1
|
Sébastien M, Paquette AL, Prowse ENP, Hendricks AG, Brouhard GJ. Doublecortin restricts neuronal branching by regulating tubulin polyglutamylation. Nat Commun 2025; 16:1749. [PMID: 39966472 PMCID: PMC11836384 DOI: 10.1038/s41467-025-56951-2] [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: 08/19/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Doublecortin is a neuronal microtubule-associated protein that regulates microtubule structure in neurons. Mutations in Doublecortin cause lissencephaly and subcortical band heterotopia by impairing neuronal migration. We use CRISPR/Cas9 to knock-out the Doublecortin gene in induced pluripotent stem cells and differentiate the cells into cortical neurons. DCX-KO neurons show reduced velocities of nuclear movements and an increased number of neurites early in neuronal development, consistent with previous findings. Neurite branching is regulated by a host of microtubule-associated proteins, as well as by microtubule polymerization dynamics. However, EB comet dynamics are unchanged in DCX-KO neurons. Rather, we observe a significant reduction in α-tubulin polyglutamylation in DCX-KO neurons. Polyglutamylation levels and neuronal branching are rescued by expression of Doublecortin or of TTLL11, an α-tubulin glutamylase. Using U2OS cells as an orthogonal model system, we show that DCX and TTLL11 act synergistically to promote polyglutamylation. We propose that Doublecortin acts as a positive regulator of α-tubulin polyglutamylation and restricts neurite branching. Our results indicate an unexpected role for Doublecortin in the homeostasis of the tubulin code.
Collapse
Affiliation(s)
- Muriel Sébastien
- Department of Biology, McGill University, Montréal, QC, Canada
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | | | - Emily N P Prowse
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Adam G Hendricks
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Gary J Brouhard
- Department of Biology, McGill University, Montréal, QC, Canada.
| |
Collapse
|
2
|
Chew YM, Cross RA. Structural switching of tubulin in the microtubule lattice. Biochem Soc Trans 2025; 53:BST20240360. [PMID: 39910801 DOI: 10.1042/bst20240360] [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: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Microtubule (MT) dynamic instability, a cycle of growth, catastrophe, shrinkage and rescue, is driven by the switching of tubulin between two structural states, one stabilised by GTP and the other by GDP. Recent work has uncovered the ancient origins of this structural switch and revealed further fundamental elements of microtubule dynamic instability, whereby switching can be brought about by a range of allosteric effectors, propagate deep within the lattice of assembled MTs, and profoundly affect MT function. Here, we review evidence for structural switching within the MT lattice and discuss current ideas about its mechanisms.
Collapse
Affiliation(s)
- Yean-Ming Chew
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| | - Robert A Cross
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| |
Collapse
|
3
|
Afshar-Sterle S, Carli ALE, O'Keefe R, Tse J, Fischer S, Azimpour AI, Baloyan D, Elias L, Thilakasiri P, Patel O, Ferguson FM, Eissmann MF, Chand AL, Gray NS, Busuttil R, Boussioutas A, Lucet IS, Ernst M, Buchert M. DCLK1 induces a pro-tumorigenic phenotype to drive gastric cancer progression. Sci Signal 2024; 17:eabq4888. [PMID: 39288218 DOI: 10.1126/scisignal.abq4888] [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: 04/11/2022] [Revised: 03/22/2023] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a proposed driver of gastric cancer (GC) that phosphorylates serine and threonine residues. Here, we showed that the kinase activity of DCLK1 orchestrated cancer cell-intrinsic and-extrinsic processes that led to pro-invasive and pro-metastatic reprogramming of GC cells. Inhibition of the kinase activity of DCLK1 reduced the growth of subcutaneous xenograft tumors formed from MKN1 human gastric carcinoma cells in mice and decreased the abundance of the stromal markers α-Sma, vimentin, and collagen. Similar effects were seen in mice with xenograft tumors formed from MKN1 cells expressing a kinase-inactive DCLK1 mutant (MKN1D511N). MKN1D511N cells also had reduced in vitro migratory potential and stemness compared with control cells. Mice orthotopically grafted with MKN1 cells overexpressing DCLK1 (MKN1DCLK1) showed increased invasiveness and had a greater incidence of lung metastases compared with those grafted with control MKN1 cells. Mechanistically, we showed that the chemokine CXCL12 acted downstream of DCLK1 in cultured MKN1 cells and in mice subcutaneously implanted with gastric tumors formed by MKN1DCLK1 cells. Moreover, inhibition of the kinase activity of DCLK1 or the expression of DCLK1D511N reversed the pro-tumorigenic and pro-metastatic phenotype. Together, this study establishes DCLK1 as a broadly acting and potentially targetable promoter of GC.
Collapse
Affiliation(s)
- Shoukat Afshar-Sterle
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Annalisa L E Carli
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Ryan O'Keefe
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Janson Tse
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Stefanie Fischer
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Alexander I Azimpour
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - David Baloyan
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Lena Elias
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Pathum Thilakasiri
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Onisha Patel
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Fleur M Ferguson
- Department of Chemistry and Biochemistry and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Moritz F Eissmann
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Ashwini L Chand
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Rita Busuttil
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Gastroenterology, Alfred Hospital, Melbourne, VIC, Australia
| | - Alex Boussioutas
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Gastroenterology, Alfred Hospital, Melbourne, VIC, Australia
| | - Isabelle S Lucet
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Michael Buchert
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
4
|
Czajkowski ER, Zou Y, Divekar NS, Wignall SM. The doublecortin-family kinase ZYG-8DCLK1 regulates microtubule dynamics and motor-driven forces to promote the stability of C. elegans acentrosomal spindles. PLoS Genet 2024; 20:e1011373. [PMID: 39226307 PMCID: PMC11398696 DOI: 10.1371/journal.pgen.1011373] [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: 04/18/2024] [Revised: 09/13/2024] [Accepted: 07/23/2024] [Indexed: 09/05/2024] Open
Abstract
Although centrosomes help organize spindles in most cell types, oocytes of most species lack these structures. During acentrosomal spindle assembly in C. elegans oocytes, microtubule minus ends are sorted outwards away from the chromosomes where they form poles, but then these outward forces must be balanced to form a stable bipolar structure. Simultaneously, microtubule dynamics must be precisely controlled to maintain spindle length and organization. How forces and dynamics are tuned to create a stable bipolar structure is poorly understood. Here, we have gained insight into this question through studies of ZYG-8, a conserved doublecortin-family kinase; the mammalian homolog of this microtubule-associated protein is upregulated in many cancers and has been implicated in cell division, but the mechanisms by which it functions are poorly understood. We found that ZYG-8 depletion from oocytes resulted in overelongated spindles with pole and midspindle defects. Importantly, experiments with monopolar spindles revealed that ZYG-8 depletion led to excess outward forces within the spindle and suggested a potential role for this protein in regulating the force-generating motor BMK-1/kinesin-5. Further, we found that ZYG-8 is also required for proper microtubule dynamics within the oocyte spindle and that kinase activity is required for its function during both meiosis and mitosis. Altogether, our findings reveal new roles for ZYG-8 in oocytes and provide insights into how acentrosomal spindles are stabilized to promote faithful meiosis.
Collapse
Affiliation(s)
- Emily R. Czajkowski
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Yuntong Zou
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Nikita S. Divekar
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Sarah M. Wignall
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
5
|
Bagdadi N, Wu J, Delaroche J, Serre L, Delphin C, De Andrade M, Carcel M, Nawabi H, Pinson B, Vérin C, Couté Y, Gory-Fauré S, Andrieux A, Stoppin-Mellet V, Arnal I. Stable GDP-tubulin islands rescue dynamic microtubules. J Cell Biol 2024; 223:e202307074. [PMID: 38758215 PMCID: PMC11101955 DOI: 10.1083/jcb.202307074] [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: 07/18/2023] [Revised: 02/26/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024] Open
Abstract
Microtubules are dynamic polymers that interconvert between phases of growth and shrinkage, yet they provide structural stability to cells. Growth involves hydrolysis of GTP-tubulin to GDP-tubulin, which releases energy that is stored within the microtubule lattice and destabilizes it; a GTP cap at microtubule ends is thought to prevent GDP subunits from rapidly dissociating and causing catastrophe. Here, using in vitro reconstitution assays, we show that GDP-tubulin, usually considered inactive, can itself assemble into microtubules, preferentially at the minus end, and promote persistent growth. GDP-tubulin-assembled microtubules are highly stable, displaying no detectable spontaneous shrinkage. Strikingly, islands of GDP-tubulin within dynamic microtubules stop shrinkage events and promote rescues. Microtubules thus possess an intrinsic capacity for stability, independent of accessory proteins. This finding provides novel mechanisms to explain microtubule dynamics.
Collapse
Affiliation(s)
- Nassiba Bagdadi
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Juliette Wu
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Julie Delaroche
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Laurence Serre
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Christian Delphin
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Manon De Andrade
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Marion Carcel
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Homaira Nawabi
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Benoît Pinson
- Metabolic Analyses Service, TBMCore—Université de Bordeaux—CNRS UAR 3427—INSERM US005, Bordeaux, France
| | - Claire Vérin
- Université Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, FR2048, Grenoble, France
| | - Yohann Couté
- Université Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, FR2048, Grenoble, France
| | - Sylvie Gory-Fauré
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Annie Andrieux
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Virginie Stoppin-Mellet
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| | - Isabelle Arnal
- Université Grenoble Alpes, INSERM, U1216, CNRS, CEA, Grenoble Institut Neurosciences (GIN), Grenoble, France
| |
Collapse
|
6
|
Dema A, Charafeddine RA, van Haren J, Rahgozar S, Viola G, Jacobs KA, Kutys ML, Wittmann T. Doublecortin reinforces microtubules to promote growth cone advance in soft environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582626. [PMID: 38464100 PMCID: PMC10925279 DOI: 10.1101/2024.02.28.582626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Doublecortin (DCX) is a microtubule-associated protein critical for brain development. Although most highly expressed in the developing central nervous system, the molecular function of DCX in neuron morphogenesis remains unknown and controversial. We demonstrate that DCX function is intimately linked to its microtubule-binding activity. By using human induced pluripotent stem cell (hiPSC)- derived cortical i 3 Neurons genome engineered to express mEmerald-tagged DCX from the endogenous locus, we find that DCX-MT interactions become highly polarized very early during neuron morphogenesis. DCX becomes enriched only on straight microtubules in advancing growth cones with approximately 120 DCX molecules bound per micrometer of growth cone microtubule. At a similar saturation, microtubule-bound DCX molecules begin to impede lysosome transport, and thus can potentially control growth cone organelle entry. In addition, by comparing control, DCX-mEmerald and knockout DCX -/Y i 3 Neurons, we find that DCX stabilizes microtubules in the growth cone peripheral domain by reducing the microtubule catastrophe frequency and the depolymerization rate. DCX -/Y i 3 Neuron morphogenesis was inhibited in soft microenvironments that mimic the viscoelasticity of brain tissue and DCX -/Y neurites failed to grow toward brain-derived neurotrophic factor (BDNF) gradients. Together with high resolution traction force microscopy data, we propose a model in which DCX-decorated, rigid growth cone microtubules provide intracellular mechanical resistance to actomyosin generated contractile forces in soft physiological environments in which weak and transient adhesion-mediated forces in the growth cone periphery may be insufficient for productive growth cone advance. These data provide a new mechanistic understanding of how DCX mutations cause lissencephaly-spectrum brain malformations by impacting growth cone dynamics during neuron morphogenesis in physiological environments.
