Miura Y, Yokomizo T. The c-terminal region of BLT2 restricts its localization to the lateral membrane in a LIN7C-dependent manner. FASEB J 2021; 35:e21364. [PMID: 33481310 DOI: 10.1096/fj.202002640r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 11/11/2022]
Abstract
Leukotriene B4 receptor type 2 (BLT2) is a G protein-coupled receptor (GPCR) mainly expressed in epithelial cells, where it enhances barrier function. A unique characteristic of BLT2 is its restricted localization to the lateral membrane. However, the molecular mechanism underlying the localization of BLT2 to the lateral membrane and the physiological roles of laterally localized BLT2 are unknown. BLT1 is the most homologous GPCR to BLT2 and localizes to both the apical and lateral membranes. In this study, we generated chimeric receptors of BLT2 and BLT1 as well as deletion mutants of BLT2 to determine the region(s) of BLT2 responsible for its localization. Chimeric receptors containing the C-terminal domain of BLT2 localized only to the lateral membrane, and the C-terminal deletion mutant of BLT2 accumulated at the Golgi apparatus. Furthermore, the middle and C-terminal regions of BLT2 were important for maintaining epithelial barrier function. Proteomics analysis using the chimeric BLT-ascorbate peroxidase 2 biotinylation method showed that some proteins involved in intracellular protein transport, cell-cell junctions, and actin filament binding were located very close to the C-terminal domain of BLT2. Knockdown of lin-7 homolog C (LIN7C), a membrane trafficking protein, led to accumulation of BLT2 in the Golgi apparatus, resulting in diminished epithelial barrier function. These results suggest that the C-terminal region of BLT2 plays an important role in the transport of BLT2 from the Golgi apparatus to the plasma membrane in a LIN7C-dependent manner.
Collapse
Affiliation(s)
- Takuya Hara
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan.,Fuji Research Laboratories, Kowa Co., Ltd, Shizuoka, Japan
| | - Kazuko Saeki
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiromi Jinnouchi
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Regulation of the NMDA receptor by its cytoplasmic domains: (How) is the tail wagging the dog? Neuropharmacology 2021; 195:108634. [PMID: 34097949 DOI: 10.1016/j.neuropharm.2021.108634] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022]
Abstract
Excitatory neurotransmission mediated by N-methyl-d-aspartate receptors (NMDARs) is critical for synapse development, function, and plasticity in the brain. NMDARs are tetra-heteromeric cation-channels that mediate synaptic transmission and plasticity. Extensive human studies show the existence of genetic variants in NMDAR subunits genes (GRIN genes) that are associated with neurodevelopmental and neuropsychiatric disorders, including autism spectrum disorders (ASD), epilepsy (EP), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), and schizophrenia (SCZ). NMDAR subunits have a unique modular architecture with four semiautonomous domains. Here we focus on the carboxyl terminal domain (CTD), also known as the intracellular C-tail, which varies in length among the glutamate receptor subunits and is the most diverse domain in terms of amino acid sequence. The CTD shows no sequence homology to any known proteins but encodes short docking motifs for intracellular binding proteins and covalent modifications. Our review will discuss the many important functions of the CTD in regulating NMDA membrane and synaptic targeting, stabilization, degradation targeting, allosteric modulation and metabotropic signaling of the receptor. This article is part of the special issue on 'Glutamate Receptors - NMDA Receptors'.
Collapse
|
25
|
Goldstein O, Gana‐Weisz M, Shiner T, Attar R, Mordechai Y, Waldman YY, Bar‐Shira A, Thaler A, Gurevich T, Mirelman A, Giladi N, Orr‐Urtreger A. R869C mutation in molecular motor KIF17 gene is involved in dementia with Lewy bodies. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12143. [PMID: 34124335 PMCID: PMC8176903 DOI: 10.1002/dad2.12143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION THE GBA-N370S mutation is one of the most frequent risk factors for dementia with Lewy bodies (DLB) and Parkinson's disease (PD). We looked for genetic variations that contribute to the outcome in N370S-carriers, whether PD or DLB. METHODS Whole-genome sequencing of 95 Ashkenazi-N370S-carriers affected with either DLB (n = 19) or PD (n = 76) was performed, and 564 genes related to dementia and PD analyzed. RESULTS We identified enrichment of linked alleles in PINK1 locus in DLB patients (false discovery rate P = .0412). Haplotype analysis delineated 1.8 Mb interval encompassing 29 genes and 87 unique variants, of them, KIF17-R869C received the highest functional prediction score (Combined Annotation Dependent Depletion = 34). Its frequency was significantly higher in 26 DLB-N370S-carriers compared to 140 PD-N370S-carriers (odds ratio [OR] = 33.4 P = .001, and OR = 70.2 when only heterozygotes were included). DISCUSSION Because KIF17 was shown to be important for learning and memory in mice, our data further suggest, for the first time, its involvement in DLB, and possibly in human dementia.
Collapse
Affiliation(s)
- Orly Goldstein
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Mali Gana‐Weisz
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Tamara Shiner
- Cognitive Neurology Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Reut Attar
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Yael Mordechai
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | | | - Anat Bar‐Shira
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Avner Thaler
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory for Early Markers of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Tanya Gurevich
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Anat Mirelman
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory for Early Markers of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Nir Giladi
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory for Early Markers of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Avi Orr‐Urtreger
- The Genomic Research Laboratory for Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of Medicine, and Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
26
|
Gutiérrez Y, López-García S, Lario A, Gutiérrez-Eisman S, Delevoye C, Esteban JA. KIF13A drives AMPA receptor synaptic delivery for long-term potentiation via endosomal remodeling. J Cell Biol 2021; 220:212112. [PMID: 33999113 PMCID: PMC8129809 DOI: 10.1083/jcb.202003183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 02/16/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
The regulated trafficking of AMPA-type glutamate receptors (AMPARs) from dendritic compartments to the synaptic membrane in response to neuronal activity is a core mechanism for long-term potentiation (LTP). However, the contribution of the microtubule cytoskeleton to this synaptic transport is still unknown. In this work, using electrophysiological, biochemical, and imaging techniques, we have found that one member of the kinesin-3 family of motor proteins, KIF13A, is specifically required for the delivery of AMPARs to the spine surface during LTP induction. Accordingly, KIF13A depletion from hippocampal slices abolishes LTP expression. We also identify the vesicular protein centaurin-α1 as part of a motor transport machinery that is engaged with KIF13A and AMPARs upon LTP induction. Finally, we determine that KIF13A is responsible for the remodeling of Rab11-FIP2 endosomal structures in the dendritic shaft during LTP. Overall, these results identify specific kinesin molecular motors and endosomal transport machinery that catalyzes the dendrite-to-synapse translocation of AMPA receptors during synaptic plasticity.
