1
|
Lu T, Chen X, Zhang Q, Shang K, Yang X, Xiang W. Vitamin D Relieves Epilepsy Symptoms and Neuroinflammation in Juvenile Mice by Activating the mTOR Signaling Pathway via RAF1: Insights from Network Pharmacology and Molecular Docking Studies. Neurochem Res 2024; 49:2379-2392. [PMID: 38837094 DOI: 10.1007/s11064-024-04176-y] [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: 11/10/2023] [Revised: 03/15/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Epilepsy is a common neurological disorder, and the exploration of potential therapeutic drugs for its treatment is still ongoing. Vitamin D has emerged as a promising treatment due to its potential neuroprotective effects and anti-epileptic properties. This study aimed to investigate the effects of vitamin D on epilepsy and neuroinflammation in juvenile mice using network pharmacology and molecular docking, with a focus on the mammalian target of rapamycin (mTOR) signaling pathway. Experimental mouse models of epilepsy were established through intraperitoneal injection of pilocarpine, and in vitro injury models of hippocampal neurons were induced by glutamate (Glu) stimulation. The anti-epileptic effects of vitamin D were evaluated both in vivo and in vitro. Network pharmacology and molecular docking analysis were used to identify potential targets and regulatory pathways of vitamin D in epilepsy. The involvement of the mTOR signaling pathway in the regulation of mouse epilepsy by vitamin D was validated using rapamycin (RAPA). The levels of inflammatory cytokines (TNF-α, IL-1β, and IL-6) were assessed by enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. The terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) staining was used to analyze the apoptosis of hippocampal neurons. In in vivo experiments, vitamin D reduced the Racine scores of epileptic mice, prolonged the latency of epilepsy, and inhibited the production of TNF-α, IL-1β, and IL-6 in the hippocampus. Furthermore, network pharmacology analysis identified RAF1 as a potential target of vitamin D in epilepsy, which was further confirmed by molecular docking analysis. Additionally, the mTOR signaling pathway was found to be involved in the regulation of mouse epilepsy by vitamin D. In in vitro experiments, Glu stimulation upregulated the expressions of RAF1 and LC3II/LC3I, inhibited mTOR phosphorylation, and induced neuronal apoptosis. Mechanistically, vitamin D activated the mTOR signaling pathway and alleviated mouse epilepsy via RAF1, while the use of the pathway inhibitor RAPA reversed this effect. Vitamin D alleviated epilepsy symptoms and neuroinflammation in juvenile mice by activating the mTOR signaling pathway via RAF1. These findings provided new insights into the molecular mechanisms underlying the anti-epileptic effects of vitamin D and further supported its use as an adjunctive therapy for existing anti-epileptic drugs.
Collapse
Affiliation(s)
- Tiantian Lu
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Xiuling Chen
- Department of Pediatric Medicine, Affiliated Haikou Hospital of Xiangya Medical School Central South University, Haikou, 570208, China
| | - Qin Zhang
- Department of Neurosurgery, Hainan Women and Children's Medical Center, Haikou, 570312, China
| | - Kun Shang
- Institute of Deep-sea Science and Engineering, Chinese Academy of Science, Sanya, 572000, China
| | - Xiaogui Yang
- Department of Neonatology, Haikou Maternal and Child Health Hospital, Haikou, 570203, China
| | - Wei Xiang
- School of Pediatrics, Hainan Medical University, Haikou, 571199, China.
- Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570312, China.
- National Health Commission (NHC) Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, 570216, China.
| |
Collapse
|
2
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
3
|
Li RQ, Yang Y, Qiao L, Yang L, Shen DD, Zhao XJ. KIF2C: An important factor involved in signaling pathways, immune infiltration, and DNA damage repair in tumorigenesis. Biomed Pharmacother 2024; 171:116173. [PMID: 38237349 DOI: 10.1016/j.biopha.2024.116173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUNDS Poorly regulated mitosis and chromosomal instability are common characteristics in malignant tumor cells. Kinesin family member 2 C (KIF2C), also known as mitotic centromere-associated kinesin (MCAK) is an essential component during mitotic regulation. In recent years, KIF2C was shown to be dysregulated in several tumors and was involved in many aspects of tumor self-regulation. Research on KIF2C may be a new direction and target for anti-tumor therapy. OBJECT The article aims at reviewing current literatures and summarizing the research status of KIF2C in malignant tumors as well as the oncogenic signaling pathways associated with KIF2C and its role in immune infiltration. RESULT In this review, we summarize the KIF2C mechanisms and signaling pathways in different malignant tumors, and briefly describe its involvement in pathways related to classical chemotherapeutic drug resistance, such as MEK/ERK, mTOR, Wnt/β-catenin, P53 and TGF-β1/Smad pathways. KIF2C upregulation was shown to promote tumor cell migration, invasion, chemotherapy resistance and inhibit DNA damage repair. It was also highly correlated with microRNAs, and CD4 +T cell and CD8 +T cell tumor immune infiltration. CONCLUSION This review shows that KIF2C may function as a new anticancer drug target with great potential for malignant tumor treatment and the mitigation of chemotherapy resistance.