Collapse
|
7
|
Czajkowski ER, Divekar NS, Wignall SM. The doublecortin-family kinase ZYG-8 DCLK1 regulates motor activity to achieve proper force balance in C. elegans acentrosomal spindles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568242. [PMID: 38045228 PMCID: PMC10690225 DOI: 10.1101/2023.11.22.568242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Although centrosomes help organize spindles in most cell types, oocytes of most species lack these structures. During acentrosomal spindle assembly in C. elegans oocytes, microtubule minus ends are sorted outwards away from the chromosomes where they form poles, but then these outward forces must be balanced to form a stable bipolar structure. How proper force balance is achieved in these spindles is not known. Here, we have gained insight into this question through studies of ZYG-8, a conserved doublecortin-family kinase; the mammalian homolog of this microtubule-associated protein is upregulated in many cancers and has been implicated in cell division, but the mechanisms by which it functions are poorly understood. Interestingly, we found that ZYG-8 depletion from oocytes resulted in spindles that were over-elongated, suggesting that there was excess outward force following ZYG-8 removal. Experiments with monopolar spindles confirmed this hypothesis and revealed a role for ZYG-8 in regulating the force-generating motor BMK-1/kinesin-5. Importantly, further investigation revealed that kinase activity is required for the function of ZYG-8 in both meiosis and mitosis. Altogether, our results support a model in which ZYG-8 regulates motor-driven forces within the oocyte spindle, thus identifying a new function for a doublecortin-family protein in cell division.
Collapse
Affiliation(s)
- Emily R Czajkowski
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Nikita S Divekar
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Sarah M Wignall
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| |
Collapse
|
8
|
Song X, Cui L, Wu M, Wang S, Song Y, Liu Z, Xue Z, Chen W, Zhang Y, Li H, Sun L, Liang X. DCX-EMAP is a core organizer for the ultrastructure of Drosophila mechanosensory organelles. J Cell Biol 2023; 222:e202209116. [PMID: 37651176 PMCID: PMC10471123 DOI: 10.1083/jcb.202209116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Mechanoreceptor cells develop specialized mechanosensory organelles (MOs), where force-sensitive channels and supporting structures are organized in an orderly manner to detect forces. It is intriguing how MOs are formed. Here, we address this issue by studying the MOs of fly ciliated mechanoreceptors. We show that the main structure of the MOs is a compound cytoskeleton formed of short microtubules and electron-dense materials (EDMs). In a knock-out mutant of DCX-EMAP, this cytoskeleton is nearly absent, suggesting that DCX-EMAP is required for the formation of the MOs and in turn fly mechanotransduction. Further analysis reveals that DCX-EMAP expresses in fly ciliated mechanoreceptors and localizes to the MOs. Moreover, it plays dual roles by promoting the assembly/stabilization of the microtubules and the accumulation of the EDMs in the MOs. Therefore, DCX-EMAP serves as a core ultrastructural organizer of the MOs, and this finding provides novel molecular insights as to how fly MOs are formed.
Collapse
Affiliation(s)
- Xuewei Song
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lihong Cui
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Menghua Wu
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shan Wang
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yinlong Song
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhen Liu
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhaoyu Xue
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wei Chen
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yingjie Zhang
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hui Li
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Landi Sun
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
- Guangzhou Laboratory, Guangzhou, China
| | - Xin Liang
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Sinks MR, Morrison DE, Ramdev RA, Lentzou S, Spritzer MD. Cell proliferation and cell death levels in the dentate gyrus correlate with home range size among adult male meadow voles. Neuroscience 2023:S0306-4522(23)00231-2. [PMID: 37245693 DOI: 10.1016/j.neuroscience.2023.05.018] [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: 11/21/2022] [Revised: 05/07/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
Neurogenesis occurs throughout adulthood within the dentate gyrus, and evidence indicates that these new neurons play a critical role in both spatial and social memory. However, a vast majority of past research on adult neurogenesis has involved experiments with captive mice and rats, making the generalizability of results to natural settings questionable. We assessed the connection between adult neurogenesis and memory by measuring the home range size of wild-caught, free-ranging meadow voles (Microtus pennsylvanicus). Adult male voles (n = 18) were captured, fitted with radio collars, and released back into their natural habitat, where each vole's home range was assessed using 40 radio-telemetry fixes over the course of 5 evenings. Voles were then recaptured, and brain tissue was collected. Cellular markers of cell proliferation (pHisH3, Ki67), neurogenesis (DCX), and pyknosis were labeled on histological sections and then quantified using either fluorescent or light microscopy. Voles with larger home ranges had significantly higher pHisH3+ cell densities within the granule cell layer and subgranular zone (GCL+SGZ) of the dentate gyrus and higher Ki67+ cell densities in the dorsal GCL+SGZ. Voles with larger ranges also had significantly higher pyknotic cell densities in the entire GCL+SGZ and in the dorsal GCL+SGZ. These results support the hypothesis that cell proliferation and cell death within the hippocampus are involved with spatial memory formation. However, a marker of neurogenesis (DCX+) was not correlated with range size, suggesting that there may be selective cellular turnover in the dentate gyrus when a vole is ranging through its environment.
Collapse
Affiliation(s)
- Mark R Sinks
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Daryl E Morrison
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Rajan A Ramdev
- Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Stergiani Lentzou
- Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| | - Mark D Spritzer
- Department of Biology, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A; Program in Neuroscience, Middlebury College, McCardell Bicentennial Hall, Middlebury, VT 05753, U.S.A.
| |
Collapse
|
10
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Carli ALE, Hardy JM, Hoblos H, Ernst M, Lucet IS, Buchert M. Structure-Guided Prediction of the Functional Impact of DCLK1 Mutations on Tumorigenesis. Biomedicines 2023; 11:biomedicines11030990. [PMID: 36979969 PMCID: PMC10046695 DOI: 10.3390/biomedicines11030990] [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: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Doublecortin-like kinase 1 (DCLK1) is a functional serine/threonine (S/T)-kinase and a member of the doublecortin family of proteins which are characterized by their ability to bind to microtubules (MTs). DCLK1 is a proposed cancer driver gene, and its upregulation is associated with poor overall survival in several solid cancer types. However, how DCLK1 associates with MTs and how its kinase function contributes to pro-tumorigenic processes is poorly understood. This review builds on structural models to propose not only the specific functions of the domains but also attempts to predict the impact of individual somatic missense mutations on DCLK1 functions. Somatic missense mutations in DCLK1 are most frequently located within the N-terminal MT binding region and likely impact on the ability of DCLK1 to bind to αβ-tubulin and to polymerize and stabilize MTs. Moreover, the MT binding affinity of DCLK1 is negatively regulated by its auto-phosphorylation, and therefore mutations that affect kinase activity are predicted to indirectly alter MT dynamics. The emerging picture portrays DCLK1 as an MT-associated protein whose interactions with tubulin heterodimers and MTs are tightly controlled processes which, when disrupted, may confer pro-tumorigenic properties.
Collapse
Affiliation(s)
- Annalisa L E Carli
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Joshua M Hardy
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Hanadi Hoblos
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Matthias Ernst
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Isabelle S Lucet
- ACRF Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael Buchert
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
12
|
Zocchi R, Compagnucci C, Bertini E, Sferra A. Deciphering the Tubulin Language: Molecular Determinants and Readout Mechanisms of the Tubulin Code in Neurons. Int J Mol Sci 2023; 24:ijms24032781. [PMID: 36769099 PMCID: PMC9917122 DOI: 10.3390/ijms24032781] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Microtubules (MTs) are dynamic components of the cell cytoskeleton involved in several cellular functions, such as structural support, migration and intracellular trafficking. Despite their high similarity, MTs have functional heterogeneity that is generated by the incorporation into the MT lattice of different tubulin gene products and by their post-translational modifications (PTMs). Such regulations, besides modulating the tubulin composition of MTs, create on their surface a "biochemical code" that is translated, through the action of protein effectors, into specific MT-based functions. This code, known as "tubulin code", plays an important role in neuronal cells, whose highly specialized morphologies and activities depend on the correct functioning of the MT cytoskeleton and on its interplay with a myriad of MT-interacting proteins. In recent years, a growing number of mutations in genes encoding for tubulins, MT-interacting proteins and enzymes that post-translationally modify MTs, which are the main players of the tubulin code, have been linked to neurodegenerative processes or abnormalities in neural migration, differentiation and connectivity. Nevertheless, the exact molecular mechanisms through which the cell writes and, downstream, MT-interacting proteins decipher the tubulin code are still largely uncharted. The purpose of this review is to describe the molecular determinants and the readout mechanisms of the tubulin code, and briefly elucidate how they coordinate MT behavior during critical neuronal events, such as neuron migration, maturation and axonal transport.
