101
|
Wu S, Li H, Wang L, Mak N, Wu X, Ge R, Sun F, Cheng CY. Motor Proteins and Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:131-159. [PMID: 34453735 DOI: 10.1007/978-3-030-77779-1_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Unlike the intermediate filament- and septin-based cytoskeletons which are apolar structures, the microtubule (MT) and actin cytoskeletons are polarized structures in mammalian cells and tissues including the testis, most notable in Sertoli cells. In the testis, these cytoskeletons that stretch across the epithelium of seminiferous tubules and lay perpendicular to the basement membrane of tunica propria serve as tracks for corresponding motor proteins to support cellular cargo transport. These cargoes include residual bodies, phagosomes, endocytic vesicles and most notably developing spermatocytes and haploid spermatids which lack the ultrastructures of motile cells (e.g., lamellipodia, filopodia). As such, these developing germ cells require the corresponding motor proteins to facilitate their transport across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Due to the polarized natures of these cytoskeletons with distinctive plus (+) and minus (-) end, directional cargo transport can take place based on the use of corresponding actin- or MT-based motor proteins. These include the MT-based minus (-) end directed motor proteins: dyneins, and the plus (+) end directed motor proteins: kinesins, as well as the actin-based motor proteins: myosins, many of which are plus (+) end directed but a few are also minus (-) end directed motor proteins. Recent studies have shown that these motor proteins are essential to support spermatogenesis. In this review, we briefly summarize and evaluate these recent findings so that this information will serve as a helpful guide for future studies and for planning functional experiments to better understand their role mechanistically in supporting spermatogenesis.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Huitao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Nathan Mak
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA
| | - Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Zhejiang, China
| | - Fei Sun
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - C Yan Cheng
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
102
|
Zaniewski TM, Gicking AM, Fricks J, Hancock WO. A kinetic dissection of the fast and superprocessive kinesin-3 KIF1A reveals a predominant one-head-bound state during its chemomechanical cycle. J Biol Chem 2020; 295:17889-17903. [PMID: 33082143 PMCID: PMC7939386 DOI: 10.1074/jbc.ra120.014961] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
The kinesin-3 family contains the fastest and most processive motors of the three neuronal transport kinesin families, yet the sequence of states and rates of kinetic transitions that comprise the chemomechanical cycle and give rise to their unique properties are poorly understood. We used stopped-flow fluorescence spectroscopy and single-molecule motility assays to delineate the chemomechanical cycle of the kinesin-3, KIF1A. Our bacterially expressed KIF1A construct, dimerized via a kinesin-1 coiled-coil, exhibits fast velocity and superprocessivity behavior similar to WT KIF1A. We established that the KIF1A forward step is triggered by hydrolysis of ATP and not by ATP binding, meaning that KIF1A follows the same chemomechanical cycle as established for kinesin-1 and -2. The ATP-triggered half-site release rate of KIF1A was similar to the stepping rate, indicating that during stepping, rear-head detachment is an order of magnitude faster than in kinesin-1 and kinesin-2. Thus, KIF1A spends the majority of its hydrolysis cycle in a one-head-bound state. Both the ADP off-rate and the ATP on-rate at physiological ATP concentration were fast, eliminating these steps as possible rate-limiting transitions. Based on the measured run length and the relatively slow off-rate in ADP, we conclude that attachment of the tethered head is the rate-limiting transition in the KIF1A stepping cycle. Thus, KIF1A's activity can be explained by a fast rear-head detachment rate, a rate-limiting step of tethered-head attachment that follows ATP hydrolysis, and a relatively strong electrostatic interaction with the microtubule in the weakly bound post-hydrolysis state.
Collapse
Affiliation(s)
- Taylor M Zaniewski
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Allison M Gicking
- Department of Biomedical Engineering and Bioengineering Graduate Program, Pennsylvania State University, University Park, Pennsylvania, USA
| | - John Fricks
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, Arizona, USA
| | - William O Hancock
- Department of Biomedical Engineering and Bioengineering Graduate Program, Pennsylvania State University, University Park, Pennsylvania, USA.