Collapse
Affiliation(s)
- Rui-Qing Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lin Qiao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China.
| | - Dan-Dan Shen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Jing Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Ruiz-Reig N, Hakanen J, Tissir F. Connecting neurodevelopment to neurodegeneration: a spotlight on the role of kinesin superfamily protein 2A (KIF2A). Neural Regen Res 2024; 19:375-379. [PMID: 37488893 PMCID: PMC10503618 DOI: 10.4103/1673-5374.375298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 07/26/2023] Open
Abstract
Microtubules play a central role in cytoskeletal changes during neuronal development and maintenance. Microtubule dynamics is essential to polarity and shape transitions underlying neural cell division, differentiation, motility, and maturation. Kinesin superfamily protein 2A is a member of human kinesin 13 gene family of proteins that depolymerize and destabilize microtubules. In dividing cells, kinesin superfamily protein 2A is involved in mitotic progression, spindle assembly, and chromosome segregation. In postmitotic neurons, it is required for axon/dendrite specification and extension, neuronal migration, connectivity, and survival. Humans with kinesin superfamily protein 2A mutations suffer from a variety of malformations of cortical development, epilepsy, autism spectrum disorder, and neurodegeneration. In this review, we discuss how kinesin superfamily protein 2A regulates neuronal development and function, and how its deregulation causes neurodevelopmental and neurological disorders.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Université catholique de Louvain, Institute of neuroscience, Brussels, Belgium
| | - Janne Hakanen
- Université catholique de Louvain, Institute of neuroscience, Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of neuroscience, Brussels, Belgium
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
5
|
Costa FV, Zabegalov KN, Kolesnikova TO, de Abreu MS, Kotova MM, Petersen EV, Kalueff AV. Experimental models of human cortical malformations: from mammals to 'acortical' zebrafish. Neurosci Biobehav Rev 2023; 155:105429. [PMID: 37863278 DOI: 10.1016/j.neubiorev.2023.105429] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Human neocortex controls and integrates cognition, emotions, perception and complex behaviors. Aberrant cortical development can be triggered by multiple genetic and environmental factors, causing cortical malformations. Animal models, especially rodents, are a valuable tool to probe molecular and physiological mechanisms of cortical malformations. Complementing rodent studies, the zebrafish (Danio rerio) is an important model organism in biomedicine. Although the zebrafish (like other fishes) lacks neocortex, here we argue that this species can still be used to model various aspects and brain phenomena related to human cortical malformations. We also discuss novel perspectives in this field, covering both advantages and limitations of using mammalian and zebrafish models in cortical malformation research. Summarizing mounting evidence, we also highlight the importance of translationally-relevant insights into the pathogenesis of cortical malformations from animal models, and discuss future strategies of research in the field.
Collapse
Affiliation(s)
- Fabiano V Costa
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Konstantin N Zabegalov
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Tatiana O Kolesnikova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Maria M Kotova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Allan V Kalueff
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; Ural Federal University, Yekaterinburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia.
| |
Collapse
|
6
|
Zhao X, Chen T, Fu B, Fu Z, Xu K, Zhou W. Mutations obstructing ATP's emplacement in KIF2A nucleotide-binding pocket causes parenchymal malformations, motor developmental delay, with intellectual disability. Mol Genet Genomic Med 2023; 11:e2225. [PMID: 37331001 PMCID: PMC10568378 DOI: 10.1002/mgg3.2225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND KIF2A-related tubulinopathy (MIM: #615411) is a very rare disorder that was clinically characterized as microcephaly, epilepsy, motor developmental disorder (MDD), and various malformations of cortical development, but intellectual disability (ID) or global developmental delay (GDD) was rarely reported in the patients. METHODS Quad whole-exome sequencing (WES) was performed on the proband, the older brother, and their parents. Sanger sequencing was used to verify the candidate gene variant. RESULTS The proband, a 23-month-old boy, was previously diagnosed with GDD, and his brother, aged nine years, had ID; both were born to a healthy couple. Quad-WES detected a novel heterozygous KIF2A variant, c.1318G>A (p.G440R), in both the brothers but not in the parents. In-silico analysis revealed that the variants G440R and G318R (which were previously reported in the only reported patient with GDD) lead to markedly enlarged side chains and hinder ATP's emplacement in the NBD pocket. CONCLUSIONS The type of KIF2A variants that sterically hinder ATP emplacing in KIF2A NBD pocket may be associated with the intellectual disability phenotype; however, further studies are needed. Findings in this case also suggest a rare parental germline mosaicism of KIF2A G440R.