Collapse
Affiliation(s)
- Riccardo Zocchi
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Claudia Compagnucci
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Research Hospital, IRCCS, 00146 Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| | - Antonella Sferra
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| |
Collapse
|
13
|
Fu X, Rao L, Li P, Liu X, Wang Q, Son AI, Gennerich A, Liu JSH. Doublecortin and JIP3 are neural-specific counteracting regulators of dynein-mediated retrograde trafficking. eLife 2022; 11:e82218. [PMID: 36476638 PMCID: PMC9799976 DOI: 10.7554/elife.82218] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Mutations in the microtubule (MT)-binding protein doublecortin (DCX) or in the MT-based molecular motor dynein result in lissencephaly. However, a functional link between DCX and dynein has not been defined. Here, we demonstrate that DCX negatively regulates dynein-mediated retrograde transport in neurons from Dcx-/y or Dcx-/y;Dclk1-/- mice by reducing dynein's association with MTs and disrupting the composition of the dynein motor complex. Previous work showed an increased binding of the adaptor protein C-Jun-amino-terminal kinase-interacting protein 3 (JIP3) to dynein in the absence of DCX. Using purified components, we demonstrate that JIP3 forms an active motor complex with dynein and its cofactor dynactin with two dyneins per complex. DCX competes with the binding of the second dynein, resulting in a velocity reduction of the complex. We conclude that DCX negatively regulates dynein-mediated retrograde transport through two critical interactions by regulating dynein binding to MTs and regulating the composition of the dynein motor complex.
Collapse
Affiliation(s)
- Xiaoqin Fu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouChina
| | - Lu Rao
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouChina
| | - Xinglei Liu
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Qi Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
| | - Alexander I Son
- Center for Neuroscience Research, Children's National Research Institute, Children's National HospitalWashingtonUnited States
| | - Arne Gennerich
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Judy Shih-Hwa Liu
- Department of Neurology, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown UniversityProvidenceUnited States
| |
Collapse
|
14
|
Chhetri D, Vengadassalapathy S, Venkadassalapathy S, Balachandran V, Umapathy VR, Veeraraghavan VP, Jayaraman S, Patil S, Iyaswamy A, Palaniyandi K, Gnanasampanthapandian D. Pleiotropic effects of DCLK1 in cancer and cancer stem cells. Front Mol Biosci 2022; 9:965730. [PMID: 36250024 PMCID: PMC9560780 DOI: 10.3389/fmolb.2022.965730] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Doublecortin-like kinase 1 (DCLK1), a protein molecule, has been identified as a tumor stem cell marker in the cancer cells of gastrointestinal, pancreas, and human colon. DCLK1 expression in cancers, such as breast carcinoma, lung carcinoma, hepatic cell carcinoma, tuft cells, and human cholangiocarcinoma, has shown a way to target the DCLK1 gene and downregulate its expression. Several studies have discussed the inhibition of tumor cell proliferation along with neoplastic cell arrest when the DCLK1 gene, which is expressed in both cancer and normal cells, was targeted successfully. In addition, previous studies have shown that DCLK1 plays a vital role in various cancer metastases. The correlation of DCLK1 with numerous stem cell receptors, signaling pathways, and genes suggests its direct or an indirect role in promoting tumorigenesis. Moreover, the impact of DCLK1 was found to be related to the functioning of an oncogene. The downregulation of DCLK1 expression by using targeted strategies, such as embracing the use of siRNA, miRNA, CRISPR/Cas9 technology, nanomolecules, specific monoclonal antibodies, and silencing the pathways regulated by DCLK1, has shown promising results in both in vitro and in vivo studies on gastrointestinal (GI) cancers. In this review, we will discuss about the present understanding of DCLK1 and its role in the progression of GI cancer and metastasis.
Collapse
Affiliation(s)
- Dibyashree Chhetri
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Srinivasan Vengadassalapathy
- Department of Pharmacology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | | | - Varadharaju Balachandran
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Sree Balaji Dental College and Hospital, Chennai, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| | - Ashok Iyaswamy
- Centre for Parkinsons Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| |
Collapse
|
15
|
Hoff KJ, Aiken JE, Gutierrez MA, Franco SJ, Moore JK. Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics. eLife 2022; 11:76189. [PMID: 35511030 PMCID: PMC9236607 DOI: 10.7554/elife.76189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Heterozygous, missense mutations in α- or β-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether a mutation’s impact at the molecular and cellular levels scale with the severity of brain malformation. Here, we focus on two mutations at the valine 409 residue of TUBA1A, V409I, and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A-V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and a decrease in the number of neurite retraction events in primary rat neuronal cultures. These neuronal phenotypes are accompanied by increased microtubule acetylation and polymerization rates. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends in budding yeast and ablate tubulin binding to TOG (tumor overexpressed gene) domains. In each assay tested, the TUBA1A-V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A-V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations disrupt microtubule regulation typically conferred by XMAP215 proteins during neuronal morphogenesis and migration, and this impact on tubulin activity at the molecular level scales with the impact at the cellular and tissue levels. Proteins are molecules made up of long chains of building blocks called amino acids. When a mutation changes one of these amino acids, it can lead to the protein malfunctioning, which can have many effects at the cell and tissue level. Given that human proteins are made up of 20 different amino acids, each building block in a protein could mutate to any of the other 19 amino acids, and each mutations could have different effects. Tubulins are proteins that form microtubules, thin tubes that help give cells their shape and allow them to migrate. These proteins are added or removed to microtubules depending on the cell’s needs, meaning that microtubules can grow or shrink depending on the situation. Mutations in the tubulin proteins have been linked to malformations of varying severities involving the formation of ridges and folds on the surface of the brain, including lissencephaly, pachygyria or polymicrogyria. Hoff et al. wanted to establish links between tubulin mutations and the effects observed at both cell and tissue level in the brain. They focused on two mutations in the tubulin protein TUBA1A that affect the amino acid in position 409 in the protein, which is normally a valine. One of the mutations turns this valine into an amino acid called isoleucine. This mutation is associated with pachygyria, which leads to the brain developing few ridges that are broad and flat. The second mutation turns the valine into an alanine, and is linked to lissencephaly, a more severe condition in which the brain develops no ridges, appearing smooth. Hoff et al. found that both mutations interfere with the development of the brain by stopping neurons from migrating properly, which prevents them from forming the folds in the brain correctly. At the cellular level, the mutations lead to tubulins becoming harder to remove from microtubules, making microtubules more stable than usual. This results in longer microtubules that are harder for the cell to shorten or destroy as needed. Additionally, Hoff et al. showed that the mutant versions of TUBA1A have weaker interactions with a protein called XMAP215, which controls the addition of tubulin to microtubules. This causes the microtubules to grow uncontrollably. Hoff et al. also established that the magnitude of the effects of each mutation on microtubule growth scale with the severity of the disorder they cause. Specifically, cells in which TUBA1A is not mutated have microtubules that grow at a normal rate, and lead to typical brain development. Meanwhile, cells carrying the mutation that turns a valine into an alanine, which is linked to the more severe condition lissencephaly, have microtubules that grow very fast. Finally, cells in which the valine is mutated to an isoleucine – the mutation associated with the less severe malformation pachygyria – have microtubules that grow at an intermediate rate. These findings provide a link between mutations in tubulin proteins and larger effects on cell movement that lead to brain malformations. Additionally, they also link the severity of the malformation to the severity of the microtubule defect caused by each mutation. Further work could examine whether microtubule stabilization is also seen in other similar diseases, which, in the long term, could reveal ways to detect and treat these illnesses.
Collapse
Affiliation(s)
- Katelyn J Hoff
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jayne E Aiken
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Mark A Gutierrez
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Santos J Franco
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jeffrey K Moore
- University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
16
|
Rafiei A, Cruz Tetlalmatzi S, Edrington CH, Lee L, Crowder DA, Saltzberg DJ, Sali A, Brouhard G, Schriemer DC. Doublecortin engages the microtubule lattice through a cooperative binding mode involving its C-terminal domain. eLife 2022; 11:66975. [PMID: 35485925 PMCID: PMC9122500 DOI: 10.7554/elife.66975] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/07/2022] [Indexed: 11/17/2022] Open
Abstract
Doublecortin (DCX) is a microtubule (MT)-associated protein that regulates MT structure and function during neuronal development and mutations in DCX lead to a spectrum of neurological disorders. The structural properties of MT-bound DCX that explain these disorders are incompletely determined. Here, we describe the molecular architecture of the DCX–MT complex through an integrative modeling approach that combines data from X-ray crystallography, cryo-electron microscopy, and a high-fidelity chemical crosslinking method. We demonstrate that DCX interacts with MTs through its N-terminal domain and induces a lattice-dependent self-association involving the C-terminal structured domain and its disordered tail, in a conformation that favors an open, domain-swapped state. The networked state can accommodate multiple different attachment points on the MT lattice, all of which orient the C-terminal tails away from the lattice. As numerous disease mutations cluster in the C-terminus, and regulatory phosphorylations cluster in its tail, our study shows that lattice-driven self-assembly is an important property of DCX.
Collapse
Affiliation(s)
- Atefeh Rafiei
- Department of Chemistry, University of Calgary, Calgary, Canada
| | | | | | - Linda Lee
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - D Alex Crowder
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Daniel J Saltzberg
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, United States
| | - Gary Brouhard
- Department of Biology, McGill University, Montreal, Canada
| | - David C Schriemer
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| |
Collapse
|
17
|
Kase Y, Sato T, Okano Y, Okano H. The GADD45G/p38 MAPK/CDC25B signaling pathway enhances neurite outgrowth by promoting microtubule polymerization. iScience 2022; 25:104089. [PMID: 35497000 PMCID: PMC9042895 DOI: 10.1016/j.isci.2022.104089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/21/2021] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
GADD45G, one of the genes containing the human-specific conserved deletion enhancer-sequence (hCONDEL), has contributed to the evolution of the human cerebrum, but its function in human neurons has not been established. Here, we show that the GADD45G/p38 MAPK/CDC25B signaling pathway promotes neurite outgrowth in human neurons by facilitating microtubule polymerization. This pathway ultimately promotes dephosphorylation of phosphorylated CRMP2 which in turn promotes microtubule assembly. We also found that GADD45G was highly expressed in developing human cerebral specimens. In addition, RK-682, which is the inhibitor of a phosphatase of p38 MAPK and was found in Streptomyces sp., was shown to promote microtubule polymerization and neurite outgrowth by enhancing p38 MAPK/CDC25B signaling. These in vitro and in vivo results indicate that GADD45G/p38 MAPK/CDC25B enhances neurite outgrowth in human neurons.