Collapse
Affiliation(s)
- Yolanda Gutiérrez
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Sergio López-García
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Argentina Lario
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| | - Cédric Delevoye
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Cell and Tissue Imaging Facility, Paris, France
| | - José A Esteban
- Molecular Neuropathology Unit, Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid, Spain
| |
Collapse
|
27
|
Aiken J, Holzbaur ELF. Cytoskeletal regulation guides neuronal trafficking to effectively supply the synapse. Curr Biol 2021; 31:R633-R650. [PMID: 34033795 DOI: 10.1016/j.cub.2021.02.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The development and proper function of the brain requires the formation of highly complex neuronal circuitry. These circuits are shaped from synaptic connections between neurons and must be maintained over a lifetime. The formation and continued maintenance of synapses requires accurate trafficking of presynaptic and postsynaptic components along the axon and dendrite, respectively, necessitating deliberate and specialized delivery strategies to replenish essential synaptic components. Maintenance of synaptic transmission also requires readily accessible energy stores, produced in part by localized mitochondria, that are tightly regulated with activity level. In this review, we focus on recent developments in our understanding of the cytoskeletal environment of axons and dendrites, examining how local regulation of cytoskeletal dynamics and organelle trafficking promotes synapse-specific delivery and plasticity. These new insights shed light on the complex and coordinated role that cytoskeletal elements play in establishing and maintaining neuronal circuitry.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Koppers M, Farías GG. Organelle distribution in neurons: Logistics behind polarized transport. Curr Opin Cell Biol 2021; 71:46-54. [PMID: 33706233 DOI: 10.1016/j.ceb.2021.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 12/20/2022]
Abstract
Highly polarized neurons need to carefully regulate the distribution of organelles and other cargoes into their two morphologically and functionally distinct domains, the somatodendritic and axonal compartments, to maintain proper neuron homeostasis. An outstanding question in the field is how organelles reach their correct destination. Long-range transport along microtubules, driven by motors, ensures a fast and controlled availability of organelles in axons and dendrites, but it remains largely unclear what rules govern their transport into the correct compartment. Here, we review the emerging concepts of polarized cargo trafficking in neurons, highlighting the role of microtubule organization, microtubule-associated proteins, and motor proteins and discuss compartment-specific inclusion and exclusion mechanisms as well as the regulation of correct coupling of cargoes to motor proteins.
Collapse
Affiliation(s)
- Max Koppers
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
| | - Ginny G Farías
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands.
| |
Collapse
|
29
|
Gauthier KD, Rocheleau CE. Golgi localization of the LIN-2/7/10 complex points to a role in basolateral secretion of LET-23 EGFR in the Caenorhabditiselegans vulval precursor cells. Development 2021; 148:dev194167. [PMID: 33526581 PMCID: PMC10692275 DOI: 10.1242/dev.194167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/25/2021] [Indexed: 12/28/2022]
Abstract
The evolutionarily conserved LIN-2 (CASK)/LIN-7 (Lin7A-C)/LIN-10 (APBA1) complex plays an important role in regulating spatial organization of membrane proteins and signaling components. In Caenorhabditiselegans, the complex is essential for the development of the vulva by promoting the localization of the sole Epidermal growth factor receptor (EGFR) ortholog LET-23 to the basolateral membrane of the vulva precursor cells where it can specify the vulval cell fate. To understand how the LIN-2/7/10 complex regulates receptor localization, we determined its expression and localization during vulva development. We found that LIN-7 colocalizes with LET-23 EGFR at the basolateral membrane, whereas the LIN-2/7/10 complex colocalizes with LET-23 EGFR at cytoplasmic punctae that mostly overlap with the Golgi. Furthermore, LIN-10 recruits LIN-2, which in turn recruits LIN-7. We demonstrate that the complex forms in vivo with a particularly strong interaction and colocalization between LIN-2 and LIN-7, consistent with them forming a subcomplex. Thus, the LIN-2/7/10 complex forms on the Golgi on which it likely targets LET-23 EGFR trafficking to the basolateral membrane rather than functioning as a tether.
Collapse
Affiliation(s)
- Kimberley D Gauthier
- Division of Endocrinology and Metabolism, Department of Medicine, and Department of Anatomy and Cell Biology, McGill University; and the Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Christian E Rocheleau
- Division of Endocrinology and Metabolism, Department of Medicine, and Department of Anatomy and Cell Biology, McGill University; and the Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
30
|
Xie MX, Cao XY, Zeng WA, Lai RC, Guo L, Wang JC, Xiao YB, Zhang X, Chen D, Liu XG, Zhang XL. ATF4 selectively regulates heat nociception and contributes to kinesin-mediated TRPM3 trafficking. Nat Commun 2021; 12:1401. [PMID: 33658516 PMCID: PMC7930092 DOI: 10.1038/s41467-021-21731-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 02/09/2021] [Indexed: 12/30/2022] Open
Abstract
Effective treatments for patients suffering from heat hypersensitivity are lacking, mostly due to our limited understanding of the pathogenic mechanisms underlying this disorder. In the nervous system, activating transcription factor 4 (ATF4) is involved in the regulation of synaptic plasticity and memory formation. Here, we show that ATF4 plays an important role in heat nociception. Indeed, loss of ATF4 in mouse dorsal root ganglion (DRG) neurons selectively impairs heat sensitivity. Mechanistically, we show that ATF4 interacts with transient receptor potential cation channel subfamily M member-3 (TRPM3) and mediates the membrane trafficking of TRPM3 in DRG neurons in response to heat. Loss of ATF4 also significantly decreases the current and KIF17-mediated trafficking of TRPM3, suggesting that the KIF17/ATF4/TRPM3 complex is required for the neuronal response to heat stimuli. Our findings unveil the non-transcriptional role of ATF4 in the response to heat stimuli in DRG neurons.
Collapse
Affiliation(s)
- Man-Xiu Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Xian-Ying Cao
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
- State Key Laboratory of Marine Resources Utilization of South China Sea, 58 Renmin Avenue, Haikou, China
| | - Wei-An Zeng
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Ren-Chun Lai
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Lan Guo
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
| | - Jun-Chao Wang
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Yi-Bin Xiao
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China
| | - Xi Zhang
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China
| | - Di Chen
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
| | - Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China.
| | - Xiao-Long Zhang
- Medical Research Center of Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Rd. 2, Guangzhou, China.
| |
Collapse
|
31
|
Gauthier KD, Rocheleau CE. LIN-10 can promote LET-23 EGFR signaling and trafficking independently of LIN-2 and LIN-7. Mol Biol Cell 2021; 32:788-799. [PMID: 33566630 PMCID: PMC8108513 DOI: 10.1091/mbc.e20-07-0490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
During Caenorhabditis elegans larval development, an inductive signal mediated by the LET-23 EGFR (epidermal growth factor receptor), specifies three of six vulva precursor cells (VPCs) to adopt vulval cell fates. An evolutionarily conserved complex consisting of PDZ domain-containing scaffold proteins LIN-2 (CASK), LIN-7 (Lin7 or Veli), and LIN-10 (APBA1 or Mint1) (LIN-2/7/10) mediates basolateral LET-23 EGFR localization in the VPCs to permit signal transmission and development of the vulva. We recently found that the LIN-2/7/10 complex likely forms at Golgi ministacks; however, the mechanism through which the complex targets the receptor to the basolateral membrane remains unknown. Here we found that overexpression of LIN-10 or LIN-7 can compensate for loss of their complex components by promoting LET-23 EGFR signaling through previously unknown complex-independent and receptor-dependent pathways. In particular, LIN-10 can independently promote basolateral LET-23 EGFR localization, and its complex-independent function uniquely requires its PDZ domains that also regulate its localization to Golgi. These studies point to a novel complex-independent function for LIN-7 and LIN-10 that broadens our understanding of how this complex regulates targeted sorting of membrane proteins.