Collapse
Affiliation(s)
- Xiuying Zhao
- Department of Pediatricsthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Tao Chen
- Department of NeurologyHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Binsha Fu
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Zhifu Fu
- Department of Children's RehabilitationHainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University)HaikouChina
| | - Kaishou Xu
- Department of RehabilitationGuangzhou Women and Children's Medical Center/National Children's Medical Center for South Central RegionGuangzhouChina
| | - Wei Zhou
- Department of Pediatricsthe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- Neonatal Intensive Care UnitGuangzhou Women and Children's Medical Center/National Children's Medical Center for South Central RegionGuangzhouChina
| |
Collapse
|
7
|
Wan Y, Morikawa M, Morikawa M, Iwata S, Naseer MI, Ahmed Chaudhary AG, Tanaka Y, Hirokawa N. KIF4 regulates neuronal morphology and seizure susceptibility via the PARP1 signaling pathway. J Cell Biol 2023; 222:e202208108. [PMID: 36482480 PMCID: PMC9735414 DOI: 10.1083/jcb.202208108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
Epilepsy is a common neurological disease worldwide, and one of its causes is genetic abnormalities. Here, we identified a point mutation in KIF4A, a member of kinesin superfamily molecular motors, in patients with neurological disorders such as epilepsy, developmental delay, and intellectual disability. KIF4 is involved in the poly (ADP-ribose) polymerase (PARP) signaling pathway, and the mutation (R728Q) strengthened its affinity with PARP1 through elongation of the KIF4 coiled-coil domain. Behavioral tests showed that KIF4-mutant mice exhibited mild developmental delay with lower seizure threshold. Further experiments revealed that the KIF4 mutation caused aberrant morphology in dendrites and spines of hippocampal pyramidal neurons through PARP1-TrkB-KCC2 pathway. Furthermore, supplementing NAD, which activates PARP1, could modulate the TrkB-KCC2 pathway and rescue the seizure susceptibility phenotype of the mutant mice. Therefore, these findings indicate that KIF4 is engaged in a fundamental mechanism regulating seizure susceptibility and could be a potential target for epilepsy treatment.
Collapse
Affiliation(s)
- Yuansong Wan
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Momo Morikawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Manatsu Morikawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Suguru Iwata
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Yosuke Tanaka
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Ruiz-Reig N, García-Sánchez D, Schakman O, Gailly P, Tissir F. Inhibitory synapse dysfunction and epileptic susceptibility associated with KIF2A deletion in cortical interneurons. Front Mol Neurosci 2023; 15:1110986. [PMID: 36733270 PMCID: PMC9887042 DOI: 10.3389/fnmol.2022.1110986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Malformation of cortical development (MCD) is a family of neurodevelopmental disorders, which usually manifest with intellectual disability and early-life epileptic seizures. Mutations in genes encoding microtubules (MT) and MT-associated proteins are one of the most frequent causes of MCD in humans. KIF2A is an atypical kinesin that depolymerizes MT in ATP-dependent manner and regulates MT dynamics. In humans, single de novo mutations in KIF2A are associated with MCD with epileptic seizures, posterior pachygyria, microcephaly, and partial agenesis of corpus callosum. In this study, we conditionally ablated KIF2A in forebrain inhibitory neurons and assessed its role in development and function of inhibitory cortical circuits. We report that adult mice with specific deletion of KIF2A in GABAergic interneurons display abnormal behavior and increased susceptibility to epilepsy. KIF2A is essential for tangential migration of cortical interneurons, their positioning in the cerebral cortex, and for formation of inhibitory synapses in vivo. Our results shed light on how KIF2A deregulation triggers functional alterations in neuronal circuitries and contributes to epilepsy.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium,*Correspondence: Nuria Ruiz-Reig, Fadel Tissir, ;
| | | | - Olivier Schakman
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Philippe Gailly
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Fadel Tissir
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium,College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar,*Correspondence: Nuria Ruiz-Reig, Fadel Tissir, ;
| |
Collapse
|
9
|
Yao M, Qu H, Han Y, Cheng CY, Xiao X. Kinesins in Mammalian Spermatogenesis and Germ Cell Transport. Front Cell Dev Biol 2022; 10:837542. [PMID: 35547823 PMCID: PMC9083010 DOI: 10.3389/fcell.2022.837542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
In mammalian testes, the apical cytoplasm of each Sertoli cell holds up to several dozens of germ cells, especially spermatids that are transported up and down the seminiferous epithelium. The blood-testis barrier (BTB) established by neighboring Sertoli cells in the basal compartment restructures on a regular basis to allow preleptotene/leptotene spermatocytes to pass through. The timely transfer of germ cells and other cellular organelles such as residual bodies, phagosomes, and lysosomes across the epithelium to facilitate spermatogenesis is important and requires the microtubule-based cytoskeleton in Sertoli cells. Kinesins, a superfamily of the microtubule-dependent motor proteins, are abundantly and preferentially expressed in the testis, but their functions are poorly understood. This review summarizes recent findings on kinesins in mammalian spermatogenesis, highlighting their potential role in germ cell traversing through the BTB and the remodeling of Sertoli cell-spermatid junctions to advance spermatid transport. The possibility of kinesins acting as a mediator and/or synchronizer for cell cycle progression, germ cell transit, and junctional rearrangement and turnover is also discussed. We mostly cover findings in rodents, but we also make special remarks regarding humans. We anticipate that this information will provide a framework for future research in the field.