Collapse
Affiliation(s)
- Yoshitaka Kase
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tsukika Sato
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuji Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Corresponding author
| |
Collapse
|
18
|
Multiple asters organize the yolk microtubule network during dclk2-GFP zebrafish epiboly. Sci Rep 2022; 12:4072. [PMID: 35260695 PMCID: PMC8904445 DOI: 10.1038/s41598-022-07747-7] [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: 11/23/2021] [Accepted: 02/21/2022] [Indexed: 11/25/2022] Open
Abstract
It is known that the organization of microtubule (MT) networks in cells is orchestrated by subcellular structures named MT organizing centers (MTOCs). In this work, we use Light Sheet Fluorescence and Confocal Microscopy to investigate how the MT network surrounding the spherical yolk is arranged in the dclk2-GFP zebrafish transgenic line. We found that during epiboly the MT network is organized by multiple aster-like MTOCS. These structures form rings around the yolk sphere. Importantly, in wt embryos, aster-like MTOCs are only found upon pharmacological or genetic induction. Using our microscopy approach, we underscore the variability in the number of such asters in the transgenic line and report on the variety of global configurations of the yolk MT network. The asters’ morphology, dynamics, and their distribution in the yolk sphere are also analyzed. We propose that these features are tightly linked to epiboly timing and geometry. Key molecules are identified which support this asters role as MTOCs, where MT nucleation and growth take place. We conclude that the yolk MT network of dclk2-GFP transgenic embryos can be used as a model to organize microtubules in a spherical geometry by means of multiple MTOCs.
Collapse
|
19
|
Foster HE, Ventura Santos C, Carter AP. A cryo-ET survey of microtubules and intracellular compartments in mammalian axons. J Cell Biol 2022; 221:e202103154. [PMID: 34878519 PMCID: PMC7612188 DOI: 10.1083/jcb.202103154] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/28/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
The neuronal axon is packed with cytoskeletal filaments, membranes, and organelles, many of which move between the cell body and axon tip. Here, we used cryo-electron tomography to survey the internal components of mammalian sensory axons. We determined the polarity of the axonal microtubules (MTs) by combining subtomogram classification and visual inspection, finding MT plus and minus ends are structurally similar. Subtomogram averaging of globular densities in the MT lumen suggests they have a defined structure, which is surprising given they likely contain the disordered protein MAP6. We found the endoplasmic reticulum in axons is tethered to MTs through multiple short linkers. We surveyed membrane-bound cargos and describe unexpected internal features such as granules and broken membranes. In addition, we detected proteinaceous compartments, including numerous virus-like capsid particles. Our observations outline novel features of axonal cargos and MTs, providing a platform for identification of their constituents.
Collapse
|
20
|
DeOliveira-Mello L, Vicente I, Gonzalez-Nunez V, Santos-Ledo A, Velasco A, Arévalo R, Lara JM, Mack AF. Doublecortin in the Fish Visual System, a Specific Protein of Maturing Neurons. BIOLOGY 2022; 11:biology11020248. [PMID: 35205114 PMCID: PMC8869232 DOI: 10.3390/biology11020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 11/30/2022]
Abstract
Simple Summary Doublecortin (DCX) is an essential protein in the development of the central nervous system and in lamination of the mammalian cortex. It is known that the expression of DCX is restricted to newborn neurons. The visual system of teleost fish has been postulated as an ideal model since it continuously grows throughout the animal’s life. Here, we report a comparative expression analysis of DCX between two teleost fish species as well as a bioinformatic analysis with other animal groups. Our results demonstrate that DCX is very useful for identifying new neurons in the visual systems of Astatotilapia burtoni, but is absent in Danio rerio. Abstract Doublecortin (DCX) is a microtubule associated protein, essential for correct central nervous system development and lamination in the mammalian cortex. It has been demonstrated to be expressed in developing—but not in mature—neurons. The teleost visual system is an ideal model to study mechanisms of adult neurogenesis due to its continuous life-long growth. Here, we report immunohistochemical, in silico, and western blot analysis to detect the DCX protein in the visual system of teleost fish. We clearly determined the expression of DCX in newly generated cells in the retina of the cichlid fish Astatotilapia burtoni, but not in the cyprinid fish Danio rerio. Here, we show that DCX is not associated with migrating cells but could be related to axonal growth. This work brings to light the high conservation of DCX sequences between different evolutionary groups, which make it an ideal marker for maturing neurons in various species. The results from different techniques corroborate the absence of DCX expression in zebrafish. In A. burtoni, DCX is very useful for identifying new neurons in the transition zone of the retina. In addition, this marker can be applied to follow axons from maturing neurons through the neural fiber layer, optic nerve head, and optic nerve.
Collapse
Affiliation(s)
- Laura DeOliveira-Mello
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| | - Isabel Vicente
- Department of Agriculture, Food and Environment, University of Pisa, 56124 Pisa, Italy;
| | - Veronica Gonzalez-Nunez
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Adrian Santos-Ledo
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Almudena Velasco
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Rosario Arévalo
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Juan M. Lara
- Institute of Neurosciences of Castilla and León, University of Salamanca, 37007 Salamanca, Spain; (V.G.-N.); (A.S.-L.); (A.V.); (R.A.); (J.M.L.)
- Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Andreas F. Mack
- Institute of Clinical Anatomy and Cell Analysis, Eberhard-Karls Universität Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
21
|
Amygdala DCX and blood Cdk14 are implicated as cross-species indicators of individual differences in fear, extinction, and resilience to trauma exposure. Mol Psychiatry 2022; 27:956-966. [PMID: 34728797 PMCID: PMC9058038 DOI: 10.1038/s41380-021-01353-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 09/18/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022]
Abstract
Doublecortin (DCX) has long been implicated in, and employed as a marker for, neurogenesis, yet little is known about its function in non-neurogenic brain regions, including the amygdala. This study sought first to explore, in rodents, whether fear learning and extinction modulate amygdala DCX expression and, second, to assess the utility of peripheral DCX correlates as predictive biomarkers of trauma response in rodents and humans. Pavlovian conditioning was found to alter DCX protein levels in mice 24 h later, resulting in higher DCX expression associated with enhanced learning in paradigms examining both the acquisition and extinction of fear (p < 0.001). This, in turn, is associated with differences in freezing on subsequent fear expression tests, and the same relationship between DCX and fear extinction was replicated in rats (p < 0.001), with higher amygdala DCX levels associated with more rapid extinction of fear. RNAseq of amygdala and blood from mice identified 388 amygdala genes that correlated with DCX (q < 0.001) and which gene ontology analyses revealed were significantly over-represented for neurodevelopmental processes. In blood, DCX-correlated genes included the Wnt signaling molecule Cdk14 which was found to predict freezing during both fear acquisition (p < 0.05) and brief extinction protocols (p < 0.001). High Cdk14 measured in blood immediately after testing was also associated with less freezing during fear expression testing (p < 0.01). Finally, in humans, Cdk14 expression in blood taken shortly after trauma was found to predict resilience in males for up to a year post-trauma (p < 0.0001). These data implicate amygdala DCX in fear learning and suggest that Cdk14 may serve as a predictive biomarker of trauma response.
Collapse
|
22
|
McAlear TS, Bechstedt S. The mitotic spindle protein CKAP2 potently increases formation and stability of microtubules. eLife 2022; 11:72202. [PMID: 35029146 PMCID: PMC8798059 DOI: 10.7554/elife.72202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cells increase microtubule dynamics to make large rearrangements to their microtubule cytoskeleton during cell division. Changes in microtubule dynamics are essential for the formation and function of the mitotic spindle, and misregulation can lead to aneuploidy and cancer. Using in vitro reconstitution assays we show that the mitotic spindle protein Cytoskeleton-Associated Protein 2 (CKAP2) has a strong effect on nucleation of microtubules by lowering the critical tubulin concentration 100-fold. CKAP2 increases the apparent rate constant ka of microtubule growth by 50-fold and increases microtubule growth rates. In addition, CKAP2 strongly suppresses catastrophes. Our results identify CKAP2 as the most potent microtubule growth factor to date. These finding help explain CKAP2's role as an important spindle protein, proliferation marker, and oncogene.
Collapse
Affiliation(s)
- Thomas S McAlear
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| |
Collapse
|
23
|
Roy A, Kundu M, Chakrabarti S, Patel DR, Pahan K. Oleamide, a Sleep-Inducing Supplement, Upregulates Doublecortin in Hippocampal Progenitor Cells via PPARα. J Alzheimers Dis 2021; 84:1747-1762. [PMID: 34744082 PMCID: PMC10075226 DOI: 10.3233/jad-215124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Doublecortin (DCX), a microtubule associated protein, has emerged as a central biomarker of hippocampal neurogenesis. However, molecular mechanisms by which DCX is regulated are poorly understood. OBJECTIVE Since sleep is involved with the acquisition of memory and oleamide or 9-Octadecenamide (OCT) is a sleep-inducing supplement in human, we examined whether OCT could upregulate DCX in hippocampal progenitor cells (HPCs). METHODS We employed real-time PCR, western blot, immunostaining, chromatin immunoprecipitation, lentiviral transduction in HPCs, and the calcium influx assay. RESULTS OCT directly upregulated the transcription of Dcx in HPCs via activation of peroxisome proliferator-activated receptor α (PPARα), a lipid-lowering transcription factor. We observed that, HPCs of Ppara-null mice displayed significant impairment in DCX expression and neuronal differentiation as compared to that of wild-type mice. Interestingly, treatment with OCT stimulated the differentiation process of HPCs in wild-type, but not Ppara-null mice. Reconstruction of PPARα in mouse Ppara-null HPCs restored the expression of DCX, which was further stimulated with OCT treatment. In contrast, a dominant-negative mutant of PPARα significantly attenuated the stimulatory effect of OCT on DCX expression and suppressed neuronal differentiation of human neural progenitor cells. Furthermore, RNA microarray, STRING, chromatin immunoprecipitation, site-directed mutagenesis, and promoter reporter assay have identified DCX as a new target of PPARα. CONCLUSION These results indicate that OCT, a sleep supplement, directly controls the expression of DCX and suggest that OCT may be repurposed for stimulating the hippocampal neurogenesis.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sudipta Chakrabarti
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Dhruv R Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
24
|
Hakanen J, Parmentier N, Sommacal L, Garcia-Sanchez D, Aittaleb M, Vertommen D, Zhou L, Ruiz-Reig N, Tissir F. The Celsr3-Kif2a axis directs neuronal migration in the postnatal brain. Prog Neurobiol 2021; 208:102177. [PMID: 34582949 DOI: 10.1016/j.pneurobio.2021.102177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/12/2021] [Accepted: 09/20/2021] [Indexed: 12/27/2022]
Abstract
The tangential migration of immature neurons in the postnatal brain involves consecutive migration cycles and depends on constant remodeling of the cell cytoskeleton, particularly in the leading process (LP). Despite the identification of several proteins with permissive and empowering functions, the mechanisms that specify the direction of migration remain largely unknown. Here, we report that planar cell polarity protein Celsr3 orients neuroblasts migration from the subventricular zone (SVZ) to olfactory bulb (OB). In Celsr3-forebrain conditional knockout mice, neuroblasts loose directionality and few can reach the OB. Celsr3-deficient neuroblasts exhibit aberrant branching of LP, de novo LP formation, and decreased growth rate of microtubules (MT). Mechanistically, we show that Celsr3 interacts physically with Kif2a, a MT depolymerizing protein and that conditional inactivation of Kif2a in the forebrain recapitulates the Celsr3 knockout phenotype. Our findings provide evidence that Celsr3 and Kif2a cooperatively specify the directionality of neuroblasts tangential migration in the postnatal brain.