| |
Collapse
|
103
|
Jung M, Kim D, Mun JY. Direct Visualization of Actin Filaments and Actin-Binding Proteins in Neuronal Cells. Front Cell Dev Biol 2020; 8:588556. [PMID: 33324645 PMCID: PMC7726226 DOI: 10.3389/fcell.2020.588556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
Actin networks and actin-binding proteins (ABPs) are most abundant in the cytoskeleton of neurons. The function of ABPs in neurons is nucleation of actin polymerization, polymerization or depolymerization regulation, bundling of actin through crosslinking or stabilization, cargo movement along actin filaments, and anchoring of actin to other cellular components. In axons, ABP–actin interaction forms a dynamic, deep actin network, which regulates axon extension, guidance, axon branches, and synaptic structures. In dendrites, actin and ABPs are related to filopodia attenuation, spine formation, and synapse plasticity. ABP phosphorylation or mutation changes ABP–actin binding, which regulates axon or dendritic plasticity. In addition, hyperactive ABPs might also be expressed as aggregates of abnormal proteins in neurodegeneration. Those changes cause many neurological disorders. Here, we will review direct visualization of ABP and actin using various electron microscopy (EM) techniques, super resolution microscopy (SRM), and correlative light and electron microscopy (CLEM) with discussion of important ABPs in neuron.
Collapse
Affiliation(s)
- Minkyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Doory Kim
- Department of Chemistry, Research Institute for Convergence of Basic Sciences, Institute of Nano Science and Technology, Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
104
|
Zhao F, Feng Y, Zhang X, Liu X, Li A. Kinesin Superfamily Member 18B (KIF18B) Promotes Cell Proliferation in Colon Adenocarcinoma. Cancer Manag Res 2020; 12:12769-12778. [PMID: 33335427 PMCID: PMC7737937 DOI: 10.2147/cmar.s261894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/20/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The role of kinesin superfamily proteins (KIFs) has been reported in a variety of tumors and KIFs contributed to the proliferation of cancer cells. But few studies were focus on colon adenocarcinoma. METHODS Through bioinformatics analysis and immunohistochemistry (IHC) assays, the expression of KIF18B in colon adenocarcinoma tissues was determined. Stable KIF18B-depleted cell lines were constructed using lentivirus-mediated shRNA of KIF18B. Cell colony formation assay and CCK8 assay were performed to assess cell proliferation degree, and the expression level of KI67 and PCNA was used to indicate cell proliferation in vitro and verified using xenograft tumors in vivo. RESULTS KIF18B is highly expressed in colon adenocarcinoma tissues and has a negative correlation with the prognosis and tumor grade of colon adenocarcinoma. Interfering with KIF18B inhibits cell proliferation in vitro and in vivo. CONCLUSION KIF18B can be used as a prognostic marker for colon adenocarcinoma and may be a therapeutic target for colon adenocarcinoma treatment.
Collapse
Affiliation(s)
- Fei Zhao
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Yunzhang Feng
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Xueqiang Zhang
- HanDan Central Hospital President's Office, HanDan Central Hospital, Heibei, People's Republic of China
| | - Xiaohui Liu
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Aili Li
- Department of Gynaecology, HanDan Central Hospital, Heibei, People’s Republic of China
| |
Collapse
|
105
|
Yang Y, Liu X, Li R, Zhang M, Wang H, Qu Y. Kinesin family member 3A inhibits the carcinogenesis of non-small cell lung cancer and prolongs survival. Oncol Lett 2020; 20:348. [PMID: 33123259 PMCID: PMC7586287 DOI: 10.3892/ol.2020.12211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/21/2020] [Indexed: 12/27/2022] Open
Abstract
Kinesin family member 3A (KIF3A) plays a crucial role in the carcinogenesis of different types of human cancer. The present study aimed to identify the role of KIF3A in the carcinogenesis of non-small cell lung cancer (NSCLC). KIF3A protein expression was determined in 163 patients with NSCLC using immunohistochemistry staining. The prognosis of patients with NSCLC was determined using Kaplan-Meier survival and Cox regression analyses. The function of KIF3A on the carcinogenesis and metastasis of NSCLC was determined in vitro. Furthermore, a protein-protein interaction (PPI) network of KIF3A was constructed and the potential interacting molecules were identified using bioinformatic analysis. The protein expression levels of KIF3A were significantly lower in the NSCLC tissues compared with that in the adjacent tissues, and low KIF3A expression level was associated with unfavorable survival outcomes in patients with NSCLC. Furthermore, KIF3A knockdown increased proliferation, invasion and metastasis, and inhibited apoptosis of NSCLC cells. KIF3A was demonstrated to interact with intraflagellar transport 57 (IFT57) in the PPI network. In addition, validation analyses indicated that KIF3A mRNA expression levels were positively correlated with IFT57 mRNA expression levels in clinical NSCLC samples and NSCLC cell lines. Taken together, the results of the present study suggested that KIF3A is a key tumor suppressor gene for carcinogenesis and metastasis of NSCLC, it may also function as a biomarker and interacts with IFT57 in the progression of NSCLC.