Collapse
Affiliation(s)
- Kimberley D Gauthier
- Division of Endocrinology and Metabolism, Department of Medicine, and Department of Anatomy and Cell Biology, McGill University, Montreal, QC H4A 3J1, Canada.,Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Christian E Rocheleau
- Division of Endocrinology and Metabolism, Department of Medicine, and Department of Anatomy and Cell Biology, McGill University, Montreal, QC H4A 3J1, Canada.,Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
32
|
Sun D, Richard MA, Musani SK, Sung YJ, Winkler TW, Schwander K, Chai JF, Guo X, Kilpeläinen TO, Vojinovic D, Aschard H, Bartz TM, Bielak LF, Brown MR, Chitrala K, Hartwig FP, Horimoto AR, Liu Y, Manning AK, Noordam R, Smith AV, Harris SE, Kühnel B, Lyytikäinen LP, Nolte IM, Rauramaa R, van der Most PJ, Wang R, Ware EB, Weiss S, Wen W, Yanek LR, Arking DE, Arnett DK, Barac A, Boerwinkle E, Broeckel U, Chakravarti A, Chen YDI, Cupples LA, Davigulus ML, de las Fuentes L, de Mutsert R, de Vries PS, Delaney JA, Diez Roux AV, Dörr M, Faul JD, Fretts AM, Gallo LC, Grabe HJ, Gu CC, Harris TB, Hartman CC, Heikkinen S, Ikram MA, Isasi C, Johnson WC, Jonas JB, Kaplan RC, Komulainen P, Krieger JE, Levy D, Liu J, Lohman K, Luik AI, Martin LW, Meitinger T, Milaneschi Y, O’Connell JR, Palmas WR, Peters A, Peyser PA, Pulkki-Råback L, Raffel LJ, Reiner AP, Rice K, Robinson JG, Rosendaal FR, Schmidt CO, Schreiner PJ, Schwettmann L, Shikany JM, Shu XO, Sidney S, Sims M, Smith JA, Sotoodehnia N, Strauch K, Tai ES, Taylor KD, Uitterlinden AG, van Duijn CM, Waldenberger M, Wee HL, Wei WB, Wilson G, Xuan D, Yao J, Zeng D, Zhao W, Zhu X, Zonderman AB, Becker DM, Deary IJ, Gieger C, Lakka TA, Lehtimäki T, North KE, Oldehinkel AJ, Penninx BW, Snieder H, Wang YX, Weir DR, Zheng W, Evans MK, Gauderman WJ, Gudnason V, Horta BL, Liu CT, Mook-Kanamori DO, Morrison AC, Pereira AC, Psaty BM, Amin N, Fox ER, Kooperberg C, Sim X, Bierut L, Rotter JI, Kardia SL, Franceschini N, Rao DC, Fornage M. Multi-Ancestry Genome-wide Association Study Accounting for Gene-Psychosocial Factor Interactions Identifies Novel Loci for Blood Pressure Traits. HGG ADVANCES 2021; 2:100013. [PMID: 34734193 PMCID: PMC8562625 DOI: 10.1016/j.xhgg.2020.100013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Psychological and social factors are known to influence blood pressure (BP) and risk of hypertension and associated cardiovascular diseases. To identify novel BP loci, we carried out genome-wide association meta-analyses of systolic, diastolic, pulse, and mean arterial BP taking into account the interaction effects of genetic variants with three psychosocial factors: depressive symptoms, anxiety symptoms, and social support. Analyses were performed using a two-stage design in a sample of up to 128,894 adults from 5 ancestry groups. In the combined meta-analyses of Stages 1 and 2, we identified 59 loci (p value <5e-8), including nine novel BP loci. The novel associations were observed mostly with pulse pressure, with fewer observed with mean arterial pressure. Five novel loci were identified in African ancestry, and all but one showed patterns of interaction with at least one psychosocial factor. Functional annotation of the novel loci supports a major role for genes implicated in the immune response (PLCL2), synaptic function and neurotransmission (LIN7A, PFIA2), as well as genes previously implicated in neuropsychiatric or stress-related disorders (FSTL5, CHODL). These findings underscore the importance of considering psychological and social factors in gene discovery for BP, especially in non-European populations.
Collapse
Affiliation(s)
- Daokun Sun
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Melissa A. Richard
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Solomon K. Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg 93040, Germany
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
- Département de Génomes et Génétique, Institut Pasteur, Paris 75015, France
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA 98195, USA
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
| | - Michael R. Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kumaraswamy Chitrala
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Fernando P. Hartwig
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas RS 96010-610, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1TH, UK
| | - Andrea R.V.R. Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 01246-903, Brazil
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
| | - Albert V. Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48108, USA
- Icelandic Heart Association, Kopavogur 201, Iceland
| | - Sarah E. Harris
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33101, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33101, Finland
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
| | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Rujia Wang
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Erin B. Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17489, Germany
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald 17475, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lisa R. Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dan E. Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donna K. Arnett
- Dean’s Office, University of Kentucky College of Public Health, Lexington, KY 40563, USA
| | - Ana Barac
- MedStar Heart and Vascular Institute, Washington, DC 20010, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aravinda Chakravarti
- Center for Human Genetics and Genomics, New York University School of Medicine, New York, NY 10016, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- NHLBI Framingham Heart Study, Framingham, MA 01702, USA
| | - Martha L. Davigulus
- Division of Minority Health, Department of Epidemiology, University of Illinois, Chicago, IL, USA
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | - Ana V. Diez Roux
- Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, PA 19104, USA
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald 17475, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald 17489, Germany
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Amanda M. Fretts
- Cardiovascular Health Research Unit, Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA 98195, USA
| | - Linda C. Gallo
- Department of Psychology, San Diego State University, San Diego, CA 92182, USA
| | - Hans Jörgen Grabe
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald 17475, Germany
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald 17489, Germany
| | - C. Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Catharina C.A. Hartman
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio 70100, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus 70100, Finland
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Department of Neurology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Carmen Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Jost Bruno Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim 68167, Germany
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY 10461, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
| | - Jose E. Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 01246-903, Brazil
| | - Daniel Levy
- NHLBI Framingham Heart Study, Framingham, MA 01702, USA
- Department of Medicine, Boston University, Boston, MA 02118, USA
| | - Lifelines Cohort Study
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetic Epidemiology, University of Regensburg, Regensburg 93040, Germany
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 119228, Singapore
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA
- Département de Génomes et Génétique, Institut Pasteur, Paris 75015, France
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas RS 96010-610, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol BS8 1TH, UK
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 01246-903, Brazil
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48108, USA
- Icelandic Heart Association, Kopavogur 201, Iceland
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33101, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33101, Finland
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17489, Germany
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald 17475, Germany
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Dean’s Office, University of Kentucky College of Public Health, Lexington, KY 40563, USA
- MedStar Heart and Vascular Institute, Washington, DC 20010, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Center for Human Genetics and Genomics, New York University School of Medicine, New York, NY 10016, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- NHLBI Framingham Heart Study, Framingham, MA 01702, USA
- Division of Minority Health, Department of Epidemiology, University of Illinois, Chicago, IL, USA
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
- College of Pharmacy, University of Manitoba, Winnipeg MB R3E 0T5, Canada
- Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, PA 19104, USA
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald 17489, Germany
- Cardiovascular Health Research Unit, Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA 98195, USA
- Department of Psychology, San Diego State University, San Diego, CA 92182, USA
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald 17489, Germany
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio 70100, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus 70100, Finland
