1
|
Momotyuk E, Ebrahim N, Shakirova K, Dashinimaev E. Role of the cytoskeleton in cellular reprogramming: effects of biophysical and biochemical factors. Front Mol Biosci 2025; 12:1538806. [PMID: 40123979 PMCID: PMC11926148 DOI: 10.3389/fmolb.2025.1538806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
The cytoskeleton plays a crucial role in regulating cellular behavior, acting as both a structural framework and a mediator of mechanical and biochemical signals that influence cell fate. In the context of cellular reprogramming, modifications to the cytoskeleton can have profound effects on lineage commitment and differentiation efficiency. This review explores the impact of mechanical forces such as substrate stiffness, topography, extracellular fluid viscosity, and cell seeding density on cytoskeletal organization and mechanotransduction pathways, including Rho/ROCK and YAP/TAZ signaling. Additionally, we examine the influence of biochemical agents that modulate cytoskeletal dynamics, such as actin and microtubule polymerization inhibitors, and their effects on stem cell differentiation. By understanding how cytoskeletal remodeling governs cellular identity, this review highlights potential strategies for improving reprogramming efficiency and directing cell fate by manipulating mechanical and biochemical cues.
Collapse
Affiliation(s)
| | | | | | - Erdem Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
2
|
Schmied C, Ebner M, Samsó P, Van Der Veen R, Haucke V, Lehmann M. OrgaMapper: a robust and easy-to-use workflow for analyzing organelle positioning. BMC Biol 2024; 22:220. [PMID: 39343900 PMCID: PMC11440938 DOI: 10.1186/s12915-024-02015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Eukaryotic cells are highly compartmentalized by a variety of organelles that carry out specific cellular processes. The position of these organelles within the cell is elaborately regulated and vital for their function. For instance, the position of lysosomes relative to the nucleus controls their degradative capacity and is altered in pathophysiological conditions. The molecular components orchestrating the precise localization of organelles remain incompletely understood. A confounding factor in these studies is the fact that organelle positioning is surprisingly non-trivial to address e.g., perturbations that affect the localization of organelles often lead to secondary phenotypes such as changes in cell or organelle size. These phenotypes could potentially mask effects or lead to the identification of false positive hits. To uncover and test potential molecular components at scale, accurate and easy-to-use analysis tools are required that allow robust measurements of organelle positioning. RESULTS Here, we present an analysis workflow for the faithful, robust, and quantitative analysis of organelle positioning phenotypes. Our workflow consists of an easy-to-use Fiji plugin and an R Shiny App. These tools enable users without background in image or data analysis to (1) segment single cells and nuclei and to detect organelles, (2) to measure cell size and the distance between detected organelles and the nucleus, (3) to measure intensities in the organelle channel plus one additional channel, (4) to measure radial intensity profiles of organellar markers, and (5) to plot the results in informative graphs. Using simulated data and immunofluorescent images of cells in which the function of known factors for lysosome positioning has been perturbed, we show that the workflow is robust against common problems for the accurate assessment of organelle positioning such as changes of cell shape and size, organelle size and background. CONCLUSIONS OrgaMapper is a versatile, robust, and easy-to-use automated image analysis workflow that can be utilized in microscopy-based hypothesis testing and screens. It effectively allows for the mapping of the intracellular space and enables the discovery of novel regulators of organelle positioning.
Collapse
Affiliation(s)
- Christopher Schmied
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany.
- Present address: EU-OPENSCREEN ERIC, Robert-Roessle-Straße 10, Berlin, 13125, Germany.
| | - Michael Ebner
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany
| | - Paula Samsó
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany
| | - Rozemarijn Van Der Veen
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany
- Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, 14195, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut Für Molekulare Pharmakologie (FMP), Robert-Roessle-Straße 10, Berlin, 13125, Germany
| |
Collapse
|
3
|
Banerjee S, Zhao Q, Wang B, Qin J, Yuan X, Lou Z, Zheng W, Li H, Wang X, Cheng X, Zhu Y, Lin F, Yang F, Xu J, Munshi A, Das P, Zhou Y, Mandal K, Wang Y, Ayub M, Hirokawa N, Xi Y, Chen G, Li C. A novel in-frame deletion in KIF5C gene causes infantile onset epilepsy and psychomotor retardation. MedComm (Beijing) 2024; 5:e469. [PMID: 38525108 PMCID: PMC10960728 DOI: 10.1002/mco2.469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 03/26/2024] Open
Abstract
Motor proteins, encoded by Kinesin superfamily (KIF) genes, are critical for brain development and plasticity. Increasing studies reported KIF's roles in neurodevelopmental disorders. Here, a 6 years and 3 months-old Chinese boy with markedly symptomatic epilepsy, intellectual disability, brain atrophy, and psychomotor retardation was investigated. His parents and younger sister were phenotypically normal and had no disease-related family history. Whole exome sequencing identified a novel heterozygous in-frame deletion (c.265_267delTCA) in exon 3 of the KIF5C in the proband, resulting in the removal of evolutionarily highly conserved p.Ser90, located in its ATP-binding domain. Sanger sequencing excluded the proband's parents and family members from harboring this variant. The activity of ATP hydrolysis in vitro was significantly reduced as predicted. Immunofluorescence studies showed wild-type KIF5C was widely distributed throughout the cytoplasm, while mutant KIF5C was colocalized with microtubules. The live-cell imaging of the cargo-trafficking assay revealed that mutant KIF5C lost the peroxisome-transporting ability. Drosophila models also confirmed p.Ser90del's essential role in nervous system development. This study emphasized the importance of the KIF5C gene in intracellular cargo-transport as well as germline variants that lead to neurodevelopmental disorders and might enable clinicians for timely and accurate diagnosis and disease management in the future.
Collapse
Affiliation(s)
- Santasree Banerjee
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
- Department of GeneticsCollege of Basic Medical SciencesJilin UniversityChangchunChina
- Department of GeneticsUniversity of DelhiNew DelhiIndia
| | - Qiang Zhao
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Bo Wang
- Department of PediatricsShenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jiale Qin
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Xin Yuan
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Ziwei Lou
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Weizeng Zheng
- Department of RadiologyWomen's HospitalZhejiang University School of MedicineHangzhouChina
| | - Huanguo Li
- Department of RadiologyHangzhou Hospital of Traditional Chinese MedicineHangzhouChina
| | - Xiaojun Wang
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Xiawei Cheng
- School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Yu Zhu
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Fan Lin
- Department of Cell BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Fan Yang
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Junyu Xu
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Anjana Munshi
- Department of Human Genetics and Molecular MedicineCentral University of PunjabBathindaIndia
| | - Parimal Das
- Centre for Genetic DisordersBanaras Hindu UniversityVaranasiIndia
| | - Yuanfeng Zhou
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Kausik Mandal
- Department of Medical GeneticsSanjay Gandhi Postgraduate Institute of Medical SciencesLucknowUttar PradeshIndia
| | - Yi Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Muhammad Ayub
- Department of PsychiatryUniversity College LondonLondonUK
| | - Nobutaka Hirokawa
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Yongmei Xi
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Guangfu Chen
- Department of PediatricsShenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Chen Li
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
- Alibaba‐Zhejiang University Joint Research Center of Future Digital HealthcareHangzhouChina
| |
Collapse
|
4
|
Gümüşderelioğlu S, Resch L, Brock T, Luxton GWG, Cope H, Tan QKG, Hopkins C, Starr DA. A humanized Caenorhabditis elegans model of hereditary spastic paraplegia-associated variants in KLC4. Dis Model Mech 2023; 16:dmm050076. [PMID: 37565267 PMCID: PMC10481945 DOI: 10.1242/dmm.050076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a group of degenerative neurological disorders. We identified a variant in human kinesin light chain 4 (KLC4) that is suspected to be associated with autosomal-dominant HSP. How this and other variants relate to pathologies is unknown. We created a humanized Caenorhabditis elegans model in which klc-2 was replaced by human KLC4 (referred to as hKLC4) and assessed the extent to which hKLC4 retained function in the worm. We observed a slight decrease in motility but no nuclear migration defects in the humanized worms, suggesting that hKLC4 retains much of the function of klc-2. Five hKLC4 variants were introduced into the humanized model. The clinical variant led to early lethality, with significant defects in nuclear migration when homozygous and a weak nuclear migration defect when heterozygous, possibly correlating with the clinical finding of late-onset HSP when the proband was heterozygous. Thus, we were able to establish humanized C. elegans as an animal model for HSP and to use it to test the significance of five variants of uncertain significance in the human gene KLC4.
Collapse
Affiliation(s)
- Selin Gümüşderelioğlu
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | - G. W. Gant Luxton
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Heidi Cope
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Queenie K.-G. Tan
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Daniel A. Starr
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
5
|
Gümüşderelioğlu S, Resch L, Brock T, Luxton GWG, Tan QKG, Hopkins C, Starr DA. A humanized Caenorhabditis elegans model of Hereditary Spastic Paraplegia-associated variants in kinesin light chain KLC4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523106. [PMID: 36789438 PMCID: PMC9928042 DOI: 10.1101/2023.01.07.523106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hereditary spastic paraplegia (HSP) is a group of degenerative neurological disorders. We identified a variant in human kinesin light chain KLC4 that is suspected to be associated with autosomal dominant HSP. How this and other variants relate to pathologies is unknown. We created a humanized C. elegans model where klc- 2 was replaced with human KLC4 and assessed the extent to which hKLC4 retained function in the worm. We observed a slight decrease in motility but no nuclear migration defects in the humanized worms, suggesting that hKLC4 retains much of the function of klc-2 . Five hKLC4 variants were introduced into the humanized model. The clinical variant led to early lethality with significant defects in nuclear migration when homozygous, and a weak nuclear migration defect when heterozygous, possibly correlating with the clinical finding of late onset HSP when the proband was heterozygous. Thus, we were able to establish humanized C. elegans as an animal model for HSP and use it to test the significance of five variants of uncertain significance in the human gene KLC4 . Summary Statement We identified a variant in KLC4 associated with Hereditary Spastic Paraplegia. The variant had physiological relevance in a humanized C. elegans model where we replaced klc-2 with human KLC4 .