Collapse
Affiliation(s)
- Mingxia Yao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Haoyang Qu
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Yating Han
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang Xiao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China.,Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
10
|
Higgs VE, Das RM. Establishing neuronal polarity: microtubule regulation during neurite initiation. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac007. [PMID: 38596701 PMCID: PMC10913830 DOI: 10.1093/oons/kvac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 04/11/2024]
Abstract
The initiation of nascent projections, or neurites, from the neuronal cell body is the first stage in the formation of axons and dendrites, and thus a critical step in the establishment of neuronal architecture and nervous system development. Neurite formation relies on the polarized remodelling of microtubules, which dynamically direct and reinforce cell shape, and provide tracks for cargo transport and force generation. Within neurons, microtubule behaviour and structure are tightly controlled by an array of regulatory factors. Although microtubule regulation in the later stages of axon development is relatively well understood, how microtubules are regulated during neurite initiation is rarely examined. Here, we discuss how factors that direct microtubule growth, remodelling, stability and positioning influence neurite formation. In addition, we consider microtubule organization by the centrosome and modulation by the actin and intermediate filament networks to provide an up-to-date picture of this vital stage in neuronal development.
Collapse
Affiliation(s)
- Victoria E Higgs
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Raman M Das
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
11
|
Jin Q, Li J, Yang F, Feng L, Du X. Circular RNA circKIF2A Contributes to the Progression of Neuroblastoma Through Regulating PRPS1 Expression by Sponging miR-377-3p. Biochem Genet 2022; 60:1380-1401. [PMID: 35039981 DOI: 10.1007/s10528-021-10174-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
Neuroblastoma is a malignant tumor originating from the primitive neural crest. Circular RNA (circRNA) Kinesin Superfamily Protein 2A (circKIF2A, also known as hsa_circ_0129276) has been reported to be upregulated in neuroblastoma. However, the molecular mechanism of circKIF2A participated in neuroblastoma is poorly defined. We analyzed the expression levels of circKIF2A, microRNA-377-3p (miR-377-3p), and phosphoribosyl pyrophosphate synthetase 1 (PRPS1) in neuroblastoma tissues and cell lines (SK-N-AS and LAN-6) and explored their roles. The expression levels of CircKIF2A and PRPS1 were increased and that of miR-377-3p were decreased in 21 neuroblastoma tissues and cells. Functionally, the silencing of circKIF2A inhibited cell proliferation, migration, invasion, and glycolysis, boosted apoptosis in neuroblastoma cells in vitro, and blocked the growth of subcutaneously transplanted tumors in nude mice. Mechanically, circKIF2A could work as a sponge of miR-377-3p to enhance PRPS1 expression. CircKIF2A knockdown impedes cell proliferation, metastasis, and glycolysis partly by regulating the miR-377-3p/PRPS1 axis, suggesting that targeting circKIF2A can be a feasible therapeutic strategy for neuroblastoma.
Collapse
Affiliation(s)
- Quan Jin
- Department of Pediatrics, Xiantao first people's Hospital Affiliated to Changjiang University, Shazui Street, Xiantao, 433000, Hubei, China
| | - Jianmu Li
- Department of Pediatrics, Xiantao first people's Hospital Affiliated to Changjiang University, Shazui Street, Xiantao, 433000, Hubei, China
| | - Fan Yang
- Department of Pediatrics, Xiantao first people's Hospital Affiliated to Changjiang University, Shazui Street, Xiantao, 433000, Hubei, China
| | - Lingling Feng
- Department of Pediatrics, Xiantao first people's Hospital Affiliated to Changjiang University, Shazui Street, Xiantao, 433000, Hubei, China
| | - Xin Du
- Department of Pediatrics, Xiantao first people's Hospital Affiliated to Changjiang University, Shazui Street, Xiantao, 433000, Hubei, China.
| |
Collapse
|
12
|
Wnt signaling recruits KIF2A to the spindle to ensure chromosome congression and alignment during mitosis. Proc Natl Acad Sci U S A 2021; 118:2108145118. [PMID: 34417301 DOI: 10.1073/pnas.2108145118] [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] [Indexed: 12/16/2022] Open
Abstract
Canonical Wnt signaling plays critical roles in development and tissue renewal by regulating β-catenin target genes. Recent evidence showed that β-catenin-independent Wnt signaling is also required for faithful execution of mitosis. However, the targets and specific functions of mitotic Wnt signaling still remain uncharacterized. Using phosphoproteomics, we identified that Wnt signaling regulates the microtubule depolymerase KIF2A during mitosis. We found that Dishevelled recruits KIF2A via its N-terminal and motor domains, which is further promoted upon LRP6 signalosome formation during cell division. We show that Wnt signaling modulates KIF2A interaction with PLK1, which is critical for KIF2A localization at the spindle. Accordingly, inhibition of basal Wnt signaling leads to chromosome misalignment in somatic cells and pluripotent stem cells. We propose that Wnt signaling monitors KIF2A activity at the spindle poles during mitosis to ensure timely chromosome alignment. Our findings highlight a function of Wnt signaling during cell division, which could have important implications for genome maintenance, notably in stem cells.