Collapse
Affiliation(s)
- Janne Hakanen
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Nicolas Parmentier
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Leonie Sommacal
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Dario Garcia-Sanchez
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Mohamed Aittaleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Didier Vertommen
- Université catholique de Louvain, de Duve Institute, Massprot Platform, Brussels, Belgium
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, PR China
| | - Nuria Ruiz-Reig
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
25
|
Agulto RL, Rogers MM, Tan TC, Ramkumar A, Downing AM, Bodin H, Castro J, Nowakowski DW, Ori-McKenney KM. Autoregulatory control of microtubule binding in doublecortin-like kinase 1. eLife 2021; 10:e60126. [PMID: 34310279 PMCID: PMC8352597 DOI: 10.7554/elife.60126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/22/2021] [Indexed: 12/16/2022] Open
Abstract
The microtubule-associated protein, doublecortin-like kinase 1 (DCLK1), is highly expressed in a range of cancers and is a prominent therapeutic target for kinase inhibitors. The physiological roles of DCLK1 kinase activity and how it is regulated remain elusive. Here, we analyze the role of mammalian DCLK1 kinase activity in regulating microtubule binding. We found that DCLK1 autophosphorylates a residue within its C-terminal tail to restrict its kinase activity and prevent aberrant hyperphosphorylation within its microtubule-binding domain. Removal of the C-terminal tail or mutation of this residue causes an increase in phosphorylation within the doublecortin domains, which abolishes microtubule binding. Therefore, autophosphorylation at specific sites within DCLK1 has diametric effects on the molecule's association with microtubules. Our results suggest a mechanism by which DCLK1 modulates its kinase activity to tune its microtubule-binding affinity. These results provide molecular insights for future therapeutic efforts related to DCLK1's role in cancer development and progression.
Collapse
Affiliation(s)
- Regina L Agulto
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Melissa M Rogers
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Tracy C Tan
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Amrita Ramkumar
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Ashlyn M Downing
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Hannah Bodin
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Julia Castro
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | | | | |
Collapse
|
26
|
Lynch EM, Robertson S, FitzGibbons C, Reilly M, Switalski C, Eckardt A, Tey SR, Hayakawa K, Suzuki M. Transcriptome analysis using patient iPSC-derived skeletal myocytes: Bet1L as a new molecule possibly linked to neuromuscular junction degeneration in ALS. Exp Neurol 2021; 345:113815. [PMID: 34310943 DOI: 10.1016/j.expneurol.2021.113815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease in which patients gradually become paralyzed due to loss of motor function. Many genetically inheritable mutations have been linked to ALS; however, the majority of ALS patients are considered sporadic. Therefore, there is a need for a common therapy that is effective for all ALS patients. Although there is evidence of the disease beginning in the periphery at the neuromuscular junction (NMJ), the specific processes involved in skeletal muscle and at the NMJ are still largely unknown. To study common disease mechanisms in ALS skeletal muscle, we performed RNA sequencing of skeletal myocytes differentiated from induced pluripotent stem cells (iPSCs) derived from familial ALS (with C9ORF72, SOD1, or TARDBP mutations) and sporadic ALS patients. Compared to healthy control lines, the myocytes from all ALS lines showed downregulation of four genes: BET1L, DCX, GPC3, and HNRNPK. We next measured the expression levels of these four genes in hind limb muscle samples from a rat model of familial ALS (SOD1G93A transgenic) and found that only the Bet1L gene, which encodes Bet1 Golgi Vesicular Membrane Trafficking Protein Like, was commonly downregulated. Bet1L protein appeared to be localized to the basal lamina of the NMJ, with decreased expression over time in SOD1G93A transgenic rats. Importantly, the expression levels began to decrease early in the disease process. Our results indicate that loss of Bet1L at the NMJ could be of interest for better understanding ALS disease progression.
Collapse
Affiliation(s)
- Eileen M Lynch
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Claire FitzGibbons
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Megan Reilly
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Colton Switalski
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Adam Eckardt
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Sin-Ruow Tey
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Koji Hayakawa
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA; Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
27
|
Carli ALE, Afshar-Sterle S, Rai A, Fang H, O'Keefe R, Tse J, Ferguson FM, Gray NS, Ernst M, Greening DW, Buchert M. Cancer stem cell marker DCLK1 reprograms small extracellular vesicles toward migratory phenotype in gastric cancer cells. Proteomics 2021; 21:e2000098. [PMID: 33991177 DOI: 10.1002/pmic.202000098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/15/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a putative cancer stem cell marker, a promising diagnostic and prognostic maker for malignant tumors and a proposed driver gene for gastric cancer (GC). DCLK1 overexpression in a majority of solid cancers correlates with lymph node metastases, advanced disease and overall poor-prognosis. In cancer cells, DCLK1 expression has been shown to promote epithelial-to-mesenchymal transition (EMT), driving disruption of cell-cell adhesion, cell migration and invasion. Here, we report that DCLK1 influences small extracellular vesicle (sEV/exosome) biogenesis in a kinase-dependent manner. sEVs isolated from DCLK1 overexpressing human GC cell line MKN1 (MKN1OE -sEVs), promote the migration of parental (non-transfected) MKN1 cells (MKN1PAR ). Quantitative proteome analysis of MKN1OE -sEVs revealed enrichment in migratory and adhesion regulators (STRAP, CORO1B, BCAM, COL3A, CCN1) in comparison to MKN1PAR -sEVs. Moreover, using DCLK1-IN-1, a specific small molecule inhibitor of DCLK1, we reversed the increase in sEV size and concentration in contrast to other EV subtypes, as well as kinase-dependent cargo selection of proteins involved in EV biogenesis (KTN1, CHMP1A, MYO1G) and migration and adhesion processes (STRAP, CCN1). Our findings highlight a specific role of DCLK1-kinase dependent cargo selection for sEVs and shed new light on its role as a regulator of signaling in gastric tumorigenesis.
Collapse
Affiliation(s)
- Annalisa L E Carli
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Shoukat Afshar-Sterle
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia
| | - Ryan O'Keefe
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Janson Tse
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Ernst
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Buchert
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
28
|
Sorrells SF, Paredes MF, Zhang Z, Kang G, Pastor-Alonso O, Biagiotti S, Page CE, Sandoval K, Knox A, Connolly A, Huang EJ, Garcia-Verdugo JM, Oldham MC, Yang Z, Alvarez-Buylla A. Positive Controls in Adults and Children Support That Very Few, If Any, New Neurons Are Born in the Adult Human Hippocampus. J Neurosci 2021; 41:2554-2565. [PMID: 33762407 PMCID: PMC8018729 DOI: 10.1523/jneurosci.0676-20.2020] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 01/19/2023] Open
Abstract
Adult hippocampal neurogenesis was originally discovered in rodents. Subsequent studies identified the adult neural stem cells and found important links between adult neurogenesis and plasticity, behavior, and disease. However, whether new neurons are produced in the human dentate gyrus (DG) during healthy aging is still debated. We and others readily observe proliferating neural progenitors in the infant hippocampus near immature cells expressing doublecortin (DCX), but the number of such cells decreases in children and few, if any, are present in adults. Recent investigations using dual antigen retrieval find many cells stained by DCX antibodies in adult human DG. This has been interpreted as evidence for high rates of adult neurogenesis, even at older ages. However, most of these DCX-labeled cells have mature morphology. Furthermore, studies in the adult human DG have not found a germinal region containing dividing progenitor cells. In this Dual Perspectives article, we show that dual antigen retrieval is not required for the detection of DCX in multiple human brain regions of infants or adults. We review prior studies and present new data showing that DCX is not uniquely expressed by newly born neurons: DCX is present in adult amygdala, entorhinal and parahippocampal cortex neurons despite being absent in the neighboring DG. Analysis of available RNA-sequencing datasets supports the view that DG neurogenesis is rare or absent in the adult human brain. To resolve the conflicting interpretations in humans, it is necessary to identify and visualize dividing neuronal precursors or develop new methods to evaluate the age of a neuron at the single-cell level.