Collapse
Affiliation(s)
- Selin Gümüşderelioğlu
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | | | | | - G W Gant Luxton
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Queenie K-G Tan
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | - Daniel A Starr
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| |
Collapse
|
6
|
Guo W, Ale TA, Sun S, Sanchez JE, Li L. A Comprehensive Study on the Electrostatic Properties of Tubulin-Tubulin Complexes in Microtubules. Cells 2023; 12:238. [PMID: 36672172 PMCID: PMC9857020 DOI: 10.3390/cells12020238] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023] Open
Abstract
Microtubules are key players in several stages of the cell cycle and are also involved in the transportation of cellular organelles. Microtubules are polymerized by α/β tubulin dimers with a highly dynamic feature, especially at the plus ends of the microtubules. Therefore, understanding the interactions among tubulins is crucial for characterizing microtubule dynamics. Studying microtubule dynamics can help researchers make advances in the treatment of neurodegenerative diseases and cancer. In this study, we utilize a series of computational approaches to study the electrostatic interactions at the binding interfaces of tubulin monomers. Our study revealed that among all the four types of tubulin-tubulin binding modes, the electrostatic attractive interactions in the α/β tubulin binding are the strongest while the interactions of α/α tubulin binding in the longitudinal direction are the weakest. Our calculations explained that due to the electrostatic interactions, the tubulins always preferred to form α/β tubulin dimers. The interactions between two protofilaments are the weakest. Thus, the protofilaments are easily separated from each other. Furthermore, the important residues involved in the salt bridges at the binding interfaces of the tubulins are identified, which illustrates the details of the interactions in the microtubule. This study elucidates some mechanistic details of microtubule dynamics and also identifies important residues at the binding interfaces as potential drug targets for the inhibition of cancer cells.
Collapse
Affiliation(s)
- Wenhan Guo
- Computational Science Program, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Tolulope Ayodeji Ale
- Computational Science Program, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Shengjie Sun
- Computational Science Program, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Jason E. Sanchez
- Computational Science Program, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Lin Li
- Computational Science Program, University of Texas at El Paso, El Paso, TX 79902, USA
- Department of Physics, University of Texas at El Paso, El Paso, TX 79902, USA
| |
Collapse
|
7
|
KIF5C deficiency causes abnormal cortical neuronal migration, dendritic branching, and spine morphology in mice. Pediatr Res 2022; 92:995-1002. [PMID: 34966180 DOI: 10.1038/s41390-021-01922-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 12/13/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Malformation of cortical development (MCD) includes a variety of developmental disorders that are common causes of neurodevelopmental delay and epilepsy. Most recently, clinical studies found that patients carrying KIF5C mutations present early-onset MCD; however, the underlying mechanisms remain elusive. METHODS KIF5C expression level was examined in mouse primary cortical neurons and human ips-derived forebrain organoids. We studied the cortical neuronal migration, dendritic branching, and dendritic spine growth after knocking down the KIF5C gene by electroporation in vitro and in vivo. Then, we studied the transcriptome differences between the knockdown and control groups through RNA sequencing. RESULTS We observed high KIF5C expression in neurons during the early developmental stage in mice and the human brain. Kif5c deficiency results in disturbed cortical neuronal migration, dendritic, and spine growth. Finally, we found that Kif5c knockdown affected several genes associated with cortical neuronal development in vitro. CONCLUSIONS These results suggested a critical role for Kif5c in cortical development, providing insights into underlying pathogenic factors of kinesins in MCD. IMPACT KIF5C mutation-related MCD might be caused by abnormal early cortical neuronal development. Kif5c deficiency led to abnormal cortical neuronal dendritic and spine growth and neuronal migration. Our findings explain how Kif5c deficiency is involved in the aberrant development of cortical neurons and provide a new perspective for the pathology of MCD.
Collapse
|
8
|
Xie Y, Li L. Computational Study on E-Hooks of Tubulins in the Binding Process with Kinesin. Int J Mol Sci 2022; 23:ijms23042035. [PMID: 35216151 PMCID: PMC8877516 DOI: 10.3390/ijms23042035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/10/2022] Open
Abstract
Cargo transport within cells is essential to healthy cells, which requires microtubules-based motors, including kinesin. The C-terminal tails (E-hooks) of alpha and beta tubulins of microtubules have been proven to play important roles in interactions between the kinesins and tubulins. Here, we implemented multi-scale computational methods in E-hook-related analyses, including flexibility investigations of E-hooks, binding force calculations at binding interfaces between kinesin and tubulins, electrostatic potential calculations on the surface of kinesin and tubulins. Our results show that E-hooks have several functions during the binding process: E-hooks utilize their own high flexibilities to increase the chances of reaching a kinesin; E-hooks help tubulins to be more attractive to kinesin. Besides, we also observed the differences between alpha and beta tubulins: beta tubulin shows a higher flexibility than alpha tubulin; beta tubulin generates stronger attractive forces (about twice the strengths) to kinesin at different distances, no matter with E-hooks in the structure or not. Those facts may indicate that compared to alpha tubulin, beta tubulin contributes more to attracting and catching a kinesin to microtubule. Overall, this work sheds the light on microtubule studies, which will also benefit the treatments of neurodegenerative diseases, cancer treatments, and preventions in the future.
Collapse
Affiliation(s)
- Yixin Xie
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79912, USA;
| | - Lin Li
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79912, USA;
- Department of Physics, The University of Texas at El Paso, El Paso, TX 79912, USA
- Correspondence:
| |
Collapse
|
9
|
Chamariya R, Suvarna V. Role of KSP inhibitors as anti-cancer therapeutics: an update. Anticancer Agents Med Chem 2022; 22:2517-2538. [PMID: 35043768 DOI: 10.2174/1871520622666220119093105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/03/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
Regardless of the growing discovery of anticancer treatments, targeting cancer-specific pathways, cytotoxic therapy still maintained its abundant clinical significance based on the fact that tumours harbour a greater population of actively dividing cells than normal tissues. Conventional anti-mitotic agents or microtubule poisons acting on the major mitotic spindle protein tubulin have been effectively used in clinical settings for cancer chemotherapy over the last three decades. However, use of these drugs is associated with limited clinical utility due to serious side effects such as debilitating and dose-limiting peripheral neuropathy, myelosuppression, drug resistance and allergic reactions. Therefore, research initiatives have been undertaken to develop novel microtubule motor proteins inhibitors that can potentially circumvent the limitations associated with conventional microtubule poisons. Kinesin spindle proteins (KSP) belonging to the kinesin-5 family play a crucial role during mitosis and unregulated cell proliferation. Several evidences from preclinical studies and different phases of clinical trials have presented kinesin spindle protein as a promising target for cancer therapeutics. kinesin spindle protein inhibitors causing mitosis disruption without interfering with microtubule dynamics in non-dividing cells offer a potential therapeutic alternative for the management of several major cancer types and are devoid of side effects associated with classical anti-mitotic drugs. This review summarizes recent data highlighting progress in the discovery of targeted KSP inhibitors and presents the development of scaffolds, structure-activity relationships, and outcomes of biological, and enzyme inhibition studies. We reviewed the recent literature reports published over last decade, using various electronic database searches such as PubMed, Embase, Medline, Web of Science, and Google Scholar. Clinical trial data till 2021 was retrieved from ClinicalTrial.gov. Major chemical classes developed as selective KSP inhibitors include dihydropyrimidines, β-carbolines, carbazoles, benzimidazoles, fused aryl derivatives, pyrimidines, fused pyrimidines, quinazolines, quinolones, thiadiazolines, spiropyran and azobenzenes. Drugs such as filanesib, litronesib, ispinesib have entered clinical trials, the most advanced phase explored being Phase II. KSP inhibitors have exhibited promising results; however, continued exploration is greatly required to establish the clinical potential of KSP inhibitors.
Collapse
Affiliation(s)
- Rinkal Chamariya
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.L. Mehta Road, Vile Parle (West), Mumbai - 400056, Maharashtra, India
| | - Vasanti Suvarna
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.L. Mehta Road, Vile Parle (West), Mumbai - 400056, Maharashtra, India
| |
Collapse
|
10
|
Ospina OE, Lemmon AR, Dye M, Zdyrski C, Holland S, Stribling D, Kortyna ML, Lemmon EM. Neurogenomic divergence during speciation by reinforcement of mating behaviors in chorus frogs (Pseudacris). BMC Genomics 2021; 22:711. [PMID: 34600496 PMCID: PMC8487493 DOI: 10.1186/s12864-021-07995-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Species interactions can promote mating behavior divergence, particularly when these interactions are costly due to maladaptive hybridization. Selection against hybridization can indirectly cause evolution of reproductive isolation within species, a process termed cascade reinforcement. This process can drive incipient speciation by generating divergent selection pressures among populations that interact with different species assemblages. Theoretical and empirical studies indicate that divergent selection on gene expression networks has the potential to increase reproductive isolation among populations. After identifying candidate synaptic transmission genes derived from neurophysiological studies in anurans, we test for divergence of gene expression in a system undergoing cascade reinforcement, the Upland Chorus Frog (Pseudacris feriarum). RESULTS Our analyses identified seven candidate synaptic transmission genes that have diverged between ancestral and reinforced populations of P. feriarum, including five that encode synaptic vesicle proteins. Our gene correlation network analyses revealed four genetic modules that have diverged between these populations, two possessing a significant concentration of neurotransmission enrichment terms: one for synaptic membrane components and the other for metabolism of the neurotransmitter nitric oxide. We also ascertained that a greater number of genes have diverged in expression by geography than by sex. Moreover, we found that more genes have diverged within females as compared to males between populations. Conversely, we observed no difference in the number of differentially-expressed genes within the ancestral compared to the reinforced population between the sexes. CONCLUSIONS This work is consistent with the idea that divergent selection on mating behaviors via cascade reinforcement contributed to evolution of gene expression in P. feriarum. Although our study design does not allow us to fully rule out the influence of environment and demography, the fact that more genes diverged in females than males points to a role for cascade reinforcement. Our discoveries of divergent candidate genes and gene networks related to neurotransmission support the idea that neural mechanisms of acoustic mating behaviors have diverged between populations, and agree with previous neurophysiological studies in frogs. Increasing support for this hypothesis, however, will require additional experiments under common garden conditions. Our work points to the importance of future replicated and tissue-specific studies to elucidate the relative contribution of gene expression divergence to the evolution of reproductive isolation during incipient speciation.