- Department of Neurology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim 68167, Germany
- Beijing Institute of Ophthalmology, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Capital Medical University, Beijing, China
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Medicine, Boston University, Boston, MA 02118, USA
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138632, Singapore
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Human Genetics, Technische Universität München, Munich 81675, Germany
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam 1081 HV, the Netherlands
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of General Medicine, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Neuherberg 85764, Germany
- Faculty of Medicine, Department of Psychology and Logopedics, University of Helsinki, Helsinki 0100, Finland
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
- Departments of Epidemiology and Medicine, University of Iowa, Iowa City, IA 52242, USA
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17489, Germany
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA 94612, USA
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA 98195, USA
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universitat Munchen, Munich, 80539 Germany
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich 85764, Germany
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Jackson Heart Study, School of Public Health, Jackson State University, Jackson, MS 39217, USA
- Department of Biostatistics, University of North Carolina Gilling School of Global Public Health, Chapel Hill, NC 27599, USA
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21201, USA
- German Center for Diabetes Research (DZD e.V.), Neuherberg 85764, Germany
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio 70211, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio 70211, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, University of Tampere, Tampere 33100, Finland
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27516, USA
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA 90007, USA
- Faculty of Medicine, University of Iceland, Reykjavik 102, Iceland
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore 138632, Singapore
| | - Kurt Lohman
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Annemarie I. Luik
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Lisa W. Martin
- Division of Cardiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Human Genetics, Technische Universität München, Munich 81675, Germany
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam 1081 HV, the Netherlands
| | - Jeff R. O’Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Walter R. Palmas
- Division of General Medicine, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Neuherberg 85764, Germany
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
| | - Laura Pulkki-Råback
- Faculty of Medicine, Department of Psychology and Logopedics, University of Helsinki, Helsinki 0100, Finland
| | - Leslie J. Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, Irvine, CA 92697, USA
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Jennifer G. Robinson
- Departments of Epidemiology and Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Frits R. Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
| | - Carsten Oliver Schmidt
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald 17475, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17489, Germany
| | - Pamela J. Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lars Schwettmann
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - James M. Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xiao-ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Stephen Sidney
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA 94612, USA
| | - Mario Sims
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universitat Munchen, Munich, 80539 Germany
| | - E. Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich 85764, Germany
| | - Hwee-Lin Wee
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 119228, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Gregory Wilson
- Jackson Heart Study, School of Public Health, Jackson State University, Jackson, MS 39217, USA
| | - Deng Xuan
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Donglin Zeng
- Department of Biostatistics, University of North Carolina Gilling School of Global Public Health, Chapel Hill, NC 27599, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan B. Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21201, USA
| | - Diane M. Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ian J. Deary
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg 85764, Germany
| | - Timo A. Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Kuopio 70211, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio 70211, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33101, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, University of Tampere, Tampere 33100, Finland
| | - Kari E. North
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27516, USA
| | - Albertine J. Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Brenda W.J.H. Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam 1081 HV, the Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Ya-Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michele K. Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - W. James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 102, Iceland
| | - Bernardo L. Horta
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas RS 96010-610, Brazil
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Dennis O. Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden 2311 EZ, the Netherlands
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 01246-903, Brazil
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA 98195, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Ervin R. Fox
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48108, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC 27516, USA
| | - Dabeeru C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
33
|
Joseph NF, Swarnkar S, Puthanveettil SV. Double Duty: Mitotic Kinesins and Their Post-Mitotic Functions in Neurons. Cells 2021; 10:cells10010136. [PMID: 33445569 PMCID: PMC7827351 DOI: 10.3390/cells10010136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 01/23/2023] Open
Abstract
Neurons, regarded as post-mitotic cells, are characterized by their extensive dendritic and axonal arborization. This unique architecture imposes challenges to how to supply materials required at distal neuronal components. Kinesins are molecular motor proteins that mediate the active delivery of cellular materials along the microtubule cytoskeleton for facilitating the local biochemical and structural changes at the synapse. Recent studies have made intriguing observations that some kinesins that function during neuronal mitosis also have a critical role in post-mitotic neurons. However, we know very little about the function and regulation of such kinesins. Here, we summarize the known cellular and biochemical functions of mitotic kinesins in post-mitotic neurons.
Collapse
Affiliation(s)
- Nadine F. Joseph
- The Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research Institute, La Jolla, CA 92037, USA;
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA;
| | - Supriya Swarnkar
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA;
| | - Sathyanarayanan V Puthanveettil
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA;
- Correspondence: ; Tel.: +1-561-228-3504; Fax: +1-568-228-2249
| |
Collapse
|
34
|
Quinn SM, Vargason T, Pokhrel N, Antony E, Hahn J, Gilbert SP. KIF3A accelerates KIF3C within the kinesin-2 heterodimer to generate symmetrical phosphate release rates for each processive step. J Biol Chem 2021; 296:100020. [PMID: 33144324 PMCID: PMC7948976 DOI: 10.1074/jbc.ra120.015272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/01/2022] Open
Abstract
Heterodimeric KIF3AC is a mammalian kinesin-2 that is highly expressed in the central nervous system and associated with vesicles in neurons. KIF3AC is an intriguing member of the kinesin-2 family because the intrinsic kinetics of KIF3A and KIF3C when expressed as homodimers and analyzed in vitro are distinctively different from each other. For example, the single-molecule velocities of the engineered homodimers KIF3AA and KIF3CC are 293 and 7.5 nm/s, respectively, whereas KIF3AC has a velocity of 186 nm/s. These results led us to hypothesize that heterodimerization alters the intrinsic catalytic properties of the two heads, and an earlier computational analysis predicted that processive steps would alternate between a fast step for KIF3A followed by a slow step for KIF3C resulting in asymmetric stepping. To test this hypothesis directly, we measured the presteady-state kinetics of phosphate release for KIF3AC, KIF3AA, and KIF3CC followed by computational modeling of the KIF3AC phosphate release transients. The results reveal that KIF3A and KIF3C retain their intrinsic ATP-binding and hydrolysis kinetics. Yet within KIF3AC, KIF3A activates the rate of phosphate release for KIF3C such that the coupled steps of phosphate release and dissociation from the microtubule become more similar for KIF3A and KIF3C. These coupled steps are the rate-limiting transition for the ATPase cycle suggesting that within KIF3AC, the stepping kinetics are similar for each head during the processive run. Future work will be directed to define how these properties enable KIF3AC to achieve its physiological functions.