Collapse
|
13
|
Modeling Neurodevelopmental Disorders and Epilepsy Caused by Loss of Function of kif2a in Zebrafish. eNeuro 2021; 8:ENEURO.0055-21.2021. [PMID: 34404749 PMCID: PMC8425962 DOI: 10.1523/eneuro.0055-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
In recent years there has been extensive research on malformations of cortical development (MCDs) that result in clinical features like developmental delay, intellectual disability, and drug-resistant epilepsy (DRE). Various studies highlighted the contribution of microtubule-associated genes (including tubulin and kinesin encoding genes) in MCD development. It has been reported that de novo mutations in KIF2A, a member of the kinesin-13 family, are linked to brain malformations and DRE. Although it is known that KIF2A functions by regulating microtubule depolymerization via an ATP-driven process, in vivo implications of KIF2A loss of function remain partly unclear. Here, we present a novel kif2a knock-out zebrafish model, showing hypoactivity, habituation deficits, pentylenetetrazole-induced seizure susceptibility and microcephaly, as well as neuronal cell proliferation defects and increased apoptosis. Interestingly, kif2a−/− larvae survived until adulthood and were fertile. Notably, our kif2a zebrafish knock-out model demonstrated many phenotypic similarities to KIF2A mouse models. This study provides valuable insights into the functional importance of kif2a in zebrafish and phenotypical hallmarks related to KIF2A mutations. Ultimately, this model could be used in a future search for more effective therapies that alleviate the clinical symptoms typically associated with MCDs.
Collapse
|
14
|
Gilet JG, Ivanova EL, Trofimova D, Rudolf G, Meziane H, Broix L, Drouot N, Courraud J, Skory V, Voulleminot P, Osipenko M, Bahi-Buisson N, Yalcin B, Birling MC, Hinckelmann MV, Kwok BH, Allingham JS, Chelly J. Conditional switching of KIF2A mutation provides new insights into cortical malformation pathogeny. Hum Mol Genet 2021; 29:766-784. [PMID: 31919497 DOI: 10.1093/hmg/ddz316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/04/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
By using the Cre-mediated genetic switch technology, we were able to successfully generate a conditional knock-in mouse, bearing the KIF2A p.His321Asp missense point variant, identified in a subject with malformations of cortical development. These mice present with neuroanatomical anomalies and microcephaly associated with behavioral deficiencies and susceptibility to epilepsy, correlating with the described human phenotype. Using the flexibility of this model, we investigated RosaCre-, NestinCre- and NexCre-driven expression of the mutation to dissect the pathophysiological mechanisms underlying neurodevelopmental cortical abnormalities. We show that the expression of the p.His321Asp pathogenic variant increases apoptosis and causes abnormal multipolar to bipolar transition in newborn neurons, providing therefore insights to better understand cortical organization and brain growth defects that characterize KIF2A-related human disorders. We further demonstrate that the observed cellular phenotypes are likely to be linked to deficiency in the microtubule depolymerizing function of KIF2A.
Collapse
Affiliation(s)
- Johan G Gilet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Ekaterina L Ivanova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Daria Trofimova
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Gabrielle Rudolf
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Hamid Meziane
- CNRS UMR 7104, 67400 Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université de Strasbourg, F-67404 Illkirch-Graffenstaden, France
| | - Loic Broix
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Nathalie Drouot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Jeremie Courraud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Valerie Skory
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Paul Voulleminot
- Département de Neurologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Maria Osipenko
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Nadia Bahi-Buisson
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, 75015 Paris, France
| | - Binnaz Yalcin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Marie-Christine Birling
- CNRS UMR 7104, 67400 Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université de Strasbourg, F-67404 Illkirch-Graffenstaden, France
| | - Maria-Victoria Hinckelmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Benjamin H Kwok
- Département de médecine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Jamel Chelly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.,Service de Diagnostic Génétique, Hôpital Civil de Strasbourg, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
15
|
Puri D, Ponniah K, Biswas K, Basu A, Dey S, Lundquist EA, Ghosh-Roy A. Wnt signaling establishes the microtubule polarity in neurons through regulation of Kinesin-13. J Cell Biol 2021; 220:212396. [PMID: 34137792 DOI: 10.1083/jcb.202005080] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuronal polarization is facilitated by the formation of axons with parallel arrays of plus-end-out and dendrites with the nonuniform orientation of microtubules. In C. elegans, the posterior lateral microtubule (PLM) neuron is bipolar with its two processes growing along the anterior-posterior axis under the guidance of Wnt signaling. Here we found that loss of the Kinesin-13 family microtubule-depolymerizing enzyme KLP-7 led to the ectopic extension of axon-like processes from the PLM cell body. Live imaging of the microtubules and axonal transport revealed mixed polarity of the microtubules in the short posterior process, which is dependent on both KLP-7 and the minus-end binding protein PTRN-1. KLP-7 is positively regulated in the posterior process by planar cell polarity components of Wnt involving rho-1/rock to induce mixed polarity of microtubules, whereas it is negatively regulated in the anterior process by the unc-73/ced-10 cascade to establish a uniform microtubule polarity. Our work elucidates how evolutionarily conserved Wnt signaling establishes the microtubule polarity in neurons through Kinesin-13.