Collapse
Affiliation(s)
- Oscar E Ospina
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
- Present address: Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 13131 USF Magnolia Drive, Tampa, FL, 33612, USA
| | - Alan R Lemmon
- Department of Scientific Computing, Florida State University, 400 Dirac Science Library, Tallahassee, FL, 32306, USA
| | - Mysia Dye
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
| | - Christopher Zdyrski
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
- Present address: Genetics and Genomics Program, Iowa State University, 2437 Pammel Drive, Ames, IA, 50011, USA
| | - Sean Holland
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
| | - Daniel Stribling
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
- Present address: Department of Molecular Genetics and Microbiology, Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Michelle L Kortyna
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA
| | - Emily Moriarty Lemmon
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, 50011, Ames, IA, USA.
| |
Collapse
|
11
|
Xian Y, Xie Y, Silva SM, Karki CB, Qiu W, Li L. StructureMan: A Structure Manipulation Tool to Study Large Scale Biomolecular Interactions. Front Mol Biosci 2021; 7:627087. [PMID: 33505991 PMCID: PMC7831659 DOI: 10.3389/fmolb.2020.627087] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/10/2020] [Indexed: 11/22/2022] Open
Abstract
Studying biomolecular interactions is a crucial but challenging task. Due to their large scales, many biomolecular interactions are difficult to be simulated via all atom models. An effective approach to investigate the biomolecular interactions is highly demanded in many areas. Here we introduce a Structure Manipulation (StructureMan) program to operate the structures when studying the large-scale biomolecular interactions. This novel StructureMan tool provides comprehensive operations which can be utilized to study the interactions in various large biological systems. Combining with electrostatic calculation programs such as DelPhi and DelPhiForce, StructureMan was implemented to reveal the detailed electrostatic features in two large biological examples, the viral capsid and molecular motor-microtubule complexes. Applications on these two examples revealed interesting binding mechanisms in the viral capsid and molecular motor. Such applications demonstrated that the StructureMan can be widely used when studying the biomolecular interactions in large scale biological problems. This novel tool provides an alternative approach to efficiently study the biomolecular interactions, especially for large scale biology systems. The StructureMan tool is available at our website: http://compbio.utep.edu/static/downloads/script-for-munipulation2.zip.
Collapse
Affiliation(s)
- Yuejiao Xian
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, United States
| | - Yixin Xie
- Computational Science Program, University of Texas at El Paso, El Paso, TX, United States
| | - Sebastian Miki Silva
- Department of Physics, University of Texas at El Paso, El Paso, TX, United States
| | - Chitra B Karki
- Computational Science Program, University of Texas at El Paso, El Paso, TX, United States
| | - Weihong Qiu
- Department of Physics, Oregon State University, Corvallis, OR, United States.,Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR, United States
| | - Lin Li
- Computational Science Program, University of Texas at El Paso, El Paso, TX, United States.,Department of Physics, University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
12
|
Atherton J, Hummel JJA, Olieric N, Locke J, Peña A, Rosenfeld SS, Steinmetz MO, Hoogenraad CC, Moores CA. The mechanism of kinesin inhibition by kinesin-binding protein. eLife 2020; 9:e61481. [PMID: 33252036 PMCID: PMC7746232 DOI: 10.7554/elife.61481] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/28/2020] [Indexed: 12/25/2022] Open
Abstract
Subcellular compartmentalisation is necessary for eukaryotic cell function. Spatial and temporal regulation of kinesin activity is essential for building these local environments via control of intracellular cargo distribution. Kinesin-binding protein (KBP) interacts with a subset of kinesins via their motor domains, inhibits their microtubule (MT) attachment, and blocks their cellular function. However, its mechanisms of inhibition and selectivity have been unclear. Here we use cryo-electron microscopy to reveal the structure of KBP and of a KBP-kinesin motor domain complex. KBP is a tetratricopeptide repeat-containing, right-handed α-solenoid that sequesters the kinesin motor domain's tubulin-binding surface, structurally distorting the motor domain and sterically blocking its MT attachment. KBP uses its α-solenoid concave face and edge loops to bind the kinesin motor domain, and selected structure-guided mutations disrupt KBP inhibition of kinesin transport in cells. The KBP-interacting motor domain surface contains motifs exclusively conserved in KBP-interacting kinesins, suggesting a basis for kinesin selectivity.
Collapse
Affiliation(s)
- Joseph Atherton
- Randall Centre for Cell and Molecular Biophysics, King’s CollegeLondonUnited Kingdom
- Institute of Structural and Molecular Biology, Birkbeck, University of LondonLondonUnited Kingdom
| | - Jessica JA Hummel
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer InstitutVilligen PSISwitzerland
| | - Julia Locke
- Institute of Structural and Molecular Biology, Birkbeck, University of LondonLondonUnited Kingdom
| | - Alejandro Peña
- Institute of Structural and Molecular Biology, Birkbeck, University of LondonLondonUnited Kingdom
| | | | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer InstitutVilligen PSISwitzerland
- University of Basel, BiozentrumBaselSwitzerland
| | - Casper C Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht UniversityUtrechtNetherlands
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck, University of LondonLondonUnited Kingdom
| |
Collapse
|
13
|
Zorgniotti A, Ditamo Y, Arce CA, Bisig CG. Irreversible incorporation of L-dopa into the C-terminus of α-tubulin inhibits binding of molecular motor KIF5B to microtubules and alters mitochondrial traffic along the axon. Neurobiol Dis 2020; 147:105164. [PMID: 33171229 DOI: 10.1016/j.nbd.2020.105164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
L-dopa is the most effective drug used to date for management of Parkinson's disease symptoms. Unfortunately, long-term administration of L-dopa often results in development of motor disorders, including dyskinesias. Despite extensive research on L-dopa-induced dyskinesia, its pathogenesis remains poorly understood. We demonstrated previously that L-dopa can be post-translationally incorporated into the C-terminus of α-tubulin in living cells. In the present study, we investigated the effect of the presence of L-dopa-tubulin-enriched microtubules on mitochondrial traffic mediated by molecular motor KIF5B. Using biochemical approaches in combination with experiments on neuronal cell lines and mouse hippocampal primary cultures, we demonstrated that L-dopa incorporation into tubulin is irreversible. Transport of mitochondria along the axon was altered after L-dopa treatment of cells. In L-dopa-treated cells, mitochondria had reduced ability to reach the distal segment of the axon, spent more time in pause, and showed reduced velocity of anterograde movement. KIF5B motor, a member of the kinesin family involved in mitochondrial transport in neurons, showed reduced affinity for Dopa-tubulin-containing microtubules. Our findings, taken together, suggest that tyrosination state of tubulin (and microtubules) is altered by L-dopa incorporation into tubulin; the gradual increase in amount of altered microtubules affects microtubule functioning, impairs mitochondrial traffic and distribution, and this could be relevant in Parkinson's disease patients chronically treated with L-dopa.
Collapse
Affiliation(s)
- Agustina Zorgniotti
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - Yanina Ditamo
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - Carlos A Arce
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| | - C Gaston Bisig
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina.
| |
Collapse
|
14
|
Venugopalan PL, Esteban-Fernández de Ávila B, Pal M, Ghosh A, Wang J. Fantastic Voyage of Nanomotors into the Cell. ACS NANO 2020; 14:9423-9439. [PMID: 32701260 DOI: 10.1021/acsnano.0c05217] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Richard Feynman's 1959 vision of controlling devices at small scales and swallowing the surgeon has inspired the science-fiction Fantastic Voyage film and has played a crucial role in the rapid development of the microrobotics field. Sixty years later, we are currently witnessing a dramatic progress in this field, with artificial micro- and nanoscale robots moving within confined spaces, down to the cellular level, and performing a wide range of biomedical applications within the cellular interior while addressing the limitations of common passive nanosystems. In this review article, we discuss key recent advances in the field of micro/nanomotors toward important cellular applications. Specifically, we outline the distinct capabilities of nanoscale motors for such cellular applications and illustrate how the active movement of nanomotors leads to distinct advantages of rapid cell penetration, accelerated intracellular sensing, and effective intracellular delivery toward enhanced therapeutic efficiencies. We finalize by discussing the future prospects and key challenges that such micromotor technology face toward implementing practical intracellular applications. By increasing our knowledge of nanomotors' cell entry and of their behavior within the intracellular space, and by successfully addressing key challenges, we expect that next-generation nanomotors will lead to exciting advances toward cell-based diagnostics and therapy.
Collapse
Affiliation(s)
- Pooyath Lekshmy Venugopalan
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | | | - Malay Pal
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Ambarish Ghosh
- Centre for Nano Science and Engineering, Department of Physics, Indian Institute of Science, Bengaluru 560012, India
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
15
|
Abraham Z, Hawley E, Hayosh D, Webster-Wood VA, Akkus O. Kinesin and Dynein Mechanics: Measurement Methods and Research Applications. J Biomech Eng 2019; 140:2654261. [PMID: 28901373 DOI: 10.1115/1.4037886] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Indexed: 11/08/2022]
Abstract
Motor proteins play critical roles in the normal function of cells and proper development of organisms. Among motor proteins, failings in the normal function of two types of proteins, kinesin and dynein, have been shown to lead many pathologies, including neurodegenerative diseases and cancers. As such, it is critical to researchers to understand the underlying mechanics and behaviors of these proteins, not only to shed light on how failures may lead to disease, but also to guide research toward novel treatment and nano-engineering solutions. To this end, many experimental techniques have been developed to measure the force and motility capabilities of these proteins. This review will (a) discuss such techniques, specifically microscopy, atomic force microscopy (AFM), optical trapping, and magnetic tweezers, and (b) the resulting nanomechanical properties of motor protein functions such as stalling force, velocity, and dependence on adenosine triphosophate (ATP) concentrations will be comparatively discussed. Additionally, this review will highlight the clinical importance of these proteins. Furthermore, as the understanding of the structure and function of motor proteins improves, novel applications are emerging in the field. Specifically, researchers have begun to modify the structure of existing proteins, thereby engineering novel elements to alter and improve native motor protein function, or even allow the motor proteins to perform entirely new tasks as parts of nanomachines. Kinesin and dynein are vital elements for the proper function of cells. While many exciting experiments have shed light on their function, mechanics, and applications, additional research is needed to completely understand their behavior.