Collapse
Affiliation(s)
- Yie Yang
- Department of Clinical Laboratory, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250012, P.R. China
| | - Xiao Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mengyu Zhang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hong Wang
- Department of Thoracic Surgery, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250012, P.R. China
| | - Yiqing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
106
|
Huang C, Hu CG, Ning ZK, Huang J, Zhu ZM. Identification of key genes controlling cancer stem cell characteristics in gastric cancer. World J Gastrointest Surg 2020; 12:442-459. [PMID: 33304447 PMCID: PMC7701879 DOI: 10.4240/wjgs.v12.i11.442] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/13/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Self-renewal of gastric cancer stem cells (GCSCs) is considered to be the underlying cause of the metastasis, drug resistance, and recurrence of gastric cancer (GC).
AIM To characterize the expression of stem cell-related genes in GC.
METHODS RNA sequencing results and clinical data for gastric adenoma and adenocarcinoma samples were obtained from The Cancer Genome Atlas database, and the results of the GC mRNA expression-based stemness index (mRNAsi) were analyzed. Weighted gene coexpression network analysis was then used to find modules of interest and their key genes. Survival analysis of key genes was performed using the online tool Kaplan-Meier Plotter, and the online database Oncomine was used to assess the expression of key genes in GC.
RESULTS mRNAsi was significantly upregulated in GC tissues compared to normal gastric tissues (P < 0.0001). A total of 16 modules were obtained from the gene coexpression network; the brown module was most positively correlated with mRNAsi. Sixteen key genes (BUB1, BUB1B, NCAPH, KIF14, RACGAP1, RAD54L, TPX2, KIF15, KIF18B, CENPF, TTK, KIF4A, SGOL2, PLK4, XRCC2, and C1orf112) were identified in the brown module. The functional and pathway enrichment analyses showed that the key genes were significantly enriched in the spindle cellular component, the sister chromatid segregation biological process, the motor activity molecular function, and the cell cycle and homologous recombination pathways. Survival analysis and Oncomine analysis revealed that the prognosis of patients with GC and the expression of three genes (RAD54L, TPX2, and XRCC2) were consistently related.
CONCLUSION Sixteen key genes are primarily associated with stem cell self-renewal and cell proliferation characteristics. RAD54L, TPX2, and XRCC2 are the most likely therapeutic targets for inhibiting the stemness characteristics of GC cells.
Collapse
Affiliation(s)
- Chao Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ce-Gui Hu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zhi-Kun Ning
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zheng-Ming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
107
|
Joseph NF, Grinman E, Swarnkar S, Puthanveettil SV. Molecular Motor KIF3B Acts as a Key Regulator of Dendritic Architecture in Cortical Neurons. Front Cell Neurosci 2020; 14:521199. [PMID: 33192305 PMCID: PMC7604319 DOI: 10.3389/fncel.2020.521199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/09/2020] [Indexed: 01/08/2023] Open
Abstract
Neurons require a well-coordinated intercellular transport system to maintain their normal cellular function and morphology. The kinesin family of proteins (KIFs) fills this role by regulating the transport of a diverse array of cargos in post-mitotic cells. On the other hand, in mitotic cells, KIFs facilitate the fidelity of the cellular division machinery. Though certain mitotic KIFs function in post-mitotic neurons, little is known about them. We studied the role of a mitotic KIF (KIF3B) in neuronal architecture. We find that the RNAi mediated knockdown of KIF3B in primary cortical neurons resulted in an increase in spine density; the number of thin and mushroom spines; and dendritic branching. Consistent with the change in spine density, we observed a specific increase in the distribution of the excitatory post-synaptic protein, PSD-95 in KIF3B knockdown neurons. Interestingly, overexpression of KIF3B produced a reduction in spine density, in particular mushroom spines, and a decrease in dendritic branching. These studies suggest that KIF3B is a key determinant of cortical neuron morphology and that it functions as an inhibitory constraint on structural plasticity, further illuminating the significance of mitotic KIFs in post-mitotic neurons.