Collapse
Affiliation(s)
- Shawn F Sorrells
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Mercedes F Paredes
- Department of Neurology, University of California San Francisco, San Francisco, California 94143
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, P.R. 200032 China
| | - Gugene Kang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94143
| | - Oier Pastor-Alonso
- Department of Neurology, University of California San Francisco, San Francisco, California 94143
| | - Sean Biagiotti
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Chloe E Page
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Kadellyn Sandoval
- Department of Neurology, University of California San Francisco, San Francisco, California 94143
| | - Anthony Knox
- Department of Pathology, University of California San Francisco, San Francisco, California 94143
| | - Andrew Connolly
- Department of Pathology, University of California San Francisco, San Francisco, California 94143
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, San Francisco, California 94143
| | - Jose Manuel Garcia-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Valencia 46980, Spain
| | - Michael C Oldham
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94143
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, P.R. 200032 China
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California 94143
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California 94143
| |
Collapse
|
29
|
Li P, Li L, Yu B, Wang X, Wang Q, Lin J, Zheng Y, Zhu J, He M, Xia Z, Tu M, Liu JS, Lin Z, Fu X. Doublecortin facilitates the elongation of the somatic Golgi apparatus into proximal dendrites. Mol Biol Cell 2021; 32:422-434. [PMID: 33405953 PMCID: PMC8098852 DOI: 10.1091/mbc.e19-09-0530] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in the doublecortin (DCX) gene, which encodes a microtubule (MT)-binding protein, cause human cortical malformations, including lissencephaly and subcortical band heterotopia. A deficiency in DCX and DCX-like kinase 1 (DCLK1), a functionally redundant and structurally similar cognate of DCX, decreases neurite length and increases the number of primary neurites directly arising from the soma. The underlying mechanism is not completely understood. In this study, the elongation of the somatic Golgi apparatus into proximal dendrites, which have been implicated in dendrite patterning, was significantly decreased in the absence of DCX/DCLK1. Phosphorylation of DCX at S47 or S327 was involved in this process. DCX deficiency shifted the distribution of CLASP2 proteins to the soma from the dendrites. In addition to CLASP2, dynein and its cofactor JIP3 were abnormally distributed in DCX-deficient neurons. The association between JIP3 and dynein was significantly increased in the absence of DCX. Down-regulation of CLASP2 or JIP3 expression with specific shRNAs rescued the Golgi phenotype observed in DCX-deficient neurons. We conclude that DCX regulates the elongation of the Golgi apparatus into proximal dendrites through MT-associated proteins and motors.
Collapse
Affiliation(s)
- Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Luyao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Binyuan Yu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xinye Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qi Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jingjing Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yihui Zheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jinjin Zhu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Minzhi He
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhaonan Xia
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Mengjing Tu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Judy S Liu
- Department of Neurology, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903
| | - Zhenlang Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaoqin Fu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
30
|
Manka SW, Moores CA. Pseudo-repeats in doublecortin make distinct mechanistic contributions to microtubule regulation. EMBO Rep 2020; 21:e51534. [PMID: 33051979 PMCID: PMC7726794 DOI: 10.15252/embr.202051534] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 11/16/2022] Open
Abstract
Doublecortin (DCX) is a neuronal microtubule-associated protein (MAP) indispensable for brain development. Its flexibly linked doublecortin (DC) domains-NDC and CDC-mediate microtubule (MT) nucleation and stabilization, but it is unclear how. Using high-resolution time-resolved cryo-EM, we mapped NDC and CDC interactions with tubulin at different MT polymerization stages and studied their functional effects on MT dynamics using TIRF microscopy. Although coupled, each DC repeat within DCX appears to have a distinct role in MT nucleation and stabilization: CDC is a conformationally plastic module that appears to facilitate MT nucleation and stabilize tubulin-tubulin contacts in the nascent MT lattice, while NDC appears to be favored along the mature lattice, providing MT stabilization. Our structures of MT-bound DC domains also explain in unprecedented detail the DCX mutation-related brain defects observed in the clinic. This modular composition of DCX reflects a common design principle among MAPs where pseudo-repeats of tubulin/MT binding elements chaperone or stabilize distinct conformational transitions to regulate distinct stages of MT dynamic instability.
Collapse
Affiliation(s)
- Szymon W Manka
- Institute of Structural and Molecular BiologyDepartment of Biological Sciences, BirkbeckUniversity of LondonLondonUK
| | - Carolyn A Moores
- Institute of Structural and Molecular BiologyDepartment of Biological Sciences, BirkbeckUniversity of LondonLondonUK
| |
Collapse
|
31
|
Knossow M, Campanacci V, Khodja LA, Gigant B. The Mechanism of Tubulin Assembly into Microtubules: Insights from Structural Studies. iScience 2020; 23:101511. [PMID: 32920486 PMCID: PMC7491153 DOI: 10.1016/j.isci.2020.101511] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022] Open
Abstract
Microtubules are cytoskeletal components involved in pivotal eukaryotic functions such as cell division, ciliogenesis, and intracellular trafficking. They assemble from αβ-tubulin heterodimers and disassemble in a process called dynamic instability, which is driven by GTP hydrolysis. Structures of the microtubule and of soluble tubulin have been determined by cryo-EM and by X-ray crystallography, respectively. Altogether, these data define the mechanism of tubulin assembly-disassembly at atomic or near-atomic level. We review here the structural changes that occur during assembly, tubulin switching from a curved conformation in solution to a straight one in the microtubule core. We also present more subtle changes associated with GTP binding, leading to tubulin activation for assembly. Finally, we show how cryo-EM and X-ray crystallography are complementary methods to characterize the interaction of tubulin with proteins involved either in intracellular transport or in microtubule dynamics regulation.
Collapse
Affiliation(s)
- Marcel Knossow
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Valérie Campanacci
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Liza Ammar Khodja
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Benoît Gigant
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| |
Collapse
|
32
|
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: 0.4] [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
|
33
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
34
|
Monroy BY, Tan TC, Oclaman JM, Han JS, Simó S, Niwa S, Nowakowski DW, McKenney RJ, Ori-McKenney KM. A Combinatorial MAP Code Dictates Polarized Microtubule Transport. Dev Cell 2020; 53:60-72.e4. [PMID: 32109385 DOI: 10.1016/j.devcel.2020.01.029] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/19/2019] [Accepted: 01/27/2020] [Indexed: 01/14/2023]
Abstract
Many eukaryotic cells distribute their intracellular components asymmetrically through regulated active transport driven by molecular motors along microtubule tracks. While intrinsic and extrinsic regulation of motor activity exists, what governs the overall distribution of activated motor-cargo complexes within cells remains unclear. Here, we utilize in vitro reconstitution of purified motor proteins and non-enzymatic microtubule-associated proteins (MAPs) to demonstrate that MAPs exhibit distinct influences on the motility of the three main classes of transport motors: kinesin-1, kinesin-3, and cytoplasmic dynein. Further, we dissect how combinations of MAPs affect motors and unveil MAP9 as a positive modulator of kinesin-3 motility. From these data, we propose a general "MAP code" that has the capacity to strongly bias directed movement along microtubules and helps elucidate the intricate intracellular sorting observed in highly polarized cells such as neurons.
Collapse
Affiliation(s)
- Brigette Y Monroy
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Tracy C Tan
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Janah May Oclaman
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Jisoo S Han
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| | | | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
35
|
Martineau FS, Sahu S, Plantier V, Buhler E, Schaller F, Fournier L, Chazal G, Kawasaki H, Represa A, Watrin F, Manent JB. Correct Laminar Positioning in the Neocortex Influences Proper Dendritic and Synaptic Development. Cereb Cortex 2019; 28:2976-2990. [PMID: 29788228 PMCID: PMC6041803 DOI: 10.1093/cercor/bhy113] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Indexed: 01/28/2023] Open
Abstract
The neocortex is a 6-layered laminated structure with a precise anatomical and functional organization ensuring proper function. Laminar positioning of cortical neurons, as determined by termination of neuronal migration, is a key determinant of their ability to assemble into functional circuits. However, the exact contribution of laminar placement to dendrite morphogenesis and synapse formation remains unclear. Here we manipulated the laminar position of cortical neurons by knocking down doublecortin (Dcx), a crucial effector of migration, and show that misplaced neurons fail to properly form dendrites, spines, and functional glutamatergic and GABAergic synapses. We further show that knocking down Dcx in properly positioned neurons induces similar but milder defects, suggesting that the laminar misplacement is the primary cause of altered neuronal development. Thus, the specific laminar environment of their fated layers is crucial for the maturation of cortical neurons, and influences their functional integration into developing cortical circuits.
Collapse
Affiliation(s)
| | - Surajit Sahu
- INMED, Aix-Marseille University, INSERM U901, Marseille, France
| | | | | | | | | | | | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Alfonso Represa
- INMED, Aix-Marseille University, INSERM U901, Marseille, France
| | | | | |
Collapse
|
36
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
37
|
Lawrence EJ, Zanic M. Rescuing microtubules from the brink of catastrophe: CLASPs lead the way. Curr Opin Cell Biol 2019; 56:94-101. [PMID: 30453184 PMCID: PMC6370552 DOI: 10.1016/j.ceb.2018.10.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/12/2018] [Accepted: 10/31/2018] [Indexed: 01/11/2023]
Abstract
Microtubules are cytoskeletal polymers that dynamically remodel to perform essential cellular functions. Individual microtubules alternate between phases of growth and shrinkage via sudden transitions called catastrophe and rescue, driven by losing and regaining a stabilizing cap at the dynamic microtubule end. New in vitro studies now show that a conserved family of CLASP proteins specifically modulate microtubule catastrophe and rescue transitions. Further, recent cryo-electron microscopy approaches have elucidated new structural features of the stabilizing cap. Together, these new advances provide a clearer view on the complexity of the microtubule end and its regulation.
Collapse
Affiliation(s)
- E J Lawrence
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, United States
| | - M Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, United States; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37240, United States; Department of Biochemistry, Vanderbilt University, Nashville, TN 37240, United States.
| |
Collapse
|
38
|
Manka SW, Moores CA. Microtubule structure by cryo-EM: snapshots of dynamic instability. Essays Biochem 2018; 62:737-751. [PMID: 30315096 PMCID: PMC6281474 DOI: 10.1042/ebc20180031] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 01/24/2023]
Abstract
The development of cryo-electron microscopy (cryo-EM) allowed microtubules to be captured in their solution-like state, enabling decades of insight into their dynamic mechanisms and interactions with binding partners. Cryo-EM micrographs provide 2D visualization of microtubules, and these 2D images can also be used to reconstruct the 3D structure of the polymer and any associated binding partners. In this way, the binding sites for numerous components of the microtubule cytoskeleton-including motor domains from many kinesin motors, and the microtubule-binding domains of dynein motors and an expanding collection of microtubule associated proteins-have been determined. The effects of various microtubule-binding drugs have also been studied. High-resolution cryo-EM structures have also been used to probe the molecular basis of microtubule dynamic instability, driven by the GTPase activity of β-tubulin. These studies have shown the conformational changes in lattice-confined tubulin dimers in response to steps in the tubulin GTPase cycle, most notably lattice compaction at the longitudinal inter-dimer interface. Although work is ongoing to define a complete structural model of dynamic instability, attention has focused on the role of gradual destabilization of lateral contacts between tubulin protofilaments, particularly at the microtubule seam. Furthermore, lower resolution cryo-electron tomography 3D structures are shedding light on the heterogeneity of microtubule ends and how their 3D organization contributes to dynamic instability. The snapshots of these polymers captured using cryo-EM will continue to provide critical insights into their dynamics, interactions with cellular components, and the way microtubules contribute to cellular functions in diverse physiological contexts.