Collapse
Affiliation(s)
- Zachary Abraham
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Emma Hawley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Daniel Hayosh
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Victoria A Webster-Wood
- Mem. ASME Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 e-mail:
| | - Ozan Akkus
- Mem. ASME Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
16
|
Ogunwa TH, Laudadio E, Galeazzi R, Miyanishi T. Insights into the Molecular Mechanisms of Eg5 Inhibition by (+)-Morelloflavone. Pharmaceuticals (Basel) 2019; 12:ph12020058. [PMID: 30995725 PMCID: PMC6630617 DOI: 10.3390/ph12020058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
(+)-Morelloflavone (MF) is an antitumor biflavonoid that is found in the Garcinia species. Recently, we reported MF as a novel inhibitor of ATPase and microtubules-gliding activities of the kinesin spindle protein (Eg5) in vitro. Herein, we provide dynamical insights into the inhibitory mechanisms of MF against Eg5, which involves binding of the inhibitor to the loop5/α2/α3 allosteric pocket. Molecular dynamics simulations were carried out for 100 ns on eight complexes: Eg5-Adenosine diphosphate (Eg5-ADP), Eg5-ADP-S-trityl-l-cysteine (Eg5-ADP-STLC), Eg5-ADP-ispinesib, Eg5-ADP-MF, Eg5-Adenosine triphosphate (Eg5-ATP), Eg5-ATP-STLC, Eg5-ATP-ispinesib, and Eg5-ATP-MF complexes. Structural and energetic analyses were done using Umbrella sampling, Molecular Mechanics Poisson–Boltzmann Surface Area (MM/PBSA) method, GROMACS analysis toolkit, and virtual molecular dynamics (VMD) utilities. The results were compared with those of the known Eg5 inhibitors; ispinesib, and STLC. Our data strongly support a stable Eg5-MF complex, with significantly low binding energy and reduced flexibility of Eg5 in some regions, including loop5 and switch I. Furthermore, the loop5 Trp127 was trapped in a downward position to keep the allosteric pocket of Eg5 in the so-called “closed conformation”, comparable to observations for STLC. Altered structural conformations were also visible within various regions of Eg5, including switch I, switch II, α2/α3 helices, and the tubulin-binding region, indicating that MF might induce modifications in the Eg5 structure to compromise its ATP/ADP binding and conversion process as well as its interaction with microtubules. The described mechanisms are crucial for understanding Eg5 inhibition by MF.
Collapse
Affiliation(s)
- Tomisin Happy Ogunwa
- Department of Environmental Studies, Graduate School of Fisheries and Environmental Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | - Emiliano Laudadio
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - Roberta Galeazzi
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | - Takayuki Miyanishi
- Department of Environmental Studies, Graduate School of Fisheries and Environmental Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| |
Collapse
|
17
|
Hu X, Guiseppi-Elie A, Dinu CZ. Biomolecular interfaces based on self-assembly and self-recognition form biosensors capable of recording molecular binding and release. NANOSCALE 2019; 11:4987-4998. [PMID: 30839012 DOI: 10.1039/c8nr10090j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This research proposed to create the next generation of versatile electrochemical-based biosensors capable of monitoring target capture and release as dictated by molecular binding or unbinding. The biosensor integrates cellular machines (i.e., microtubules, structural elements of cells and kinesin molecular motors involved in cellular transport) as functional units; its assembly is based on molecular self-assembly and self-recognition. Our results demonstrate that the designed biosensor was capable of allowing detection of binding and unbinding events based on redox reactions at user-controlled electrode interfaces. The analysis also showed that the sensitivity of the designed biosensor or its ability to record such events could be user-controlled at any given time by adjusting the energy source that "fuels" the system.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Chemical and Biomedical Engineering, West Virginia University, WV, USA.
| | | | | |
Collapse
|
18
|
Structural consequences of hereditary spastic paraplegia disease-related mutations in kinesin. Proc Natl Acad Sci U S A 2018; 115:E10822-E10829. [PMID: 30366951 DOI: 10.1073/pnas.1810622115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A wide range of mutations in the kinesin motor Kif5A have been linked to a neuronal disorder called hereditary spastic paraplegia (HSP). The position of these mutations can vary, and a range of different motile behaviors have been observed, indicating that the HSP mutants can alter distinct aspects of kinesin mechanochemistry. While focusing on four key HSP-associated mutants, this study examined the structural and dynamic perturbations that arise from these mutations using a series of different computational methods, ranging from bioinformatics analyses to all-atom simulations, that account for solvent effects explicitly. We show that two catalytic domain mutations (R280S and K253N) reduce the microtubule (MT) binding affinity of the kinesin head domains appreciably, while N256S has a much smaller impact. Bioinformatics analysis suggests that the stalk mutation A361V perturbs motor dimerization. Subsequent integration of these effects into a coarse-grained structure-based model of dimeric kinesin revealed that the order-disorder transition of the neck linker is substantially affected, indicating a hampered directionality and processivity of kinesin. The present analyses therefore suggest that, in addition to kinesin-MT binding and coiled-coil dimerization, HSP mutations affecting motor stepping transitions and processivity can lead to disease.
Collapse
|
19
|
Pernigo S, Chegkazi MS, Yip YY, Treacy C, Glorani G, Hansen K, Politis A, Bui S, Dodding MP, Steiner RA. Structural basis for isoform-specific kinesin-1 recognition of Y-acidic cargo adaptors. eLife 2018; 7:38362. [PMID: 30320553 PMCID: PMC6214655 DOI: 10.7554/elife.38362] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/14/2018] [Indexed: 12/19/2022] Open
Abstract
The light chains (KLCs) of the heterotetrameric microtubule motor kinesin-1, that bind to cargo adaptor proteins and regulate its activity, have a capacity to recognize short peptides via their tetratricopeptide repeat domains (KLCTPR). Here, using X-ray crystallography, we show how kinesin-1 recognizes a novel class of adaptor motifs that we call ‘Y-acidic’ (tyrosine flanked by acidic residues), in a KLC-isoform specific manner. Binding specificities of Y-acidic motifs (present in JIP1 and in TorsinA) to KLC1TPR are distinct from those utilized for the recognition of W-acidic motifs found in adaptors that are KLC- isoform non-selective. However, a partial overlap on their receptor binding sites implies that adaptors relying on Y-acidic and W-acidic motifs must act independently. We propose a model to explain why these two classes of motifs that bind to the concave surface of KLCTPR with similar low micromolar affinity can exhibit different capacities to promote kinesin-1 activity.
Collapse
Affiliation(s)
- Stefano Pernigo
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Magda S Chegkazi
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Yan Y Yip
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Conor Treacy
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Giulia Glorani
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Kjetil Hansen
- Department of Chemistry, King's College London, London, United Kingdom
| | - Argyris Politis
- Department of Chemistry, King's College London, London, United Kingdom
| | - Soi Bui
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Mark P Dodding
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.,School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Roberto A Steiner
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
20
|
Vulinovic F, Krajka V, Hausrat TJ, Seibler P, Alvarez-Fischer D, Madoev H, Park JS, Kumar KR, Sue CM, Lohmann K, Kneussel M, Klein C, Rakovic A. Motor protein binding and mitochondrial transport are altered by pathogenic TUBB4A variants. Hum Mutat 2018; 39:1901-1915. [PMID: 30079973 DOI: 10.1002/humu.23602] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/29/2018] [Indexed: 12/21/2022]
Abstract
Mutations in TUBB4A have been identified to cause a wide phenotypic spectrum of diseases ranging from hereditary generalized dystonia with whispering dysphonia (DYT-TUBB4A) and hereditary spastic paraplegia (HSP) to leukodystrophy hypomyelination with atrophy of the basal ganglia and cerebellum (H-ABC). TUBB4A encodes the brain-specific β-tubulin isotype, β-tubulin 4A. To elucidate the pathogenic mechanisms conferred by TUBB4A mutations leading to the different phenotypes, we functionally characterized three pathogenic TUBB4A variants (c.4C>G,p.R2G; c.745G>A,p.D249N; c.811G>A, p.A271T) as representatives of the mutational and disease spectrum) in human neuroblastoma cells and human induced pluripotent stem cell (iPSC)-derived neurons. We showed that mRNA stability was not affected by any of the TUBB4A variants. Although two mutations (p.R2G and p.D249N) are located at the α/β-tubulin interdimer interface, we confirmed incorporation of all TUBB4A mutants into the microtubule network. However, we showed that the mutations p.D249N and p.A271T interfered with motor protein binding to microtubules and impaired neurite outgrowth and microtubule dynamics. Finally, TUBB4A mutations, as well as heterozygous knockout of TUBB4A, disrupted mitochondrial transport in iPSC-derived neurons. Taken together, our findings suggest that functional impairment of microtubule-associated transport is a shared pathogenic mechanism by which the TUBB4A mutations studied here cause a spectrum of diseases.
Collapse
Affiliation(s)
- Franca Vulinovic
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Victor Krajka
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Torben J Hausrat
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Harutyun Madoev
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Jin-Sung Park
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
| | - Kishore R Kumar
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, Royal North Shore Hospital and the University of Sydney, St. Leonards, New South Wales, Australia
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
21
|
Misiura M, Wang Q, Cheung MS, Kolomeisky AB. Theoretical Investigations of the Role of Mutations in Dynamics of Kinesin Motor Proteins. J Phys Chem B 2018; 122:4653-4661. [PMID: 29630822 DOI: 10.1021/acs.jpcb.8b00830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Motor proteins are active enzymatic molecules that are critically important for a variety of biological phenomena. It is known that some neurodegenerative diseases are caused by specific mutations in motor proteins that lead to their malfunctioning. Hereditary spastic paraplegia is one of such diseases, and it is associated with the mutations in the neuronal conventional kinesin gene, producing the decreased speed and processivity of this motor protein. Despite the importance of this problem, there is no clear understanding on the role of mutations in modifying dynamic properties of motor proteins. In this work, we investigate theoretically the molecular basis for negative effects of two specific mutations, N256S and R280S, on the dynamics of kinesin motor proteins. We hypothesize that these mutations might accelerate the adenosine triphosphate (ATP) release by increasing the probability of open conformations for the ATP-binding pocket. Our approach is based on the use of coarse-grained structure-based molecular dynamics simulations to analyze the conformational changes and chemical transitions in the kinesin molecule, which is also supplemented by investigation of a mesoscopic discrete-state stochastic model. Computer simulations suggest that mutations N256S and R280S can decrease the free energy difference between open and closed biochemical states, making the open conformation more stable and the ATP release faster, which is in agreement with our hypothesis. Furthermore, we show that in the case of N256S mutation, this effect is caused by disruption of interactions between α helix and switch I and loop L11 structural elements. Our computational results are qualitatively supported by the explicit analysis of the discrete-state stochastic model.