Collapse
Affiliation(s)
- Nadine F Joseph
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, United States
| | - Eddie Grinman
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, United States
| | - Supriya Swarnkar
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, United States
| | | |
Collapse
|
108
|
Azobenzene Based Photo-responsive Mechanical Actuator Fabricated by Intermolecular H-bond Interaction. CHINESE JOURNAL OF POLYMER SCIENCE 2020. [DOI: 10.1007/s10118-021-2504-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
109
|
Kitagawa A, Masuda T, Takahashi J, Tobo T, Noda M, Kuroda Y, Hu Q, Kouyama Y, Kobayashi Y, Kuramitsu S, Sato K, Fujii A, Yoshikawa Y, Wakiyama H, Shimizu D, Tsuruda Y, Eguchi H, Doki Y, Mori M, Mimori K. KIF15 Expression in Tumor-associated Monocytes Is a Prognostic Biomarker in Hepatocellular Carcinoma. Cancer Genomics Proteomics 2020; 17:141-149. [PMID: 32108036 DOI: 10.21873/cgp.20174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/01/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIM Kinesin family member 15 (KIF15) participates in the transport of macromolecules in essential cellular processes. In this study we evaluated the clinical relevance of KIF15 expression in hepatocellular carcinoma (HCC). MATERIALS AND METHODS Association between KIF15 expression and clinical outcomes in HCC patients was analyzed using three independent cohorts. Localization of KIF15 expression was assessed by immunohistochemical analysis. Co-culture experiments were performed using healthy donor peripheral blood mononuclear cells (PBMC) and HCC cell lines. RESULTS Immunohistochemical analysis showed that KIF15 was mainly expressed in inflammatory monocytes around cancer cells. Multivariate analysis indicated high KIF15 expression was an independent poor prognostic factor for survival. HCC cells with high expression of minichromosome maintenance protein 2 (MCM2) were located close to KIF15-expressing inflammatory monocytes. The proliferation ability of HCC cells was increased by co-culture with PBMC. CONCLUSION High KIF15 expression in inflammatory monocytes in tumor tissues may serve as a prognostic marker for poor outcome in HCC.
Collapse
Affiliation(s)
- Akihiro Kitagawa
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Junichi Takahashi
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Taro Tobo
- Department of Clinical Laboratory Medicine, Kyushu University, Beppu Hospital, Oita, Japan
| | - Miwa Noda
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Yosuke Kuroda
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Qingjiang Hu
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Yuta Kouyama
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Yuta Kobayashi
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shotaro Kuramitsu
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Kuniaki Sato
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Atsushi Fujii
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Yukihiro Yoshikawa
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroaki Wakiyama
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Dai Shimizu
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Yusuke Tsuruda
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University, Beppu Hospital, Oita, Japan
| |
Collapse
|
110
|
Li W, Zhang S, Yang G. Dynamic organization of intracellular organelle networks. WIREs Mech Dis 2020; 13:e1505. [PMID: 32865347 DOI: 10.1002/wsbm.1505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/06/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Intracellular organelles are membrane-bound and biochemically distinct compartments constructed to serve specialized functions in eukaryotic cells. Through extensive interactions, they form networks to coordinate and integrate their specialized functions for cell physiology. A fundamental property of these organelle networks is that they constantly undergo dynamic organization via membrane fusion and fission to remodel their internal connections and to mediate direct material exchange between compartments. The dynamic organization not only enables them to serve critical physiological functions adaptively but also differentiates them from many other biological networks such as gene regulatory networks and cell signaling networks. This review examines this fundamental property of the organelle networks from a systems point of view. The focus is exclusively on homotypic networks formed by mitochondria, lysosomes, endosomes, and the endoplasmic reticulum, respectively. First, key mechanisms that drive the dynamic organization of these networks are summarized. Then, several distinct organizational properties of these networks are highlighted. Next, spatial properties of the dynamic organization of these networks are emphasized, and their functional implications are examined. Finally, some representative molecular machineries that mediate the dynamic organization of these networks are surveyed. Overall, the dynamic organization of intracellular organelle networks is emerging as a fundamental and unifying paradigm in the internal organization of eukaryotic cells. This article is categorized under: Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Shuhao Zhang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA.,Department of Computational Biology, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