Collapse
Affiliation(s)
- Sean M Quinn
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Troy Vargason
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Nilisha Pokhrel
- The Department of Biology, Marquette University, Milwaukee, Wisconsin, USA
| | - Edwin Antony
- The Department of Biology, Marquette University, Milwaukee, Wisconsin, USA; The Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | - Juergen Hahn
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Chemical & Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA.
| | - Susan P Gilbert
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA.
| |
Collapse
|
35
|
Zhang Z, Li W, Yang G, Lu X, Qi X, Wang S, Cao C, Zhang P, Ren J, Zhao J, Zhang J, Hong S, Tan Y, Burchfield J, Yu Y, Xu T, Yao X, James D, Feng W, Chen Z. CASK modulates the assembly and function of the Mint1/Munc18-1 complex to regulate insulin secretion. Cell Discov 2020; 6:92. [PMID: 33318489 PMCID: PMC7736295 DOI: 10.1038/s41421-020-00216-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
Calcium/calmodulin-dependent protein serine kinase (CASK) is a key player in vesicle transport and release in neurons. However, its precise role, particularly in nonneuronal systems, is incompletely understood. We report that CASK functions as an important regulator of insulin secretion. CASK depletion in mouse islets/β cells substantially reduces insulin secretion and vesicle docking/fusion. CASK forms a ternary complex with Mint1 and Munc18-1, and this event is regulated by glucose stimulation in β cells. The crystal structure of the CASK/Mint1 complex demonstrates that Mint1 exhibits a unique "whip"-like structure that wraps tightly around the CASK-CaMK domain, which contains dual hydrophobic interaction sites. When triggered by CASK binding, Mint1 modulates the assembly of the complex. Further investigation revealed that CASK-Mint1 binding is critical for ternary complex formation, thereby controlling Munc18-1 membrane localization and insulin secretion. Our work illustrates the distinctive molecular basis underlying CASK/Mint1/Munc18-1 complex formation and reveals the importance of the CASK-Mint1-Munc18 signaling axis in insulin secretion.
Collapse
Affiliation(s)
- Zhe Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wei Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guang Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Xuefeng Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Qi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuting Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Can Cao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Peng Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jinqi Ren
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaxu Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Junyi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Sheng Hong
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yan Tan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - James Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yang Yu
- National Center for Protein Science Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuebiao Yao
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Laboratory for Physical Sciences at Nanoscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - David James
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Wei Feng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhengjun Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
36
|
Iwata S, Morikawa M, Takei Y, Hirokawa N. An activity-dependent local transport regulation via degradation and synthesis of KIF17 underlying cognitive flexibility. SCIENCE ADVANCES 2020; 6:6/51/eabc8355. [PMID: 33328231 PMCID: PMC7744090 DOI: 10.1126/sciadv.abc8355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/03/2020] [Indexed: 06/12/2023]
Abstract
Synaptic weight changes among postsynaptic densities within a single dendrite are regulated by the balance between localized protein degradation and synthesis. However, the molecular mechanism via these opposing regulatory processes is still elusive. Here, we showed that the molecular motor KIF17 was locally degraded and synthesized in an N-methyl-d-aspartate receptor (NMDAR)-mediated activity-dependent manner. Accompanied by the degradation of KIF17, its transport was temporarily dampened in dendrites. We also observed that activity-dependent local KIF17 synthesis driven by its 3' untranslated region (3'UTR) occurred at dendritic shafts, and the newly synthesized KIF17 moved along the dendrites. Furthermore, hippocampus-specific deletion of Kif17 3'UTR disrupted KIF17 synthesis induced by fear memory retrieval, leading to impairment in extinction of fear memory. These results indicate that the regulation of the KIF17 transport is driven by the single dendrite-restricted cycle of degradation and synthesis that underlies cognitive flexibility.
Collapse
Affiliation(s)
- Suguru Iwata
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Momo Morikawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Yosuke Takei
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
37
|
The mechanochemistry of the kinesin-2 KIF3AC heterodimer is related to strain-dependent kinetic properties of KIF3A and KIF3C. Proc Natl Acad Sci U S A 2020; 117:15632-15641. [PMID: 32571914 DOI: 10.1073/pnas.1916343117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
KIF3AC is a mammalian neuron-specific kinesin-2 implicated in intracellular cargo transport. It is a heterodimer of KIF3A and KIF3C motor polypeptides which have distinct biochemical and motile properties as engineered homodimers. Single-molecule motility assays show that KIF3AC moves processively along microtubules at a rate faster than expected given the motility rates of the KIF3AA and much slower KIF3CC homodimers. To resolve the stepping kinetics of KIF3A and KIF3C motors in homo- and heterodimeric constructs and determine their transport potential under load, we assayed motor activity using interferometric scattering microscopy and optical trapping. The distribution of stepping durations of KIF3AC molecules is described by a rate (k 1 = 11 s-1) without apparent kinetic asymmetry. Asymmetry was also not apparent under hindering or assisting mechanical loads in the optical trap. KIF3AC shows increased force sensitivity relative to KIF3AA yet is more capable of stepping against mechanical load than KIF3CC. Interestingly, the behavior of KIF3C mirrors prior studies of kinesins with increased interhead compliance. Microtubule gliding assays containing 1:1 mixtures of KIF3AA and KIF3CC result in speeds similar to KIF3AC, suggesting the homodimers mechanically impact each other's motility to reproduce the behavior of the heterodimer. Our observations are consistent with a mechanism in which the stepping of KIF3C can be activated by KIF3A in a strain-dependent manner, similar to application of an assisting load. These results suggest that the mechanochemical properties of KIF3AC can be explained by the strain-dependent kinetics of KIF3A and KIF3C.
Collapse
|
38
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Warnet XL, Bakke Krog H, Sevillano-Quispe OG, Poulsen H, Kjaergaard M. The C-terminal domains of the NMDA receptor: How intrinsically disordered tails affect signalling, plasticity and disease. Eur J Neurosci 2020; 54:6713-6739. [PMID: 32464691 DOI: 10.1111/ejn.14842] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/16/2020] [Accepted: 05/18/2020] [Indexed: 01/14/2023]
Abstract
NMDA receptors are part of the ionotropic glutamate receptor family, and are crucial for neurotransmission and memory. At the cellular level, the effects of activating these receptors include long-term potentiation (LTP) or depression (LTD). The NMDA receptor is a stringently gated cation channel permeable to Ca2+ , and it shares the molecular architecture of a tetrameric ligand-gated ion channel with the other family members. Its subunits, however, have uniquely long cytoplasmic C-terminal domains (CTDs). While the molecular gymnastics of the extracellular domains have been described in exquisite detail, much less is known about the structure and function of these CTDs. The CTDs vary dramatically in length and sequence between receptor subunits, but they all have a composition characteristic of intrinsically disordered proteins. The CTDs affect channel properties, trafficking and downstream signalling output from the receptor, and these functions are regulated by alternative splicing, protein-protein interactions, and post-translational modifications such as phosphorylation and palmitoylation. Here, we review the roles of the CTDs in synaptic plasticity with a focus on biochemical mechanisms. In total, the CTDs play a multifaceted role as a modifier of channel function, a regulator of cellular location and abundance, and signalling scaffold control the downstream signalling output.