Collapse
Affiliation(s)
| | | | - Margaret S Cheung
- Department of Physics , University of Houston , Houston , Texas 77204 , United States
| | | |
Collapse
|
22
|
Hwang W, Hyeon C. Energetic Costs, Precision, and Transport Efficiency of Molecular Motors. J Phys Chem Lett 2018; 9:513-520. [PMID: 29329502 DOI: 10.1021/acs.jpclett.7b03197] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
An efficient molecular motor would deliver cargo to the target site at a high speed and in a punctual manner while consuming a minimal amount of energy. According to a recently formulated thermodynamic principle, referred to as the thermodynamic uncertainty relation, the travel distance of a motor and its variance are, however, constrained by the free energy being consumed. Here we use the principle underlying the uncertainty relation to quantify the transport efficiency of molecular motors for varying ATP concentration ([ATP]) and applied load (f). Our analyses of experimental data find that transport efficiencies of the motors studied here are semioptimized under the cellular condition. The efficiency is significantly deteriorated for a kinesin-1 mutant that has a longer neck-linker, which underscores the importance of molecular structure. It is remarkable to recognize that, among many possible directions for optimization, biological motors have evolved to optimize the transport efficiency in particular.
Collapse
Affiliation(s)
- Wonseok Hwang
- Korea Institute for Advanced Study , Seoul 02455, Republic of Korea
| | - Changbong Hyeon
- Korea Institute for Advanced Study , Seoul 02455, Republic of Korea
| |
Collapse
|
23
|
A deterministic model for one-dimensional excluded flow with local interactions. PLoS One 2017; 12:e0182074. [PMID: 28796838 PMCID: PMC5552133 DOI: 10.1371/journal.pone.0182074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/26/2017] [Indexed: 11/21/2022] Open
Abstract
Natural phenomena frequently involve a very large number of interacting molecules moving in confined regions of space. Cellular transport by motor proteins is an example of such collective behavior. We derive a deterministic compartmental model for the unidirectional flow of particles along a one-dimensional lattice of sites with nearest-neighbor interactions between the particles. The flow between consecutive sites is governed by a “soft” simple exclusion principle and by attracting or repelling forces between neighboring particles. Using tools from contraction theory, we prove that the model admits a unique steady-state and that every trajectory converges to this steady-state. Analysis and simulations of the effect of the attracting and repelling forces on this steady-state highlight the crucial role that these forces may play in increasing the steady-state flow, and reveal that this increase stems from the alleviation of traffic jams along the lattice. Our theoretical analysis clarifies microscopic aspects of complex multi-particle dynamic processes.
Collapse
|
24
|
Li Q, King SJ, Xu J. Native kinesin-1 does not bind preferentially to GTP-tubulin-rich microtubules in vitro. Cytoskeleton (Hoboken) 2017; 74:356-366. [PMID: 28699205 DOI: 10.1002/cm.21386] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/27/2017] [Accepted: 07/04/2017] [Indexed: 11/06/2022]
Abstract
Molecular motors such as kinesin-1 work in small teams to actively shuttle cargos in cells, for example in polarized transport in axons. Here, we examined the potential regulatory role of the nucleotide state of tubulin on the run length of cargos carried by multiple kinesin motors, using an optical trapping-based in vitro assay. Based on a previous report that kinesin binds preferentially to GTP-tubulin-rich microtubules, we anticipated that multiple-kinesin cargos would run substantially greater distances along GMPCPP microtubules than along GDP microtubules. Surprisingly, we did not uncover any significant differences in run length between microtubule types. A combination of single-molecule experiments, comparison with previous theory, and classic microtubule affinity pulldown assays revealed that native kinesin-1 does not bind preferentially to GTP-tubulin-rich microtubules. The apparent discrepancy between our observations and the previous report likely reflects differences in post-translational modifications between the native motors used here and the recombinant motors examined previously. Future investigations will help shed light on the interplay between the motor's post-translational modification and the microtubule's nucleotide-binding state for transport regulation in vivo.
Collapse
Affiliation(s)
- Qiaochu Li
- Department of Physics, University of California, Merced, California
| | - Stephen J King
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida
| | - Jing Xu
- Department of Physics, University of California, Merced, California
| |
Collapse
|
25
|
Abstract
Millisecond-scale conformational transitions represent a seminal challenge for traditional molecular dynamics simulations, even with the help of high-end supercomputer architectures. Such events are particularly relevant to the study of molecular motors-proteins or abiological constructs that convert chemical energy into mechanical work. Here, we present a hybrid-simulation scheme combining an array of methods including elastic network models, transition path sampling, and advanced free-energy methods, possibly in conjunction with generalized-ensemble schemes to deliver a viable option for capturing the millisecond-scale motor steps of biological motors. The methodology is already implemented in large measure in popular molecular dynamics programs, and it can leverage the massively parallel capabilities of petascale computers. The applicability of the hybrid method is demonstrated with two examples, namely cyclodextrin-based motors and V-type ATPases.
Collapse
Affiliation(s)
- Abhishek Singharoy
- Theoretical and Computational Biophysics Group, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana, Illinois 61801
| | - Christophe Chipot
- Laboratoire International Associé Centre National de la Recherche Scientifique et University of Illinois at Urbana-Champaign, Unité Mixte de Recherche n°7565, Université de Lorraine, B.P. 70239, 54506 Vandœuvre-lès-Nancy cedex, France
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, Illinois 61801
| |
Collapse
|
26
|
Gramlich MW, Conway L, Liang WH, Labastide JA, King SJ, Xu J, Ross JL. Single Molecule Investigation of Kinesin-1 Motility Using Engineered Microtubule Defects. Sci Rep 2017; 7:44290. [PMID: 28287156 PMCID: PMC5347089 DOI: 10.1038/srep44290] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/06/2017] [Indexed: 12/05/2022] Open
Abstract
The structure of the microtubule is tightly regulated in cells via a number of microtubule associated proteins and enzymes. Microtubules accumulate structural defects during polymerization, and defect size can further increase under mechanical stresses. Intriguingly, microtubule defects have been shown to be targeted for removal via severing enzymes or self-repair. The cell’s control in defect removal suggests that defects can impact microtubule-based processes, including molecular motor-based intracellular transport. We previously demonstrated that microtubule defects influence cargo transport by multiple kinesin motors. However, mechanistic investigations of the observed effects remained challenging, since defects occur randomly during polymerization and are not directly observable in current motility assays. To overcome this challenge, we used end-to-end annealing to generate defects that are directly observable using standard epi-fluorescence microscopy. We demonstrate that the annealed sites recapitulate the effects of polymerization-derived defects on multiple-motor transport, and thus represent a simple and appropriate model for naturally-occurring defects. We found that single kinesins undergo premature dissociation, but not preferential pausing, at the annealed sites. Our findings provide the first mechanistic insight to how defects impact kinesin-based transport. Preferential dissociation on the single-molecule level has the potential to impair cargo delivery at locations of microtubule defect sites in vivo.
Collapse
Affiliation(s)
- Michael W Gramlich
- Department of Physics, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Leslie Conway
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Winnie H Liang
- Department of Physics, University of California Merced, Merced, CA 95343, USA
| | - Joelle A Labastide
- Department of Physics, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Stephen J King
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, USA
| | - Jing Xu
- Department of Physics, University of California Merced, Merced, CA 95343, USA
| | - Jennifer L Ross
- Department of Physics, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
27
|
White JA, Banerjee R, Gunawardena S. Axonal Transport and Neurodegeneration: How Marine Drugs Can Be Used for the Development of Therapeutics. Mar Drugs 2016; 14:E102. [PMID: 27213408 PMCID: PMC4882576 DOI: 10.3390/md14050102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 11/23/2022] Open
Abstract
Unlike virtually any other cells in the human body, neurons are tasked with the unique problem of transporting important factors from sites of synthesis at the cell bodies, across enormous distances, along narrow-caliber projections, to distally located nerve terminals in order to maintain cell viability. As a result, axonal transport is a highly regulated process whereby necessary cargoes of all types are packaged and shipped from one end of the neuron to the other. Interruptions in this finely tuned transport have been linked to many neurodegenerative disorders including Alzheimer's (AD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) suggesting that this pathway is likely perturbed early in disease progression. Therefore, developing therapeutics targeted at modifying transport defects could potentially avert disease progression. In this review, we examine a variety of potential compounds identified from marine aquatic species that affect the axonal transport pathway. These compounds have been shown to function in microtubule (MT) assembly and maintenance, motor protein control, and in the regulation of protein degradation pathways, such as the autophagy-lysosome processes, which are defective in many degenerative diseases. Therefore, marine compounds have great potential in developing effective treatment strategies aimed at early defects which, over time, will restore transport and prevent cell death.
Collapse
Affiliation(s)
- Joseph A White
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Rupkatha Banerjee
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY 14260, USA.
| |
Collapse
|
28
|
Karunagaran S, Subhashchandrabose S, Lee KW, Meganathan C. Investigation on the isoform selectivity of novel kinesin-like protein 1 (KIF11) inhibitor using chemical feature based pharmacophore, molecular docking, and quantum mechanical studies. Comput Biol Chem 2016; 61:47-61. [DOI: 10.1016/j.compbiolchem.2016.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/14/2015] [Accepted: 01/08/2016] [Indexed: 12/28/2022]
|
29
|
Multiscale method for modeling binding phenomena involving large objects: application to kinesin motor domains motion along microtubules. Sci Rep 2016; 6:23249. [PMID: 26988596 PMCID: PMC4796874 DOI: 10.1038/srep23249] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/03/2016] [Indexed: 11/30/2022] Open
Abstract
Many biological phenomena involve the binding of proteins to a large object. Because the electrostatic forces that guide binding act over large distances, truncating the size of the system to facilitate computational modeling frequently yields inaccurate results. Our multiscale approach implements a computational focusing method that permits computation of large systems without truncating the electrostatic potential and achieves the high resolution required for modeling macromolecular interactions, all while keeping the computational time reasonable. We tested our approach on the motility of various kinesin motor domains. We found that electrostatics help guide kinesins as they walk: N-kinesins towards the plus-end, and C-kinesins towards the minus-end of microtubules. Our methodology enables computation in similar, large systems including protein binding to DNA, viruses, and membranes.