Collapse
Affiliation(s)
- Dharmendra Puri
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Keerthana Ponniah
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Kasturi Biswas
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Atrayee Basu
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Swagata Dey
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Erik A Lundquist
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | - Anindya Ghosh-Roy
- Department of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| |
Collapse
|
16
|
Congenital hypothyroidism impairs spine growth of dentate granule cells by downregulation of CaMKIV. Cell Death Discov 2021; 7:143. [PMID: 34127648 PMCID: PMC8203692 DOI: 10.1038/s41420-021-00530-z] [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] [Received: 03/09/2021] [Accepted: 05/23/2021] [Indexed: 12/13/2022] Open
Abstract
Congenital hypothyroidism (CH), a common neonatal endocrine disorder, can result in cognitive deficits if delay in diagnose and treatment. Dentate gyrus (DG) is the severely affected subregion of the hippocampus by the CH, where the dentate granule cells (DGCs) reside in. However, how CH impairs the cognitive function via affecting DGCs and the underlying mechanisms are not fully elucidated. In the present study, the CH model of rat pups was successfully established, and the aberrant dendrite growth of the DGCs and the impaired cognitive behaviors were observed in the offspring. Transcriptome analysis of hippocampal tissues following rat CH successfully identified that calcium/calmodulin-dependent protein kinase IV (CaMKIV) was the prominent regulator involved in mediating deficient growth of DGC dendrites. CaMKIV was shown to be dynamically regulated in the DG subregion of the rats following drug-induced CH. Interference of CaMKIV expression in the primary DGCs significantly reduced the spine density of dendrites, while addition of T3 to the primary DGCs isolated from CH pups could facilitate the spine growth of dendrites. Insights into relevant mechanisms revealed that CH-mediated CaMKIV deficiency resulted in the significant decrease of phosphorylated CREB in DGCs, in association with the abnormality of dendrites. Our results have provided a distinct cell type in hippocampus that is affected by CH, which would be beneficial for the treatment of CH-induced cognitive deficiency.
Collapse
|
17
|
Gao H, Zhang Y, Li Y, Lin X. KIF2A regulates ovarian development via modulating cell cycle progression and vitollogenin levels. INSECT MOLECULAR BIOLOGY 2021; 30:165-175. [PMID: 33251618 DOI: 10.1111/imb.12685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The kinesin superfamily of proteins (KIFs) are microtubule motor proteins that use the hydrolysis of ATP to power directional movement along microtubules. KIFs induce microtubule depolymerization to regulate the length and dynamics of microtubules in a variety of cell processes and structures, including the mitotic and meiotic spindles and centriole and interphase microtubules. KIF plays a significant role in the transport of organelles, protein complexes and mRNAs. The brown planthopper (Nilaparvata lugens) is a major insect pest in rice paddy fields. Ovarian development is regulated by multiple factors, including endocrine factors. The role of KIFs in brown planthopper ovarian development remains unknown. We found that downregulation of KIF2A significantly compromised the development and eclosion of the brown planthopper, delayed ovarian cell cycle progression, disrupted ovarian development, reduced the expression of MCM genes required for DNA replication and significantly reduced the number of nuclei in the follicles. We also found a significant reduction in Vg mRNA and protein levels. We conclude that downregulation of KIF2A disrupts the cell cycle progression of cells. Alternatively, the ovarian phenotype could be an indirect effect of a compromised trophic cord. In summary, KIF2A regulates ovarian development via modulating cell cycle progression and/or vitollogenin transportation.
Collapse
Affiliation(s)
- H Gao
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Y Zhang
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Y Li
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - X Lin
- College of Life Sciences, China Jiliang University, Hangzhou, China
| |
Collapse
|
18
|
Akkaya C, Atak D, Kamacioglu A, Akarlar BA, Guner G, Bayam E, Taskin AC, Ozlu N, Ince-Dunn G. Roles of developmentally regulated KIF2A alternative isoforms in cortical neuron migration and differentiation. Development 2021; 148:dev.192674. [PMID: 33531432 DOI: 10.1242/dev.192674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/18/2021] [Indexed: 11/20/2022]
Abstract
KIF2A is a kinesin motor protein with essential roles in neural progenitor division and axonal pruning during brain development. However, how different KIF2A alternative isoforms function during development of the cerebral cortex is not known. Here, we focus on three Kif2a isoforms expressed in the developing cortex. We show that Kif2a is essential for dendritic arborization in mice and that the functions of all three isoforms are sufficient for this process. Interestingly, only two of the isoforms can sustain radial migration of cortical neurons; a third isoform, lacking a key N-terminal region, is ineffective. By proximity-based interactome mapping for individual isoforms, we identify previously known KIF2A interactors, proteins localized to the mitotic spindle poles and, unexpectedly, also translation factors, ribonucleoproteins and proteins that are targeted to organelles, prominently to the mitochondria. In addition, we show that a KIF2A mutation, which causes brain malformations in humans, has extensive changes to its proximity-based interactome, with depletion of mitochondrial proteins identified in the wild-type KIF2A interactome. Our data raises new insights about the importance of alternative splice variants during brain development.