Collapse
Affiliation(s)
- Xavier L Warnet
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Helle Bakke Krog
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Oscar G Sevillano-Quispe
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Hanne Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| |
Collapse
|
40
|
Nakayama K, Katoh Y. Architecture of the IFT ciliary trafficking machinery and interplay between its components. Crit Rev Biochem Mol Biol 2020; 55:179-196. [PMID: 32456460 DOI: 10.1080/10409238.2020.1768206] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cilia and flagella serve as cellular antennae and propellers in various eukaryotic cells, and contain specific receptors and ion channels as well as components of axonemal microtubules and molecular motors to achieve their sensory and motile functions. Not only the bidirectional trafficking of specific proteins within cilia but also their selective entry and exit across the ciliary gate is mediated by the intraflagellar transport (IFT) machinery with the aid of motor proteins. The IFT-B complex, which is powered by the kinesin-2 motor, mediates anterograde protein trafficking from the base to the tip of cilia, whereas the IFT-A complex together with the dynein-2 complex mediates retrograde protein trafficking. The BBSome complex connects ciliary membrane proteins to the IFT machinery. Defects in any component of this trafficking machinery lead to abnormal ciliogenesis and ciliary functions, and results in a broad spectrum of disorders, collectively called the ciliopathies. In this review article, we provide an overview of the architectures of the components of the IFT machinery and their functional interplay in ciliary protein trafficking.
Collapse
Affiliation(s)
- Kazuhisa Nakayama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yohei Katoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
41
|
Baliova M, Jursky F. Comparison of SynCAM1/CADM1 PDZ interactions with MUPP1 using mammalian and bacterial pull-down systems. Brain Behav 2020; 10:e01587. [PMID: 32108449 PMCID: PMC7177587 DOI: 10.1002/brb3.1587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 01/20/2020] [Accepted: 02/15/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Synaptic cell adhesion molecule 1 (SynCAM1) also known as cell adhesion molecule 1 (CADM1) is a transmembrane cell adhesion protein that operates in a variety of physiological and pathological cellular contexts, and its interaction with the PDZ signalling protein MUPP1 have been previously implicated in autism spectrum disorder (ASD). METHODS We used in vitro pull-down systems based on the bacterial and mammalian extracts to study SynCAM1/CADM1 PDZ interactions with MUPP1 at various conditions. RESULTS So far, the investigated interaction of SynCAM1/CADM1 with MUPP1 has been mostly attributed to an unspecified region of MUPP1 PDZ domains 1-5 or exclusively to domain 2, using a yeast two-hybrid system. We also confirmed the single interaction of native synaptosomal CADM1 with PDZ domain 2. However, in this work, using recombinant proteins overexpressed in bacteria, we found an in vitro pull-down conditions in which all first five domains and, to a much lesser extent, MUPP1 domains 7 and 11 significantly interacted with the whole C-terminal domain of SynCAM1/CADM1. These PDZ interactions were confirmed by a pull-down assay using the last seven amino acids of the SynCAM1/CADM1 PDZ motif and using two fusion partners. Multiple interactions were additionally replicated using the continuous N-terminal MUPP1 protein fragment, which included first five PDZ domains, containing either intact or mutated domain 2. CONCLUSIONS We hypothesize that multiple interactions might exist in vivo, representing transient low-affinity interactions or alternative binding sites on MUPP1 when domain 2 is occupied or occluded by the interaction with other ligands. This newly identified interactions extend the potential genetic mutations, possibly affecting SynCAM1/CADM1/MUPP1 function. Possible reasons for the absence of some of the identified CADM1 PDZ interactions in mammalian extracts are discussed.
Collapse
Affiliation(s)
- Martina Baliova
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Frantisek Jursky
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
42
|
Vieira M, Yong XLH, Roche KW, Anggono V. Regulation of NMDA glutamate receptor functions by the GluN2 subunits. J Neurochem 2020; 154:121-143. [PMID: 31978252 DOI: 10.1111/jnc.14970] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023]
Abstract
The N-methyl-D-aspartate receptors (NMDARs) are ionotropic glutamate receptors that mediate the flux of calcium (Ca2+ ) into the post-synaptic compartment. Ca2+ influx subsequently triggers the activation of various intracellular signalling cascades that underpin multiple forms of synaptic plasticity. Functional NMDARs are assembled as heterotetramers composed of two obligatory GluN1 subunits and two GluN2 or GluN3 subunits. Four different GluN2 subunits (GluN2A-D) are present throughout the central nervous system; however, they are differentially expressed, both developmentally and spatially, in a cell- and synapse-specific manner. Each GluN2 subunit confers NMDARs with distinct ion channel properties and intracellular trafficking pathways. Regulated membrane trafficking of NMDARs is a dynamic process that ultimately determines the number of NMDARs at synapses, and is controlled by subunit-specific interactions with various intracellular regulatory proteins. Here we review recent progress made towards understanding the molecular mechanisms that regulate the trafficking of GluN2-containing NMDARs, focusing on the roles of several key synaptic proteins that interact with NMDARs via their carboxyl termini.
Collapse
Affiliation(s)
- Marta Vieira
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Xuan Ling Hilary Yong
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
43
|
Alsabban AH, Morikawa M, Tanaka Y, Takei Y, Hirokawa N. Kinesin Kif3b mutation reduces NMDAR subunit NR2A trafficking and causes schizophrenia-like phenotypes in mice. EMBO J 2020; 39:e101090. [PMID: 31746486 PMCID: PMC6939202 DOI: 10.15252/embj.2018101090] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 10/19/2019] [Accepted: 10/22/2019] [Indexed: 01/22/2023] Open
Abstract
The transport of N-methyl-d-aspartate receptors (NMDARs) is crucial for neuronal plasticity and synapse formation. Here, we show that KIF3B, a member of the kinesin superfamily proteins (KIFs), supports the transport of vesicles simultaneously containing NMDAR subunit 2A (NR2A) and the adenomatous polyposis coli (APC) complex. Kif3b+/- neurons exhibited a reduction in dendritic levels of both NR2A and NR2B due to the impaired transport of NR2A and increased degradation of NR2B. In Kif3b+/- hippocampal slices, electrophysiological NMDAR response was found decreased and synaptic plasticity was disrupted, which corresponded to a common feature of schizophrenia (SCZ). The histological features of Kif3b+/- mouse brain also mimicked SCZ features, and Kif3b+/- mice exhibited behavioral defects in prepulse inhibition (PPI), social interest, and cognitive flexibility. Indeed, a mutation of KIF3B was specifically identified in human SCZ patients, which was revealed to be functionally defective in a rescue experiment. Therefore, we propose that KIF3B transports NR2A/APC complex and that its dysfunction is responsible for SCZ pathogenesis.