Collapse
|
30
|
Mauvezin C, Neisch AL, Ayala CI, Kim J, Beltrame A, Braden CR, Gardner MK, Hays TS, Neufeld TP. Coordination of autophagosome-lysosome fusion and transport by a Klp98A-Rab14 complex in Drosophila. J Cell Sci 2016; 129:971-82. [PMID: 26763909 DOI: 10.1242/jcs.175224] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 01/07/2016] [Indexed: 01/06/2023] Open
Abstract
Degradation of cellular material by autophagy is essential for cell survival and homeostasis, and requires intracellular transport of autophagosomes to encounter acidic lysosomes through unknown mechanisms. Here, we identify the PX-domain-containing kinesin Klp98A as a new regulator of autophagosome formation, transport and maturation in Drosophila. Depletion of Klp98A caused abnormal clustering of autophagosomes and lysosomes at the cell center and reduced the formation of starvation-induced autophagic vesicles. Reciprocally, overexpression of Klp98A redistributed autophagic vesicles towards the cell periphery. These effects were accompanied by reduced autophagosome-lysosome fusion and autophagic degradation. In contrast, depletion of the conventional kinesin heavy chain caused a similar mislocalization of autophagosomes without perturbing their fusion with lysosomes, indicating that vesicle fusion and localization are separable and independent events. Klp98A-mediated fusion required the endolysosomal GTPase Rab14, which interacted and colocalized with Klp98A, and required Klp98A for normal localization. Thus, Klp98A coordinates the movement and fusion of autophagic vesicles by regulating their positioning and interaction with the endolysosomal compartment.
Collapse
Affiliation(s)
- Caroline Mauvezin
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amanda L Neisch
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carlos I Ayala
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jung Kim
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Abigail Beltrame
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher R Braden
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Melissa K Gardner
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thomas S Hays
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology and Development, 6-160 Jackson Hall, 321 Church St. SE, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
31
|
Lincoln BL, Alabsi SH, Frendo N, Freund R, Keller LC. Drosophila Neuronal Injury Follows a Temporal Sequence of Cellular Events Leading to Degeneration at the Neuromuscular Junction. J Exp Neurosci 2015; 9:1-9. [PMID: 26512206 PMCID: PMC4612769 DOI: 10.4137/jen.s25516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 11/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide, and as the global population ages, there is a critical need to improve our understanding of the molecular and cellular mechanisms that drive neurodegeneration. At the molecular level, neurodegeneration involves the activation of complex signaling pathways that drive the active destruction of neurons and their intracellular components. Here, we use an in vivo motor neuron injury assay to acutely induce neurodegeneration in order to follow the temporal order of events that occur following injury in Drosophila melanogaster. We find that sites of injury can be rapidly identified based on structural defects to the neuronal cytoskeleton that result in disrupted axonal transport. Additionally, the neuromuscular junction accumulates ubiquitinated proteins prior to the neurodegenerative events, occurring at 24 hours post injury. Our data provide insights into the early molecular events that occur during axonal and neuromuscular degeneration in a genetically tractable model organism. Importantly, the mechanisms that mediate neurodegeneration in flies are conserved in humans. Thus, these studies have implications for our understanding of the cellular and molecular events that occur in humans and will facilitate the identification of biomedically relevant targets for future treatments.
Collapse
Affiliation(s)
- Barron L Lincoln
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Sahar H Alabsi
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Nicholas Frendo
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Robert Freund
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Lani C Keller
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| |
Collapse
|
32
|
Sosińska P, Mikuła-Pietrasik J, Książek K. The double-edged sword of long non-coding RNA: The role of human brain-specific BC200 RNA in translational control, neurodegenerative diseases, and cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 766:58-67. [DOI: 10.1016/j.mrrev.2015.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/29/2015] [Accepted: 08/28/2015] [Indexed: 12/14/2022]
|
33
|
Selmansberger M, Feuchtinger A, Zurnadzhy L, Michna A, Kaiser JC, Abend M, Brenner A, Bogdanova T, Walch A, Unger K, Zitzelsberger H, Hess J. CLIP2 as radiation biomarker in papillary thyroid carcinoma. Oncogene 2014; 34:3917-25. [DOI: 10.1038/onc.2014.311] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/16/2014] [Accepted: 08/09/2014] [Indexed: 12/16/2022]
|
34
|
Karplus M. Development of multiscale models for complex chemical systems: from H+H₂ to biomolecules (Nobel Lecture). Angew Chem Int Ed Engl 2014; 53:9992-10005. [PMID: 25066036 DOI: 10.1002/anie.201403924] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Martin Karplus
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA 02138 (USA); Laboratoire de Chimie Biophysique, ISIS, Université de Strasbourg, 67000 Strasbourg (France)
| |
Collapse
|
35
|
Atherton J, Farabella I, Yu IM, Rosenfeld SS, Houdusse A, Topf M, Moores CA. Conserved mechanisms of microtubule-stimulated ADP release, ATP binding, and force generation in transport kinesins. eLife 2014; 3:e03680. [PMID: 25209998 PMCID: PMC4358365 DOI: 10.7554/elife.03680] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/08/2014] [Indexed: 01/21/2023] Open
Abstract
Kinesins are a superfamily of microtubule-based ATP-powered motors, important for multiple, essential cellular functions. How microtubule binding stimulates their ATPase and controls force generation is not understood. To address this fundamental question, we visualized microtubule-bound kinesin-1 and kinesin-3 motor domains at multiple steps in their ATPase cycles—including their nucleotide-free states—at ∼7 Å resolution using cryo-electron microscopy. In both motors, microtubule binding promotes ordered conformations of conserved loops that stimulate ADP release, enhance microtubule affinity and prime the catalytic site for ATP binding. ATP binding causes only small shifts of these nucleotide-coordinating loops but induces large conformational changes elsewhere that allow force generation and neck linker docking towards the microtubule plus end. Family-specific differences across the kinesin–microtubule interface account for the distinctive properties of each motor. Our data thus provide evidence for a conserved ATP-driven mechanism for kinesins and reveal the critical mechanistic contribution of the microtubule interface. DOI:http://dx.doi.org/10.7554/eLife.03680.001 The interior of a cell is a hive of activity, filled with proteins and other items moving from one location to another. A network of filaments called microtubules forms tracks along which so-called motor proteins carry these items. Kinesins are one group of motor proteins, and a typical kinesin protein has one end (called the ‘motor domain’) that can attach itself to the microtubules. The other end links to the cargo being carried, and a ‘neck’ connects the two. When two of these proteins work together, flexible regions of the neck allow the two motor domains to move past one another, which enable the kinesin to essentially walk along a microtubule in a stepwise manner. To take these steps along microtubules, each kinesin motor domain in the pair must undergo alternating cycles of tight association and release from their tracks. This cycle is coordinated by binding and breaking down a molecule called ATP, which also provides the energy needed to take the next step. How the cycle of loose and tight microtubule attachment is coordinated with the release of the breakdown products of ATP, and how the energy from the ATP molecule is converted into the force that moves the motor along the microtubule, has been unclear. Atherton et al. use a technique called cryo-electron microscopy to study—in more detail than previously seen—the structure of the motor domains of two types of kinesin called kinesin-1 and kinesin-3. Images were taken at different stages of the cycle used by the motor domains to extract the energy from ATP molecules. Although the two kinesins have been thought to move along the microtubule tracks in different ways, Atherton et al. find that the core mechanism used by their motor domains is the same. When a motor domain binds to the microtubule, its shape changes, first stimulating release of the breakdown products of ATP from the previous cycle. This release makes room for a new ATP molecule to bind. The structural changes caused by ATP binding are relatively small but produce larger changes in the flexible neck region that enable individual motor domains within a kinesin pair to co-ordinate their movement and move in a consistent direction. This mechanism involves tight coupling between track binding and fuel usage and makes kinesins highly efficient motors. The structures uncovered by Atherton et al. reveal a mechanism that links microtubule binding, the energy supplied to the motor domain and the force that moves the kinesin along a microtubule. Future work will clarify whether the key features observed in the motor domains of kinesin-1 and kinesin-3 are also found in other types of kinesin motors. DOI:http://dx.doi.org/10.7554/eLife.03680.002
Collapse
Affiliation(s)
- Joseph Atherton
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, United Kingdom
| | - Irene Farabella
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, United Kingdom
| | - I-Mei Yu
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Paris, France
| | - Steven S Rosenfeld
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Paris, France
| | - Maya Topf
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, United Kingdom
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, United Kingdom
| |
Collapse
|
36
|
Karplus M. Entwicklung von Multiskalenmodellen für komplexe chemische Systeme: Von H+H2zu Biomolekülen (Nobel-Aufsatz). Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Willemsen MH, Ba W, Wissink-Lindhout WM, de Brouwer APM, Haas SA, Bienek M, Hu H, Vissers LELM, van Bokhoven H, Kalscheuer V, Nadif Kasri N, Kleefstra T. Involvement of the kinesin family members KIF4A and KIF5C in intellectual disability and synaptic function. J Med Genet 2014; 51:487-94. [PMID: 24812067 DOI: 10.1136/jmedgenet-2013-102182] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Kinesin superfamily (KIF) genes encode motor proteins that have fundamental roles in brain functioning, development, survival and plasticity by regulating the transport of cargo along microtubules within axons, dendrites and synapses. Mouse knockout studies support these important functions in the nervous system. The role of KIF genes in intellectual disability (ID) has so far received limited attention, although previous studies have suggested that many ID genes impinge on synaptic function. METHODS By applying next-generation sequencing (NGS) in ID patients, we identified likely pathogenic mutations in KIF4A and KIF5C. To further confirm the pathogenicity of these mutations, we performed functional studies at the level of synaptic function in primary rat hippocampal neurons. RESULTS AND CONCLUSIONS Four males from a single family with a disruptive mutation in the X-linked KIF4A (c.1489-8_1490delins10; p.?- exon skipping) showed mild to moderate ID and epilepsy. A female patient with a de novo missense mutation in KIF5C (c.11465A>C; p.(Glu237Lys)) presented with severe ID, epilepsy, microcephaly and cortical malformation. Knock-down of Kif4a in rat primary hippocampal neurons altered the balance between excitatory and inhibitory synaptic transmission, whereas the mutation in Kif5c affected its protein function at excitatory synapses. Our results suggest that mutations in KIF4A and KIF5C cause ID by tipping the balance between excitatory and inhibitory synaptic excitability.