Collapse
Affiliation(s)
- Cansu Akkaya
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Dila Atak
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Altug Kamacioglu
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Busra Aytul Akarlar
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Gokhan Guner
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Efil Bayam
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Ali Cihan Taskin
- Embryo Manipulation Laboratory, Animal Research Facility, Translational Medicine Research Center, Koç University, 34450 Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey
| | - Gulayse Ince-Dunn
- Department of Molecular Biology and Genetics, Koç University, 34450 Istanbul, Turkey .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
19
|
Identification of Kinesin Family Member 2A (KIF2A) as a Promising Therapeutic Target for Osteosarcoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7102757. [PMID: 33204709 PMCID: PMC7655250 DOI: 10.1155/2020/7102757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/07/2019] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
Background Osteosarcoma is known as a type of common human bone malignancy, and more therapeutic targets are still required to combat this disease. In recent years, the involvement of KIF2A in cancer progression has been widely revealed; however, its potential effect on osteosarcoma development remains unknown. This study is to assess the KIF2A expression levels in human osteosarcoma tissues and explore its potential role in osteosarcoma development. Methods Immunohistochemical (IHC) assays were conducted to evaluate the expression levels of KIF2A in a total of 74 samples of osteosarcoma tissues and adjacent nontumor tissues. According to the staining intensity in tumor tissues, patients were divided into highly expressed and low expression KIF2A groups. The possible links between the KIF2A expression and the clinical pathological features were explored and analyzed, and the effects of KIF2A on osteosarcoma cell proliferation, migration, and invasion were detected through colony formation assay, MTT assay, wound closure assay, and transwell assay, respectively. The effects of KIF2A on tumor growth and metastasis were detected by the use of animal models. Results KIF2A was highly expressed in human osteosarcoma tissues. Meanwhile, KIF2A was obviously correlated to the tumor size (P = 0.001∗) and clinical stage (P = 0.014∗) of osteosarcoma patients. Our results also revealed that the ablation of KIF2A dramatically blocked the proliferation, migration, and invasion capacity of osteosarcoma cells in vitro and blocked tumor growth and metastasis in mice. Conclusions We investigated the involvement of KIF2A in the development and metastasis of osteosarcoma and therefore thought KIF2A as a promising therapeutic target for osteosarcoma treatment.
Collapse
|
20
|
Guillaud L, El-Agamy SE, Otsuki M, Terenzio M. Anterograde Axonal Transport in Neuronal Homeostasis and Disease. Front Mol Neurosci 2020; 13:556175. [PMID: 33071754 PMCID: PMC7531239 DOI: 10.3389/fnmol.2020.556175] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Neurons are highly polarized cells with an elongated axon that extends far away from the cell body. To maintain their homeostasis, neurons rely extensively on axonal transport of membranous organelles and other molecular complexes. Axonal transport allows for spatio-temporal activation and modulation of numerous molecular cascades, thus playing a central role in the establishment of neuronal polarity, axonal growth and stabilization, and synapses formation. Anterograde and retrograde axonal transport are supported by various molecular motors, such as kinesins and dynein, and a complex microtubule network. In this review article, we will primarily discuss the molecular mechanisms underlying anterograde axonal transport and its role in neuronal development and maturation, including the establishment of functional synaptic connections. We will then provide an overview of the molecular and cellular perturbations that affect axonal transport and are often associated with axonal degeneration. Lastly, we will relate our current understanding of the role of axonal trafficking concerning anterograde trafficking of mRNA and its involvement in the maintenance of the axonal compartment and disease.
Collapse
Affiliation(s)
- Laurent Guillaud
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sara Emad El-Agamy
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Miki Otsuki
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
21
|
Gavrilovici C, Jiang Y, Kiroski I, Teskey GC, Rho JM, Nguyen MD. Postnatal Role of the Cytoskeleton in Adult Epileptogenesis. Cereb Cortex Commun 2020; 1:tgaa024. [PMID: 32864616 PMCID: PMC7446231 DOI: 10.1093/texcom/tgaa024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Mutations in cytoskeletal proteins can cause early infantile and childhood epilepsies by misplacing newly born neurons and altering neuronal connectivity. In the adult epileptic brain, cytoskeletal disruption is often viewed as being secondary to aberrant neuronal activity and/or death, and hence simply represents an epiphenomenon. Here, we review the emerging evidence collected in animal models and human studies implicating the cytoskeleton as a potential causative factor in adult epileptogenesis. Based on the emerging evidence, we propose that cytoskeletal disruption may be an important pathogenic mechanism in the mature epileptic brain.