Collapse
Affiliation(s)
- Ashwaq Hassan Alsabban
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
- Department of Biological ScienceFaculty of SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
- Unit of Neurological DisordersDepartment of Genetic MedicineFaculty of MedicinePrincess Al‐Jawhara Center of Excellence in Research of Hereditary Disorders (PACER.HD)King Abdulaziz UniversityJeddahSaudi Arabia
| | - Momo Morikawa
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Yosuke Tanaka
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Yosuke Takei
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
- Department of Anatomy and NeuroscienceFaculty of MedicineUniversity of TsukubaTsukubaIbarakiJapan
| | - Nobutaka Hirokawa
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
- Center of Excellence in Genome Medicine ResearchKing Abdulaziz UniversityJeddahSaudi Arabia
| |
Collapse
|
44
|
Deeb SK, Guzik-Lendrum S, Jeffrey JD, Gilbert SP. The ability of the kinesin-2 heterodimer KIF3AC to navigate microtubule networks is provided by the KIF3A motor domain. J Biol Chem 2019; 294:20070-20083. [PMID: 31748411 PMCID: PMC6937563 DOI: 10.1074/jbc.ra119.010725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/17/2019] [Indexed: 01/13/2023] Open
Abstract
Heterodimeric kinesin family member KIF3AC is a mammalian kinesin-2 that is highly expressed in the central nervous system and has been implicated in intracellular transport. KIF3AC is unusual in that the motility characteristics of KIF3C when expressed as a homodimer are exceeding slow, whereas homodimeric KIF3AA, as well as KIF3AC, have much faster ATPase kinetics and single molecule velocities. Heterodimeric KIF3AC and homodimeric KIF3AA and KIF3CC are processive, although the run length of KIF3AC exceeds that of KIF3AA and KIF3CC. KIF3C is of particular interest because it exhibits a signature 25-residue insert of glycine and serine residues in loop L11 of its motor domain, and this insert is not present in any other kinesin, suggesting that it confers specific properties to mammalian heterodimeric KIF3AC. To gain a better understanding of the mechanochemical potential of KIF3AC, we pursued a single molecule study to characterize the navigation ability of KIF3AC, KIF3AA, and KIF3CC when encountering microtubule intersections. The results show that all three motors exhibited a preference to remain on the same microtubule when approaching an intersection from the top microtubule, and the majority of track switches occurred from the bottom microtubule onto the top microtubule. Heterodimeric KIF3AC and homodimeric KIF3AA displayed a similar likelihood of switching tracks (36.1 and 32.3%, respectively). In contrast, KIF3CC detached at intersections (67.7%) rather than switch tracks. These results indicate that it is the properties of KIF3A that contribute largely to the ability of KIF3AC to switch microtubule tracks to navigate intersections.
Collapse
Affiliation(s)
- Stephanie K Deeb
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Stephanie Guzik-Lendrum
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Jasper D Jeffrey
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Susan P Gilbert
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| |
Collapse
|
45
|
Blackwell R, Jung D, Bukenberger M, Smith AS. The Impact of Rate Formulations on Stochastic Molecular Motor Dynamics. Sci Rep 2019; 9:18373. [PMID: 31804523 PMCID: PMC6895049 DOI: 10.1038/s41598-019-54344-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/08/2019] [Indexed: 12/19/2022] Open
Abstract
Cells are complex structures which require considerable amounts of organization via transport of large intracellular cargo. While passive diffusion is often sufficiently fast for the transport of smaller cargo, active transport is necessary to organize large structures on the short timescales necessary for biological function. The main mechanism of this transport is by cargo attachment to motors which walk in a directed fashion along intracellular filaments. There are a number of models which seek to describe the motion of motors with attached cargo, from detailed microscopic to coarse phenomenological descriptions. We focus on the intermediate-detailed discrete stochastic hopping models, and explore how cargo transport changes depending on the number of motors, motor interaction, system constraints and rate formulations, which are derived from common thermodynamic assumptions. We find that, despite obeying the same detailed balance constraint, the choice of rate formulation considerably affects the characteristics of the overall motion of the system, with one rate formulation exhibiting novel behavior of loaded motor groups moving faster than a single unloaded motor.
Collapse
Affiliation(s)
- R Blackwell
- PULS group, Physics Department and Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 3, 91058, Erlangen, Germany
| | - D Jung
- PULS group, Physics Department and Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 3, 91058, Erlangen, Germany
| | - M Bukenberger
- PULS group, Physics Department and Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 3, 91058, Erlangen, Germany
| | - A-S Smith
- PULS group, Physics Department and Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 3, 91058, Erlangen, Germany. .,Group for Computational Life Sciences, Division of Physical Chemistry, Insitut Rūder Bošković, Bijenička cesta 54, 10000, Zagreb, Croatia.
| |
Collapse
|
46
|
Yoo KS, Lee K, Oh JY, Lee H, Park H, Park YS, Kim HK. Postsynaptic density protein 95 (PSD-95) is transported by KIF5 to dendritic regions. Mol Brain 2019; 12:97. [PMID: 31753031 PMCID: PMC6873588 DOI: 10.1186/s13041-019-0520-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/04/2019] [Indexed: 11/18/2022] Open
Abstract
Postsynaptic density protein 95 (PSD-95) is a pivotal postsynaptic scaffolding protein in excitatory neurons. Although the transport and regulation of PSD-95 in synaptic regions is well understood, dendritic transport of PSD-95 before synaptic localization still remains to be clarified. To evaluate the role of KIF5, conventional kinesin, in the dendritic transport of PSD-95 protein, we expressed a transport defective form of KIF5A (ΔMD) that does not contain the N-terminal motor domain. Expression of ΔMD significantly decreased PSD-95 level in the dendrites. Consistently, KIF5 was associated with PSD-95 in in vitro and in vivo assays. This interaction was mediated by the C-terminal tail regions of KIF5A and the third PDZ domain of PSD-95. Additionally, the ADPDZ3 (the association domain of NMDA receptor and PDZ3 domain) expression significantly reduced the levels of PSD-95, glutamate receptor 1 (GluA1) in dendrites. The association between PSD-95 and KIF5A was dose-dependent on Staufen protein, suggesting that the Staufen plays a role as a regulatory role in the association. Taken together, our data suggest a new mechanism for dendritic transport of the AMPA receptor-PSD-95.
Collapse
Affiliation(s)
- Ki-Seo Yoo
- Department of Medicine and Microbiology, Graduate Program in Neuroscience, College of Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, South Korea
| | - Kina Lee
- Department of Medicine and Microbiology, Graduate Program in Neuroscience, College of Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, South Korea
| | - Jun-Young Oh
- Department of Medicine and Microbiology, Graduate Program in Neuroscience, College of Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, South Korea.,Department of Structure and Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Hyoeun Lee
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Hyungju Park
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Young Seok Park
- Department of Neurosurgery, Graduate Program in Neuroscience, College of Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, South Korea
| | - Hyong Kyu Kim
- Department of Medicine and Microbiology, Graduate Program in Neuroscience, College of Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, South Korea.
| |
Collapse
|
47
|
Bencsik N, Pusztai S, Borbély S, Fekete A, Dülk M, Kis V, Pesti S, Vas V, Szűcs A, Buday L, Schlett K. Dendritic spine morphology and memory formation depend on postsynaptic Caskin proteins. Sci Rep 2019; 9:16843. [PMID: 31727973 PMCID: PMC6856520 DOI: 10.1038/s41598-019-53317-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
CASK-interactive proteins, Caskin1 and Caskin2, are multidomain neuronal scaffold proteins. Recent data from Caskin1 knockout animals indicated only a mild role of Caskin1 in anxiety and pain perception. In this work, we show that deletion of both Caskins leads to severe deficits in novelty recognition and spatial memory. Ultrastructural analyses revealed a reduction in synaptic profiles and dendritic spine areas of CA1 hippocampal pyramidal neurons of double knockout mice. Loss of Caskin proteins impaired LTP induction in hippocampal slices, while miniature EPSCs in dissociated hippocampal cultures appeared to be unaffected. In cultured Caskin knockout hippocampal neurons, overexpressed Caskin1 was enriched in dendritic spine heads and increased the amount of mushroom-shaped dendritic spines. Chemically induced LTP (cLTP) mediated enlargement of spine heads was augmented in the knockout mice and was not influenced by Caskin1. Immunocytochemistry and immunoprecipitation confirmed that Shank2, a master scaffold of the postsynaptic density, and Caskin1 co-localized within the same complex. Phosphorylation of AMPA receptors was specifically altered by Caskin deficiency and was not elevated by cLTP treatment further. Taken together, our results prove a previously unnoticed postsynaptic role of Caskin scaffold proteins and indicate that Caskins influence learning abilities via regulating spine morphology and AMPA receptor localisation.