Collapse
Affiliation(s)
- Marjolein H Willemsen
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands
| | - Wei Ba
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Arjan P M de Brouwer
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands
| | - Stefan A Haas
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Melanie Bienek
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Hao Hu
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lisenka E L M Vissers
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Vera Kalscheuer
- Department of Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands Department of Cognitive Neuroscience, Radboud university medical center, Nijmegen, The Netherlands Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands Nijmegen Centre for Molecular Life Sciences, Institute for Genetic and Metabolic Diseases, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
38
|
Jonczyk MS, Simon M, Kumar S, Fernandes VE, Sylvius N, Mallon AM, Denny P, Andrew PW. Genetic factors regulating lung vasculature and immune cell functions associate with resistance to pneumococcal infection. PLoS One 2014; 9:e89831. [PMID: 24594938 PMCID: PMC3940657 DOI: 10.1371/journal.pone.0089831] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023] Open
Abstract
Streptococcus pneumoniae is an important human pathogen responsible for high mortality and morbidity worldwide. The susceptibility to pneumococcal infections is controlled by as yet unknown genetic factors. To elucidate these factors could help to develop new medical treatments and tools to identify those most at risk. In recent years genome wide association studies (GWAS) in mice and humans have proved successful in identification of causal genes involved in many complex diseases for example diabetes, systemic lupus or cholesterol metabolism. In this study a GWAS approach was used to map genetic loci associated with susceptibility to pneumococcal infection in 26 inbred mouse strains. As a result four candidate QTLs were identified on chromosomes 7, 13, 18 and 19. Interestingly, the QTL on chromosome 7 was located within S. pneumoniae resistance QTL (Spir1) identified previously in a linkage study of BALB/cOlaHsd and CBA/CaOlaHsd F2 intercrosses. We showed that only a limited number of genes encoded within the QTLs carried phenotype-associated polymorphisms (22 genes out of several hundred located within the QTLs). These candidate genes are known to regulate TGFβ signalling, smooth muscle and immune cells functions. Interestingly, our pulmonary histopathology and gene expression data demonstrated, lung vasculature plays an important role in resistance to pneumococcal infection. Therefore we concluded that the cumulative effect of these candidate genes on vasculature and immune cells functions as contributory factors in the observed differences in susceptibility to pneumococcal infection. We also propose that TGFβ-mediated regulation of fibroblast differentiation plays an important role in development of invasive pneumococcal disease. Gene expression data submitted to the NCBI Gene Expression Omnibus Accession No: GSE49533 SNP data submitted to NCBI dbSNP Short Genetic Variation http://www.ncbi.nlm.nih.gov/projects/SNP/snp_viewTable.cgi?handle=MUSPNEUMONIA.
Collapse
Affiliation(s)
- Magda S. Jonczyk
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Michelle Simon
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Saumya Kumar
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Vitor E. Fernandes
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Nicolas Sylvius
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | | | - Paul Denny
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Peter W. Andrew
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Paus T, Pesaresi M, French L. White matter as a transport system. Neuroscience 2014; 276:117-25. [PMID: 24508743 DOI: 10.1016/j.neuroscience.2014.01.055] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/14/2014] [Accepted: 01/29/2014] [Indexed: 12/14/2022]
Abstract
There are two ways to picture white matter: as a grid of electrical wires or a network of roads. The first metaphor captures the classical function of an axon as conductor of action potentials (and information) from one brain region to another. The second one points to the important role of axons in a bi-directional transport of biological molecules and organelles between the cell body and synapse. Given the wide variety of such cargoes, a well-functioning axonal transport is critical for a number of processes, including neurotransmission, metabolism and viability of neurons. This selective review will emphasize the need for considering axonal transport when interpreting functional consequences of inter-individual variations in the structural properties of white matter. We start by describing the space occupied by white matter and techniques used in vivo for its characterization. We then provide examples of key features of maturation and aging of white matter, as well as some of the common abnormalities observed in neurodevelopmental and neurodegenerative disorders. Next, we review work that motivated our focus on axonal diameter, and explain the relationships between transport and cytoskeleton within the axon. We will conclude by describing molecular machinery of axonal transport and genes that may contribute to inter-individual variations in axonal diameter and axonal transport.
Collapse
Affiliation(s)
- T Paus
- Rotman Research Institute, University of Toronto, Toronto, Canada.
| | - M Pesaresi
- Rotman Research Institute, University of Toronto, Toronto, Canada
| | - L French
- Rotman Research Institute, University of Toronto, Toronto, Canada
| |
Collapse
|
40
|
Coles GL, Ackerman KG. Kif7 is required for the patterning and differentiation of the diaphragm in a model of syndromic congenital diaphragmatic hernia. Proc Natl Acad Sci U S A 2013; 110:E1898-905. [PMID: 23650387 PMCID: PMC3666741 DOI: 10.1073/pnas.1222797110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a common birth defect that results in a high degree of neonatal morbidity and mortality, but its pathological mechanisms are largely unknown. Therefore, we performed a forward genetic screen in mice to identify unique genes, models, and mechanisms of abnormal diaphragm development. We identified a mutant allele of kinesin family member 7 (Kif7), the disorganized diaphragm (dd). Embryos homozygous for the dd allele possess communicating diaphragmatic hernias, central tendon patterning defects, and increased cell proliferation with diaphragmatic tissue hyperplasia. Because the patterning of the central tendon is undescribed, we analyzed the expression of genes regulating tendonogenesis in dd/dd mutant embryos, and we determined that retinoic acid (RA) signaling was misregulautted. To further investigate the role of Kif7 and RA signaling in the development of the embryonic diaphragm, we established primary mesenchymal cultures of WT embryonic day 13.5 diaphragmatic cells. We determined that RA signaling is necessary for the expression of tendon markers as well as the expression of other CDH-associated genes. Knockdown of Kif7, and retinoic acid receptors alpha (Rara), beta (Rarb), and gamma (Rarg) indicated that RA signaling is dependent on these genes to promote tendonogenesis within the embryonic diaphragm. Taken together, our results provide evidence for a model in which inhibition of RA receptor signaling promotes CDH pathogenesis through a complex gene network.
Collapse
Affiliation(s)
| | - Kate G. Ackerman
- Department of Biomedical Genetics and
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
41
|
Gunawardena S. Nanoparticles in the Brain: A Potential Therapeutic System Targeted to an Early Defect Observed in Many Neurodegenerative Diseases. Pharm Res 2013; 30:2459-74. [DOI: 10.1007/s11095-013-1037-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 03/25/2013] [Indexed: 12/14/2022]
|
42
|
Patil H, Cho KI, Lee J, Yang Y, Orry A, Ferreira PA. Kinesin-1 and mitochondrial motility control by discrimination of structurally equivalent but distinct subdomains in Ran-GTP-binding domains of Ran-binding protein 2. Open Biol 2013; 3:120183. [PMID: 23536549 PMCID: PMC3718338 DOI: 10.1098/rsob.120183] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The pleckstrin homology (PH) domain is a versatile fold that mediates a variety of protein–protein and protein–phosphatidylinositol lipid interactions. The Ran-binding protein 2 (RanBP2) contains four interspersed Ran GTPase-binding domains (RBDn= 1–4) with close structural homology to the PH domain of Bruton's tyrosine kinase. The RBD2, kinesin-binding domain (KBD) and RBD3 comprise a tripartite domain (R2KR3) of RanBP2 that causes the unfolding, microtubule binding and biphasic activation of kinesin-1, a crucial anterograde motor of mitochondrial motility. However, the interplay between Ran GTPase and R2KR3 of RanBP2 in kinesin-1 activation and mitochondrial motility is elusive. We use structure–function, biochemical, kinetic and cell-based assays with time-lapse live-cell microscopy of over 260 000 mitochondrial-motility-related events to find mutually exclusive subdomains in RBD2 and RBD3 towards Ran GTPase binding, kinesin-1 activation and mitochondrial motility regulation. The RBD2 and RBD3 exhibit Ran-GTP-independent, subdomain and stereochemical-dependent discrimination on the biphasic kinetics of kinesin-1 activation or regulation of mitochondrial motility. Further, KBD alone and R2KR3 stimulate and suppress, respectively, multiple biophysical parameters of mitochondrial motility. The regulation of the bidirectional transport of mitochondria by either KBD or R2KR3 is highly coordinated, because their kinetic effects are accompanied always by changes in mitochondrial motile events of either transport polarity. These studies uncover novel roles in Ran GTPase-independent subdomains of RBD2 and RBD3, and KBD of RanBP2, that confer antagonizing and multi-modal mechanisms of kinesin-1 activation and regulation of mitochondrial motility. These findings open new venues towards the pharmacological harnessing of cooperative and competitive mechanisms regulating kinesins, RanBP2 or mitochondrial motility in disparate human disorders.
Collapse
Affiliation(s)
- Hemangi Patil
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
43
|
El-Nassan HB. Advances in the discovery of kinesin spindle protein (Eg5) inhibitors as antitumor agents. Eur J Med Chem 2013; 62:614-31. [PMID: 23434636 DOI: 10.1016/j.ejmech.2013.01.031] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 01/22/2013] [Accepted: 01/26/2013] [Indexed: 10/27/2022]
Abstract
Cancer is considered as one of the most serious health problems. Despite the presence of many effective chemotherapeutic agents, their severe side effects together with the appearance of mutant tumors limit the use of these drugs and increase the need for new anticancer agents. Eg5 represents an attractive target for medicinal chemists since Eg5 is overexpressed in many proliferative tissues while almost no Eg5 is detected in nonproliferative tissues. Many Eg5 inhibitors displayed potent anticancer activity against some of the mutant tumors with limited side effects. The present review provides an overview about the progress in the discovery of Eg5 inhibitors especially from 2009 to 2012 as well as the clinical trials conducted on some of these inhibitors.
Collapse
Affiliation(s)
- Hala Bakr El-Nassan
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, 33 Kasr El-Aini Street, Cairo 11562, Egypt.
| |
Collapse
|
44
|
Developing nanotherapies for neurodegenerative diseases: ORMOSIL and its potential in axonal transport. Ther Deliv 2012; 3:1189-98. [PMID: 23116011 DOI: 10.4155/tde.12.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In neurons, essential components packaged into vesicles are transported down microtubules to the ends of axons (synapses) where they are utilized. Components are also transported from the synapse to the cell body. This transport pathway is crucial for normal development, cell survival and plasticity. Recent work has established that defects in transport can contribute to the initiation of neurodegenerative disease, culminating in cell death and degeneration. Thus, delivering therapeutic treatments to an early defect is critical since many current strategies target pathology that occurs at later stages in the disease. Current treatments also affect the entire organism, causing side-effects that are often more deleterious than the disease. This article discusses how engineered synthetic structures can be used to directly target axonal transport--a pathway that is affected during the early stages of disease. Studies in this area will require the exchange of fundamental knowledge between biologists, chemists and engineers to effectively manufacture novel biomaterials for medical use.