Collapse
Affiliation(s)
- Szymon W Manka
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, U.K.
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, U.K
| |
Collapse
|
39
|
Abstract
Microtubules are dynamic polymers of αβ-tubulin that are essential for intracellular organization, organelle trafficking and chromosome segregation. Microtubule growth and shrinkage occur via addition and loss of αβ-tubulin subunits, which are biochemical processes. Dynamic microtubules can also engage in mechanical processes, such as exerting forces by pushing or pulling against a load. Recent advances at the intersection of biochemistry and mechanics have revealed the existence of multiple conformations of αβ-tubulin subunits and their central role in dictating the mechanisms of microtubule dynamics and force generation. It has become apparent that microtubule-associated proteins (MAPs) selectively target specific tubulin conformations to regulate microtubule dynamics, and mechanical forces can also influence microtubule dynamics by altering the balance of tubulin conformations. Importantly, the conformational states of tubulin dimers are likely to be coupled throughout the lattice: the conformation of one dimer can influence the conformation of its nearest neighbours, and this effect can propagate over longer distances. This coupling provides a long-range mechanism by which MAPs and forces can modulate microtubule growth and shrinkage. These findings provide evidence that the interplay between biochemistry and mechanics is essential for the cellular functions of microtubules.
Collapse
Affiliation(s)
- Gary J Brouhard
- Department of Biology, McGill University, Montréal, Quebec, Canada.
| | - Luke M Rice
- Department of Biophysics, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
40
|
Shahsavani M, Pronk RJ, Falk R, Lam M, Moslem M, Linker SB, Salma J, Day K, Schuster J, Anderlid BM, Dahl N, Gage FH, Falk A. An in vitro model of lissencephaly: expanding the role of DCX during neurogenesis. Mol Psychiatry 2018; 23:1674-1684. [PMID: 28924182 DOI: 10.1038/mp.2017.175] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/09/2017] [Accepted: 07/12/2017] [Indexed: 12/22/2022]
Abstract
Lissencephaly comprises a spectrum of brain malformations due to impaired neuronal migration in the developing cerebral cortex. Classical lissencephaly is characterized by smooth cerebral surface and cortical thickening that result in seizures, severe neurological impairment and developmental delay. Mutations in the X-chromosomal gene DCX, encoding doublecortin, is the main cause of classical lissencephaly. Much of our knowledge about DCX-associated lissencephaly comes from post-mortem analyses of patient's brains, mainly since animal models with DCX mutations do not mimic the disease. In the absence of relevant animal models and patient brain specimens, we took advantage of induced pluripotent stem cell (iPSC) technology to model the disease. We established human iPSCs from two males with mutated DCX and classical lissencephaly including smooth brain and abnormal cortical morphology. The disease was recapitulated by differentiation of iPSC into neural cells followed by expression profiling and dissection of DCX-associated functions. Here we show that neural stem cells, with absent or reduced DCX protein expression, exhibit impaired migration, delayed differentiation and deficient neurite formation. Hence, the patient-derived iPSCs and neural stem cells provide a system to further unravel the functions of DCX in normal development and disease.
Collapse
Affiliation(s)
- M Shahsavani
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - R J Pronk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - R Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - M Lam
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - M Moslem
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - S B Linker
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - J Salma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - K Day
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J Schuster
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - B-M Anderlid
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - N Dahl
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - F H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - A Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Rufer AC, Kusznir E, Burger D, Stihle M, Ruf A, Rudolph MG. Domain swap in the C-terminal ubiquitin-like domain of human doublecortin. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2018; 74:450-462. [PMID: 29717716 DOI: 10.1107/s2059798318004813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/23/2018] [Indexed: 11/10/2022]
Abstract
Doublecortin, a microtubule-associated protein that is only produced during neurogenesis, cooperatively binds to microtubules and stimulates microtubule polymerization and cross-linking by unknown mechanisms. A domain swap is observed in the crystal structure of the C-terminal domain of doublecortin. As determined by analytical ultracentrifugation, an open conformation is also present in solution. At higher concentrations, higher-order oligomers of the domain are formed. The domain swap and additional interfaces observed in the crystal lattice can explain the formation of doublecortin tetramers or multimers, in line with the analytical ultracentrifugation data. Taken together, the domain swap offers a mechanism for the observed cooperative binding of doublecortin to microtubules. Doublecortin-induced cross-linking of microtubules can be explained by the same mechanism. The effect of several mutations leading to lissencephaly and double-cortex syndrome can be traced to the domain swap and the proposed self-association of doublecortin.
Collapse
Affiliation(s)
- Arne C Rufer
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Eric Kusznir
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Dominique Burger
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Martine Stihle
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Armin Ruf
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Markus G Rudolph
- pRED, Therapeutic Modalities, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| |
Collapse
|
42
|
Márquez-Valadez B, Valle-Bautista R, García-López G, Díaz NF, Molina-Hernández A. Maternal Diabetes and Fetal Programming Toward Neurological Diseases: Beyond Neural Tube Defects. Front Endocrinol (Lausanne) 2018; 9:664. [PMID: 30483218 PMCID: PMC6243582 DOI: 10.3389/fendo.2018.00664] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022] Open
Abstract
The purpose of this review was to search for experimental or clinical evidence on the effect of hyperglycemia in fetal programming to neurological diseases, excluding evident neural tube defects. The lack of timely diagnosis and the inadequate control of diabetes during pregnancy have been related with postnatal obesity, low intellectual and verbal coefficients, language and motor deficits, attention deficit with hyperactivity, problems in psychosocial development, and an increased predisposition to autism and schizophrenia. It has been proposed that several childhood or adulthood diseases have their origin during fetal development through a phenomenon called fetal programming. However, not all the relationships between the outcomes mentioned above and diabetes during gestation are clear, well-studied, or have been related to fetal programming. To understand this relationship, it is imperative to understand how developmental processes take place in health, in order to understand how the functional cytoarchitecture of the central nervous system takes place; to identify changes prompted by hyperglycemia, and to correlate them with the above postnatal impaired functions. Although changes in the establishment of patterns during central nervous system fetal development are related to a wide variety of neurological pathologies, the mechanism by which several maternal conditions promote fetal alterations that contribute to impaired neural development with postnatal consequences are not clear. Animal models have been extremely useful in studying the effect of maternal pathologies on embryo and fetal development, since obtaining central nervous system tissue in humans with normal appearance during fetal development is an important limitation. This review explores the state of the art on this topic, to help establish the way forward in the study of fetal programming under hyperglycemia and its impact on neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Berenice Márquez-Valadez
- Department of Physiology and Cell Development, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rocío Valle-Bautista
- Department of Physiology and Cell Development, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Guadalupe García-López
- Department of Physiology and Cell Development, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Néstor Fabián Díaz
- Department of Physiology and Cell Development, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Anayansi Molina-Hernández
- Department of Physiology and Cell Development, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
- *Correspondence: Anayansi Molina-Hernández
| |
Collapse
|
43
|
Ramkumar A, Jong BY, Ori-McKenney KM. ReMAPping the microtubule landscape: How phosphorylation dictates the activities of microtubule-associated proteins. Dev Dyn 2017; 247:138-155. [PMID: 28980356 DOI: 10.1002/dvdy.24599] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Classical microtubule-associated proteins (MAPs) were originally identified based on their co-purification with microtubules assembled from mammalian brain lysate. They have since been found to perform a range of functions involved in regulating the dynamics of the microtubule cytoskeleton. Most of these MAPs play integral roles in microtubule organization during neuronal development, microtubule remodeling during neuronal activity, and microtubule stabilization during neuronal maintenance. As a result, mutations in MAPs contribute to neurodevelopmental disorders, psychiatric conditions, and neurodegenerative diseases. MAPs are post-translationally regulated by phosphorylation depending on developmental time point and cellular context. Phosphorylation can affect the microtubule affinity, cellular localization, or overall function of a particular MAP and can thus have profound implications for neuronal health. Here we review MAP1, MAP2, MAP4, MAP6, MAP7, MAP9, tau, and DCX, and how each is regulated by phosphorylation in neuronal physiology and disease. Developmental Dynamics 247:138-155, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amrita Ramkumar
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | - Brigette Y Jong
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | | |
Collapse
|
44
|
Ayanlaja AA, Xiong Y, Gao Y, Ji G, Tang C, Abdikani Abdullah Z, Gao D. Distinct Features of Doublecortin as a Marker of Neuronal Migration and Its Implications in Cancer Cell Mobility. Front Mol Neurosci 2017; 10:199. [PMID: 28701917 PMCID: PMC5487455 DOI: 10.3389/fnmol.2017.00199] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Neuronal migration is a critical process in the development of the nervous system. Defects in the migration of the neurons are associated with diseases like lissencephaly, subcortical band heterotopia (SBH), and pachygyria. Doublecortin (DCX) is an essential factor in neurogenesis and mutations in this protein impairs neuronal migration leading to several pathological conditions. Although, DCX is capable of modulating and stabilizing microtubules (MTs) to ensure effective migration, the mechanisms involved in executing these functions remain poorly understood. Meanwhile, there are existing gaps regarding the processes that underlie tumor initiation and progression into cancer as well as the ability to migrate and invade normal cells. Several studies suggest that DCX is involved in cancer metastasis. Unstable interactions between DCX and MTs destabilizes cytoskeletal organization leading to disorganized movements of cells, a process which may be implicated in the uncontrolled migration of cancer cells. However, the underlying mechanism is complex and require further clarification. Therefore, exploring the importance and features known up to date about this molecule will broaden our understanding and shed light on potential therapeutic approaches for the associated neurological diseases. This review summarizes current knowledge about DCX, its features, functions, and relationships with other proteins. We also present an overview of its role in cancer cells and highlight the importance of studying its gene mutations.