Collapse
Affiliation(s)
- Norbert Bencsik
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Szilvia Pusztai
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Sándor Borbély
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.,Institute of Cognitive Neuroscience and Psychology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Fekete
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Metta Dülk
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Viktor Kis
- Department Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Szabolcs Pesti
- Department Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Virág Vas
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Attila Szűcs
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - László Buday
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Department Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
48
|
Yang R, Bostick Z, Garbouchian A, Luisi J, Banker G, Bentley M. A novel strategy to visualize vesicle-bound kinesins reveals the diversity of kinesin-mediated transport. Traffic 2019; 20:851-866. [PMID: 31461551 PMCID: PMC7714429 DOI: 10.1111/tra.12692] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023]
Abstract
In mammals, 15 to 20 kinesins are thought to mediate vesicle transport. Little is known about the identity of vesicles moved by each kinesin or the functional significance of such diversity. To characterize the transport mediated by different kinesins, we developed a novel strategy to visualize vesicle-bound kinesins in living cells. We applied this method to cultured neurons and systematically determined the localization and transport parameters of vesicles labeled by different members of the Kinesin-1, -2, and -3 families. We observed vesicle labeling with nearly all kinesins. Only six kinesins bound vesicles that undergo long-range transport in neurons. Of these, three had an axonal bias (KIF5B, KIF5C and KIF13B), two were unbiased (KIF1A and KIF1Bβ), and one transported only in dendrites (KIF13A). Overall, the trafficking of vesicle-bound kinesins to axons or dendrites did not correspond to their motor domain preference, suggesting that on-vesicle regulation is crucial for kinesin targeting. Surprisingly, several kinesins were associated with populations of somatodendritic vesicles that underwent little long-range transport. This assay should be broadly applicable for investigating kinesin function in many cell types.
Collapse
Affiliation(s)
- Rui Yang
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
- Department of Biochemistry, Duke University, Durham, North Carolina
| | - Zoe Bostick
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| | - Alex Garbouchian
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| | - Julie Luisi
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
| | - Gary Banker
- The Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon
| | - Marvin Bentley
- Department of Biological Sciences and the Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
49
|
Raynaud F, Homburger V, Seveno M, Vigy O, Moutin E, Fagni L, Perroy J. SNAP23-Kif5 complex controls mGlu1 receptor trafficking. J Mol Cell Biol 2019; 10:423-436. [PMID: 29762713 DOI: 10.1093/jmcb/mjy031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/11/2018] [Indexed: 11/13/2022] Open
Abstract
Metabotropic glutamate receptors are expressed at excitatory synapses and control synaptic transmission in mammalian brain. These receptors are involved in numerous patho-physiological functions. However, little is known about the molecular determinants responsible for their intracellular transport and membrane targeting. Here we investigated the nature of the molecular motor and adaptor protein responsible for trafficking and membrane localization of the group I metabotropic glutamate mGlu1 postsynaptic receptor in cultured hippocampal neurons. In proteomic studies, we identified the synaptosome-associated protein 23 (SNAP23) and the molecular motor Kif5 kinesin as proteins interacting with mGlu1 receptor. We showed that SNAP23, but not Kif5, directly interacts with mGlu1 receptor carboxyl terminus. Using a recombination approach to impair or enhance the interaction between SNAP23 and Kif5, we found that the SNAP23-Kif5 complex controls the trafficking of mGlu1 receptor along microtubules. Additional fluorescence recovery after cleavage experiments allowed us to identify a role of the complex in the receptor cell surface targeting. In conclusion, our study indicates that along dendritic processes Kif5-SNAP23 complex contributes to proper mGlu1 receptor trafficking and cell surface expression.
Collapse
Affiliation(s)
| | | | - Martial Seveno
- BioCampus Montpellier, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Oana Vigy
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Enora Moutin
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Laurent Fagni
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Julie Perroy
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| |
Collapse
|
50
|
Göl MF, Erdoğan FF, Bayramov KK, Mehmetbeyoğlu E, Özkul Y. Assessment of genes involved in behavior, learning, memory, and synaptic plasticity following status epilepticus in rats. Epilepsy Behav 2019; 98:101-109. [PMID: 31326869 DOI: 10.1016/j.yebeh.2019.06.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE In this study, it was aimed to evaluate cognitive and behavioral changes after status epilepticus (SE) induced by pentylenetetrazole in immature rats via Morris water maze and open-field area tests and to assess alterations in expression of 84 key genes involved in synaptic plasticity after SE. METHOD The study was conducted on 30 immature rats (12-days old). The rats were assigned into groups as control and experiment (SE) groups. The SE was induced by pentylenetetrazole in 12-days old rats. In addition, experiment group was divided into two groups as mature (n = 8) and immature SE (n = 8) subgroups. Again, the control group was divided into two groups as mature (n = 7) and immature control (n = 7) subgroups. Hippocampal tissue samples were prepared, and expression of 84 key genes involved in synaptic plasticity was assessed in Genome and Stem Cell Center of Erciyes University before behavioral tests in immature rats (22-days old) and after open-filed area and Morris water maze tests in mature rats (72-days old) in both experiment and control groups. RESULTS No significant difference was detected in behavioral tests assessing spatial memory and learning among groups. Significant differences were detected, ARC (activity-regulated cytoskeleton-associated protein), BDNF (brain-derived neurotrophic factor), MAPK1 (mitogen-activated protein kinase 1), NR4A1 (nuclear receptor subfamily 4 group A member 1), PPP3CA (protein phosphatase 3 catalytic subunit alpha), RGS2 (regulator of G protein signaling 2), and TNF (tumor necrosis factor) gene expressions between control and experiment groups in immature rats whereas in ADCY8 (adenylate cyclase 8), BDNF (brain-derived neurotrophic factor), EGR4 (early growth response 4), and KIF17 (kinesin family member 17) gene expressions between control and experiment groups in mature rats. DISCUSSION In this study, differences detected in gene expressions of synaptic plasticity after SE indicate in which steps of synaptic plasticity may be problematic in epileptogenesis. The gene expressions in this study may be considered as potential biomarkers; however, epileptogenesis is a dynamic process and cannot be explained through a single mechanism. Future studies on epileptogenesis and studies specifically designed to evaluate genes detected in our study will further elucidate synaptic plasticity in epilepsy and epileptogenesis.
Collapse
Affiliation(s)
- Mehmet Fatih Göl
- Department of Neurology, Kayseri City Hospital, Kayseri, Turkey.
| | - Füsun Ferda Erdoğan
- Department of Neurology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | | | | | - Yusuf Özkul
- Department of Medical Genetics, Erciyes University Faculty of Medicine, Kayseri, Turkey
| |
Collapse
|