Collapse
|
45
|
Nam W, Epureanu BI. The effects of viscoelastic fluid on kinesin transport. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2012; 24:375103. [PMID: 22842317 DOI: 10.1088/0953-8984/24/37/375103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Kinesins are molecular motors which transport various cargoes in the cytoplasm of cells and are involved in cell division. Previous models for kinesins have only targeted their in vitro motion. Thus, their applicability is limited to kinesin moving in a fluid with low viscosity. However, highly viscoelastic fluids have considerable effects on the movement of kinesin. For example, the high viscosity modifies the relation between the load and the speed of kinesin. While the velocity of kinesin has a nonlinear dependence with respect to the load in environments with low viscosity, highly viscous forces change that behavior. Also, the elastic nature of the fluid changes the velocity of kinesin. The new mechanistic model described in this paper considers the viscoelasticity of the fluid using subdiffusion. The approach is based on a generalized Langevin equation and fractional Brownian motion. Results show that a single kinesin has a maximum velocity when the ratio between the viscosity and elasticity is about 0.5. Additionally, the new model is able to capture the transient dynamics, which allows the prediction of the motion of kinesin under time varying loads.
Collapse
Affiliation(s)
- Woochul Nam
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109-2125, USA
| | | |
Collapse
|
46
|
Zhu H, Lee HY, Tong Y, Hong BS, Kim KP, Shen Y, Lim KJ, Mackenzie F, Tempel W, Park HW. Crystal structures of the tetratricopeptide repeat domains of kinesin light chains: insight into cargo recognition mechanisms. PLoS One 2012; 7:e33943. [PMID: 22470497 PMCID: PMC3314626 DOI: 10.1371/journal.pone.0033943] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 02/23/2012] [Indexed: 01/07/2023] Open
Abstract
Kinesin-1 transports various cargos along the axon by interacting with the cargos through its light chain subunit. Kinesin light chains (KLC) utilize its tetratricopeptide repeat (TPR) domain to interact with over 10 different cargos. Despite a high sequence identity between their TPR domains (87%), KLC1 and KLC2 isoforms exhibit differential binding properties towards some cargos. We determined the structures of human KLC1 and KLC2 tetratricopeptide repeat (TPR) domains using X-ray crystallography and investigated the different mechanisms by which KLCs interact with their cargos. Using isothermal titration calorimetry, we attributed the specific interaction between KLC1 and JNK-interacting protein 1 (JIP1) cargo to residue N343 in the fourth TRP repeat. Structurally, the N343 residue is adjacent to other asparagines and lysines, creating a positively charged polar patch within the groove of the TPR domain. Whereas, KLC2 with the corresponding residue S328 did not interact with JIP1. Based on these finding, we propose that N343 of KLC1 can form "a carboxylate clamp" with its neighboring asparagine to interact with JIP1, similar to that of HSP70/HSP90 organizing protein-1's (HOP1) interaction with heat shock proteins. For the binding of cargos shared by KLC1 and KLC2, we propose a different site located within the groove but not involving N343. We further propose a third binding site on KLC1 which involves a stretch of polar residues along the inter-TPR loops that may form a network of hydrogen bonds to JIP3 and JIP4. Together, these results provide structural insights into possible mechanisms of interaction between KLC TPR domains and various cargo proteins.
Collapse
Affiliation(s)
- Haizhong Zhu
- Structural Genomics Consortium, Toronto, Ontario, Canada
| | - Han Youl Lee
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| | - Yufeng Tong
- Structural Genomics Consortium, Toronto, Ontario, Canada
| | - Bum-Soo Hong
- Structural Genomics Consortium, Toronto, Ontario, Canada
| | - Kyung-Phil Kim
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| | - Yang Shen
- Structural Genomics Consortium, Toronto, Ontario, Canada
| | - Kyung Jik Lim
- Philip Pocock Catholic Secondary School, Mississauga, Ontario, Canada
| | | | - Wolfram Tempel
- Structural Genomics Consortium, Toronto, Ontario, Canada
| | - Hee-Won Park
- Structural Genomics Consortium, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
47
|
Klejnot M, Kozielski F. Structural insights into human Kif7, a kinesin involved in Hedgehog signalling. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2012; 68:154-9. [PMID: 22281744 PMCID: PMC3266853 DOI: 10.1107/s0907444911053042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 12/09/2011] [Indexed: 02/16/2023]
Abstract
Kif7, a member of the kinesin 4 superfamily, is implicated in a variety of diseases including Joubert, hydrolethalus and acrocallosal syndromes. It is also involved in primary cilium formation and the Hedgehog signalling pathway and may play a role in cancer. Its activity is crucial for embryonic development. Kif7 and Kif27, a closely related kinesin in the same subfamily, are orthologues of the Drosophila melanogaster kinesin-like protein Costal-2 (Cos2). In vertebrates, they work together to fulfil the role of the single Cos2 gene in Drosophila. Here, the high-resolution structure of the human Kif7 motor domain is reported and is compared with that of conventional kinesin, the founding member of the kinesin superfamily. These data are a first step towards structural characterization of a kinesin-4 family member and of this interesting molecular motor of medical significance.
Collapse
Affiliation(s)
- Marta Klejnot
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, Scotland
| | - Frank Kozielski
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow G61 1BD, Scotland
| |
Collapse
|
48
|
Strunze S, Engelke MF, Wang IH, Puntener D, Boucke K, Schleich S, Way M, Schoenenberger P, Burckhardt CJ, Greber UF. Kinesin-1-mediated capsid disassembly and disruption of the nuclear pore complex promote virus infection. Cell Host Microbe 2012; 10:210-23. [PMID: 21925109 DOI: 10.1016/j.chom.2011.08.010] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 03/23/2011] [Accepted: 08/01/2011] [Indexed: 01/01/2023]
Abstract
Many viruses deliver their genomes into the host cell nucleus for replication. However, the size restrictions of the nuclear pore complex (NPC), which regulates the passage of proteins, nucleic acids, and solutes through the nuclear envelope, require virus capsid uncoating before viral DNA can access the nucleus. We report a microtubule motor kinesin-1-mediated and NPC-supported mechanism of adenovirus uncoating. The capsid binds to the NPC filament protein Nup214 and kinesin-1 light-chain Klc1/2. The nucleoporin Nup358, which is bound to Nup214/Nup88, interacts with the kinesin-1 heavy-chain Kif5c to indirectly link the capsid to the kinesin motor. Kinesin-1 disrupts capsids docked at Nup214, which compromises the NPC and dislocates nucleoporins and capsid fragments into the cytoplasm. NPC disruption increases nuclear envelope permeability as indicated by the nuclear influx of large cytoplasmic dextran polymers. Thus, kinesin-1 uncoats viral DNA and compromises NPC integrity, allowing viral genomes nuclear access to promote infection.
Collapse
Affiliation(s)
- Sten Strunze
- Institute of Molecular Life Sciences, University of Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
van den Berg R, Hoogenraad CC. Molecular motors in cargo trafficking and synapse assembly. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:173-96. [PMID: 22351056 DOI: 10.1007/978-3-7091-0932-8_8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Every production process, be it cellular or industrial, depends on a constant supply of energy and resources. Synapses, specialized junctions in the central nervous system through which neurons signal to each other, are no exception to this rule. In order to form new synapses and alter the strength of synaptic transmission, neurons need a regulatory mechanism to deliver and remove synaptic proteins at synaptic sites. Neurons make use of active transport driven by molecular motor proteins to move synaptic cargo over either microtubules (kinesin, dynein) or actin filaments (myosin) to their specific site of action. These mechanisms are crucial for the initial establishment of synaptic specializations during synaptogenesis and for activity-dependent changes in synaptic strength during plasticity. In this chapter, we address the organization of the neuronal cytoskeleton, focus on synaptic cargo transport activities that operate in axons and dendrites, and discuss the spatial and temporal regulation of motor protein-based transport.
Collapse
Affiliation(s)
- Robert van den Berg
- Cell Biology, Utrecht University, Padualaan 8, 3584CH, Utrecht, The Netherlands
| | | |
Collapse
|
50
|
Molecular characterization of a KIF3A-like kinesin gene in the testis of the Chinese fire-bellied newt Cynops orientalis. Mol Biol Rep 2011; 39:4207-14. [PMID: 21773941 DOI: 10.1007/s11033-011-1206-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/11/2011] [Indexed: 10/18/2022]
Abstract
KIF3A, the subunit within the kinesin-2 superfamily, is a typically N-terminal motor protein, which is involved in membranous organelle and intraflagellar transport. During spermatogenesis, KIF3A plays a critical role in the formation of flagella and cilia. KIF3A is also related to the left-right asymmetry, the signal pathway, DNA damage and tumorigenesis. We used RT-PCR and in situ hybridization to clone the kif3a gene, and we identified its function in the testis of the Chinese fire-bellied newt Cynops orientalis (termed as co-kif3a). The full-length sequence of co-kif3a was 2193 bp, containing a 56 bp 5'UTR, 2073 bp ORF encoding a protein of 691 amino acids and a 64 bp 3'UTR. The secondary structure analysis showed that co-KIF3A had three motor domains, representing the N-terminal motor domain (1-400 aa), α-helix domain (400-600 aa) and C-terminal tail domain (600-691 aa). The amino acid sequence of co-KIF3A shared an identity of 55.9%, 90.9%, 89.9%, 91.3% and 85.7% with its counterparts in Aedes aegypti, Mus musculus, Xenopus tropicalis, Homo sapiens and Danio rerio, respectively. The calculated molecular weight of the putative co-KIF3A was 79 kDa and its estimated isoelectric point was 6.8. RT-PCR result showed that co-kif3a was expressed in several examined tissues, with a high level in the testis and low levels in liver, muscle and ovum. Kif3a was weakly expressed in the heart and spleen, and barely detected in the intestine. In situ hybridization analysis demonstrated that in early spermatid co-kif3a was expressed around the nuclear membrane. When the tail began to emerge in the middle spermatid, mRNA transcript was abundantly concentrated in the flagellum. The mRNA signal was still very strong along all the flagellum in late spermatid. In mature spermatid, the message was weak. Therefore, co-KIF3A probably plays a functional role in the spermiogenesis of C. orientalis.
Collapse
|