Collapse
Affiliation(s)
- Abiola A Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Ye Xiong
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Yue Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - GuangQuan Ji
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Zamzam Abdikani Abdullah
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - DianShuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| |
Collapse
|
45
|
Yang C, Wang L, Xing X, Gao Y, Guo L. Seasonal variation in telencephalon cell proliferation in adult female tsinling dwarf skinks (Scincella tsinlingensis). Brain Res 2017; 1662:7-15. [PMID: 28237546 DOI: 10.1016/j.brainres.2017.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/08/2017] [Accepted: 02/14/2017] [Indexed: 11/25/2022]
Abstract
In adult mammals, neurogenesis is limited to specific niches in the brain, but considerable adult neurogenesis occurs in many brain regions in non-mammalian vertebrates. Non-mammalian vertebrates provide invaluable comparative material for understanding the core mechanisms of adult neural stem cell maintenance and fate, but phylogenetic differences in adult neurogenesis remain poorly understood. Here we examine cell proliferation seasonality in the telencephalon of adult female tsinling dwarf skinks (Scincella tsinlingensis) by injecting wild animals caught in summer, autumn and spring, and animals caught in autumn and raised under winter conditions, with 5-Bromo-2'-deoxyuridine (BrdU). Then, 24h, 7d and 28d after BrdU administration we examined brain tissue and quantified BrdU-labeled cells as a marker of neuronal proliferation. The highest number of labeled cells in the telencephalon was found in the 7d group. BrdU-positive cells were widely distributed in the anterior olfactory nucleus (AON), medial cortex (MC), dorsal cortex (DC), lateral cortex (LC), dorsal ventricular ridge (DVR), septum (SP), striatum (STR) and nucleus sphericus (NS). No BrdU-positive cells were detected in olfactory bulbs or elsewhere in the telencephalon. The highest proliferative levels were found in the AON in autumn. The NS exhibited relatively high levels of cell proliferation. The proliferative rate in the AON fluctuated seasonally as autumn>summer>spring>winter. Glial fibrillary acidic protein-positive cells were widely distributed in the telencephalon and their fibrous processes extended into brain parenchyma and anchored in the meninges. Doublecortin-positive newborn neurons of the subventricular zone appeared to migrate into the cerebral cortex via the radial migratory stream. Cell proliferation in the telencephalon of adult female S. tsinlingensis fluctuates seasonally, especially in regions related to olfactory memory. This is the first demonstration of proliferative activity in the telencephalon of a skink.
Collapse
Affiliation(s)
- Chun Yang
- School of Life Sciences, Shanxi Normal University, Linfen, Shanxi 041000, China.
| | - Limin Wang
- School of Life Sciences, Shanxi Normal University, Linfen, Shanxi 041000, China
| | - Xiangyang Xing
- School of Life Sciences, Shanxi Normal University, Linfen, Shanxi 041000, China
| | - Yanyan Gao
- School of Life Sciences, Shanxi Normal University, Linfen, Shanxi 041000, China
| | - Li Guo
- School of Life Sciences, Shanxi Normal University, Linfen, Shanxi 041000, China
| |
Collapse
|
46
|
Bowne-Anderson H, Hibbel A, Howard J. Regulation of Microtubule Growth and Catastrophe: Unifying Theory and Experiment. Trends Cell Biol 2016; 25:769-779. [PMID: 26616192 DOI: 10.1016/j.tcb.2015.08.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/27/2015] [Accepted: 08/27/2015] [Indexed: 10/22/2022]
Abstract
Recent studies have found that microtubule-associated proteins (MAPs) can regulate the dynamical properties of microtubules in unexpected ways. For most MAPs, there is an inverse relationship between their effects on the speed of growth and the frequency of catastrophe, the conversion of a growing microtubule to a shrinking one. Such a negative correlation is predicted by the standard GTP-cap model, which posits that catastrophe is due to loss of a stabilizing cap of GTP-tubulin at the end of a growing microtubule. However, many other MAPs, notably Kinesin-4 and combinations of EB1 with XMAP215, contradict this general rule. In this review, we show that a more nuanced, but still simple, GTP-cap model, can account for the diverse regulatory activities of MAPs.
Collapse
Affiliation(s)
| | - Anneke Hibbel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany; ETH Zurich, Institute for Biochemistry, HPM E8.1, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | | |
Collapse
|
47
|
Brouhard GJ. Dynamic instability 30 years later: complexities in microtubule growth and catastrophe. Mol Biol Cell 2016; 26:1207-10. [PMID: 25823928 PMCID: PMC4454169 DOI: 10.1091/mbc.e13-10-0594] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Microtubules are not like other polymers. Whereas polymers such as F-actin will grow continuously as long as the subunit concentration is high enough, a steadily growing microtubule can suddenly shrink even when there is ample αβ-tubulin around. This remarkable behavior was discovered in 1984 when Tim Mitchison and Marc Kirschner deduced that microtubules switch from growth to shrinkage when they lose their GTP caps. Here, I review the canonical explanation of dynamic instability that was fleshed out in the years after its discovery. Many aspects of this explanation have been recently subverted, particularly those related to how GTP-tubulin forms polymers and why GTP hydrolysis disrupts them. I describe these developments and speculate on how our explanation of dynamic instability can be changed to accommodate them.
Collapse
Affiliation(s)
- Gary J Brouhard
- Department of Biology, McGill University, Montréal, QC H3A 1B1, Canada
| |
Collapse
|
48
|
|
49
|
Ching AS, Ahmad-Annuar A. A Perspective on the Role of microRNA-128 Regulation in Mental and Behavioral Disorders. Front Cell Neurosci 2015; 9:465. [PMID: 26696825 PMCID: PMC4677093 DOI: 10.3389/fncel.2015.00465] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/16/2015] [Indexed: 12/18/2022] Open
Abstract
MiRNAs are short, non-coding RNA molecules that regulate gene expression post-transcriptionally. Over the past decade, misregulated miRNA pathways have been associated with various diseases such as cancer, neurodegenerative diseases, and neurodevelopmental disorders. In this article, we aim to discuss the role played by miR-128 in neuropsychiatric disorders, and highlight potential target genes from an in silico analysis of predicted miR-128 targets. We also discuss the differences of target gene determination based on a bioinformatics or empirical approach. Using data from TargetScan and published reports, we narrowed the miR-128 target gene list to those that are known to be associated with neuropsychiatric disorders, and found that these genes can be classified into 29 gene clusters and are mostly enriched in cancer and MAPK signaling pathways. We also highlight some recent studies on several of the miR-128 targets which should be investigated further as potential candidate genes for therapeutic interventions.
Collapse
Affiliation(s)
- Ai-Sze Ching
- Department of Biomedical Science, Faculty of Medicine, University of Malaya Kuala Lumpur, Malaysia
| | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya Kuala Lumpur, Malaysia
| |
Collapse
|
50
|
Chen L, Chuang M, Koorman T, Boxem M, Jin Y, Chisholm AD. Axon injury triggers EFA-6 mediated destabilization of axonal microtubules via TACC and doublecortin like kinase. eLife 2015; 4. [PMID: 26339988 PMCID: PMC4596636 DOI: 10.7554/elife.08695] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023] Open
Abstract
Axon injury triggers a series of changes in the axonal cytoskeleton that are prerequisites for effective axon regeneration. In Caenorhabditis elegans the signaling protein Exchange Factor for ARF-6 (EFA-6) is a potent intrinsic inhibitor of axon regrowth. Here we show that axon injury triggers rapid EFA-6-dependent inhibition of axonal microtubule (MT) dynamics, concomitant with relocalization of EFA-6. EFA-6 relocalization and axon regrowth inhibition require a conserved 18-aa motif in its otherwise intrinsically disordered N-terminal domain. The EFA-6 N-terminus binds the MT-associated proteins TAC-1/Transforming-Acidic-Coiled-Coil, and ZYG-8/Doublecortin-Like-Kinase, both of which are required for regenerative growth cone formation, and which act downstream of EFA-6. After injury TAC-1 and EFA-6 transiently relocalize to sites marked by the MT minus end binding protein PTRN-1/Patronin. We propose that EFA-6 acts as a bifunctional injury-responsive regulator of axonal MT dynamics, acting at the cell cortex in the steady state and at MT minus ends after injury. DOI:http://dx.doi.org/10.7554/eLife.08695.001 In the nervous system, cells called neurons carry information around the body. These cells have long thin projections called axons that allow the information to pass very quickly along the cell to junctions with other neurons. Neurons in adult mammals are limited in their ability to regenerate, so any damage to axons, for example, due to a stroke or a brain injury, tends to be permanent. Therefore, an important goal in neuroscience research is to discover the genes and proteins that are involved in regenerating axons as this may make it possible to develop new therapies. An internal scaffold called the cytoskeleton supports the three-dimensional shape of the axons. Changes in the cytoskeleton are required to allow neurons to regenerate axons after injury, and drugs that stabilize filaments called microtubules in the cytoskeleton can promote these changes. Chen et al. used a technique called laser microsurgery to sever individual axons in a roundworm known as C. elegans and then observed whether these axons could regenerate. The experiments reveal that a protein called EFA-6 blocks the regeneration of neurons by preventing rearrangements in the cytoskeleton. EFA-6 is normally found at the membrane that surrounds the neuron. However, Chen et al. show that when the axon is damaged, this protein rapidly moves to areas near the ends of microtubule filaments. EFA-6 interacts with two other proteins that are associated with microtubules and are required for axons to be able to regenerate. Chen et al.'s findings demonstrate that several proteins that regulate microtubule filaments play a key role in regenerating axons. All three of these proteins are found in humans and other animals so they have the potential to be targeted by drug therapies in future. The next challenge is to understand the details of how EFA-6 activity is affected by axon injury, and how this alters the cytoskeleton. DOI:http://dx.doi.org/10.7554/eLife.08695.002
Collapse
Affiliation(s)
- Lizhen Chen
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, United States.,University of California, San Diego, La Jolla, United States
| | - Marian Chuang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Thijs Koorman
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Mike Boxem
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, United States.,University of California, San Diego, La Jolla, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, United States
| | - Andrew D Chisholm
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|