Collapse
Affiliation(s)
- Cezar Gavrilovici
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Ivana Kiroski
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - G Campbell Teskey
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Jong M Rho
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| |
Collapse
|
22
|
Tempes A, Weslawski J, Brzozowska A, Jaworski J. Role of dynein-dynactin complex, kinesins, motor adaptors, and their phosphorylation in dendritogenesis. J Neurochem 2020; 155:10-28. [PMID: 32196676 DOI: 10.1111/jnc.15010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
One of the characteristic features of different classes of neurons that is vital for their proper functioning within neuronal networks is the shape of their dendritic arbors. To properly develop dendritic trees, neurons need to accurately control the intracellular transport of various cellular cargo (e.g., mRNA, proteins, and organelles). Microtubules and motor proteins (e.g., dynein and kinesins) that move along microtubule tracks play an essential role in cargo sorting and transport to the most distal ends of neurons. Equally important are motor adaptors, which may affect motor activity and specify cargo that is transported by the motor. Such transport undergoes very dynamic fine-tuning in response to changes in the extracellular environment and synaptic transmission. Such regulation is achieved by the phosphorylation of motors, motor adaptors, and cargo, among other mechanisms. This review focuses on the contribution of the dynein-dynactin complex, kinesins, their adaptors, and the phosphorylation of these proteins in the formation of dendritic trees by maturing neurons. We primarily review the effects of the motor activity of these proteins in dendrites on dendritogenesis. We also discuss less anticipated mechanisms that contribute to dendrite growth, such as dynein-driven axonal transport and non-motor functions of kinesins.
Collapse
Affiliation(s)
- Aleksandra Tempes
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jan Weslawski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agnieszka Brzozowska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
23
|
Seira O, Liu J, Assinck P, Ramer M, Tetzlaff W. KIF2A characterization after spinal cord injury. Cell Mol Life Sci 2019; 76:4355-4368. [PMID: 31041455 PMCID: PMC11105463 DOI: 10.1007/s00018-019-03116-2] [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: 10/11/2018] [Revised: 04/05/2019] [Accepted: 04/24/2019] [Indexed: 01/23/2023]
Abstract
Axons in the central nervous system (CNS) typically fail to regenerate after injury. This failure is multi-factorial and caused in part by disruption of the axonal cytoskeleton. The cytoskeleton, in particular microtubules (MT), plays a critical role in axonal transport and axon growth during development. In this regard, members of the kinesin superfamily of proteins (KIFs) regulate the extension of primary axons toward their targets and control the growth of collateral branches. KIF2A negatively regulates axon growth through MT depolymerization. Using three different injury models to induce SCI in adult rats, we examined the temporal and cellular expression of KIF2A in the injured spinal cord. We observed a progressive increase of KIF2A expression with maximal levels at 10 days to 8 weeks post-injury as determined by Western blot analysis. KIF2A immunoreactivity was present in axons, spinal neurons and mature oligodendrocytes adjacent to the injury site. Results from the present study suggest that KIF2A at the injured axonal tips may contribute to neurite outgrowth inhibition after injury, and that its increased expression in inhibitory spinal neurons adjacent to the injury site might contribute to an intrinsic wiring-control mechanism associated with neuropathic pain. Further studies will determine whether KIF2A may be a potential target for the development of regeneration-promoting or pain-preventing therapies.
Collapse
Affiliation(s)
- Oscar Seira
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada.
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada.
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
| | - Peggy Assinck
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Graduate Program in Neuroscience, University of British Columbia (UBC), Vancouver, Canada
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Matt Ramer
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
- Department of Surgery, University of British Columbia (UBC), Vancouver, Canada
| |
Collapse
|
24
|
Del Castillo U, Norkett R, Gelfand VI. Unconventional Roles of Cytoskeletal Mitotic Machinery in Neurodevelopment. Trends Cell Biol 2019; 29:901-911. [PMID: 31597609 DOI: 10.1016/j.tcb.2019.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022]
Abstract
At first look, cell division and neurite formation seem to be two different, essential biological processes. However, both processes require extensive reorganization of the cytoskeleton, and especially microtubules. Remarkably, in recent years, independent work from several groups has shown that multiple cytoskeletal components previously considered specific for the mitotic machinery play important roles in neurite initiation and extension. In this review article, we describe how several cytoplasmic and mitotic microtubule motors, components of mitotic kinetochores, and cortical actin participate in reorganization of the microtubule network required to form and maintain axons and dendrites. The emerging similarities between these two biological processes will certainly generate new insights into the mechanisms generating the unique morphology of neurons.
Collapse
Affiliation(s)
- Urko Del Castillo
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Rosalind Norkett
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Vladimir I Gelfand
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|