111
|
KIF15 contributes to cell proliferation and migration in breast cancer. Hum Cell 2020; 33:1218-1228. [PMID: 32578050 DOI: 10.1007/s13577-020-00392-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/11/2020] [Indexed: 01/14/2023]
Abstract
A number of kinesin proteins (KIFs) have been implicated in the development of multiple cancers. However, little is known about the expression and function of KIF15 in human breast cancer. Herein, we detected KIF15 expression in breast cancer tissues and paired adjacent normal tissues using immunohistochemistry and quantitative real-time polymerase chain reaction analysis, and the correlation of KIF15 expression with clinicopathological parameters was evaluated statistically. The role of KIF15 in cell proliferation, migration, tumor growth and metastasis of breast cancer cells was investigated in vitro and in vivo, and we explored potential molecular mechanisms underlying the effects of KIF15 in breast cancer through western blot analysis. The results revealed that increased KIF15 expression in breast cancer tissues were positively related with tumor size, lymph node metastasis and TNM stage, and higher KIF15 expression predicts a worse prognosis of patients with breast cancer. Furthermore, KIF15 knockdown markedly attenuated breast cancer cell proliferation, migration, tumor growth and metastasis in vitro and in vivo, and silenced KIF15 expression significantly inhibited the expression of phosphorylated AKT, phosphorylated JNK, and cyclin D1, while both p53 and p21 protein expressions were strongly enhanced. These results suggest that KIF15 is a potential oncogene in human breast cancer.
Collapse
|
112
|
Sheng J, Xue X, Jiang K. Knockdown of Kinase Family 15 Inhibits Cancer Cell Proliferation In vitro and its Clinical Relevance in Triple-Negative Breast Cancer. Curr Mol Med 2020; 19:147-155. [PMID: 30854965 DOI: 10.2174/1566524019666190308122108] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE Breast cancer is the most prevalent malignancy and the leading cause of death among women. Triple-negative breast cancer (TNBC) is a subtype of breast cancer and shows a distinctly aggressive nature with higher rates of relapse and shorter overall survival in the metastatic setting compared to other subtypes of breast cancer. This study aimed to assess the effect of KIF15 on various clinicopathological characteristics, survival analysis, and cell proliferation in triple-negative breast cancer, which has not been reported to our knowledge. METHODS A total of 165 patients with triple-negative breast cancer were enrolled and clinical data were obtained, Mann-Whitney U analysis was performed to assess the correlation between the expression of KIF15 and clinical pathological characteristics of TNBC patients. Survival analysis was performed by Kaplan-Meier analysis and Log-rank test. The expression levels of KIF15 in cancer tissues and adjacent tissues were evaluated via Sign test. Lentivirus was used to down-regulate the expression of KIF15 in TNBC cells. The cell proliferation, colony formation capacity and apoptosis were examined by MTT, Giemsa staining and flow cytometry assay, respectively. RESULTS Our results showed that, among the 165 TNBC patients, the expression of KIF15 was positive correlation with clinicopathological features of TNBC. In addition, KIF15 low-expression group showed higher disease-free survival than KIF15 highexpression group and univariate analysis showed that KIF15 high-expression group appeared higher mortality than KIF low-expression group (P ≤ 0.05). Meanwhile, the expression levels of KIF15 in cancer tissue notably up-regulated in comparison with adjacent tissue. In vitro, knockdown of KIF15 significantly promoted cell apoptosis and suppressed cell proliferation and colony formation of TNBC cells. CONCLUSION By utilizing survival analysis, we found that high-expression of KIF15 in the TNBC samples were associated with poorer overall survival, while the anti-tumor effect of KIF15 knockdown was also confirmed at the cellular level in vitro. Taken together, KIF15 can be applied as a potential diagnostic and therapeutic target in TNBC.
Collapse
Affiliation(s)
- Jiayu Sheng
- Department of Breast Diseases, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Xiaohong Xue
- Department of Breast Diseases, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| | - Ke Jiang
- Department of Breast Diseases, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, China
| |
Collapse
|
113
|
Zhang SF, Zhang K, Cheng HM, Lin L, Wang DZ. Comparative transcriptomics reveals colony formation mechanism of a harmful algal bloom species Phaeocystis globosa. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 719:137454. [PMID: 32114233 DOI: 10.1016/j.scitotenv.2020.137454] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
Phaeocystis globosa is a major causative agent of harmful algal blooms in the global ocean, featuring a complex polymorphic life cycle alternating between free-living solitary cells and colonial cells. Colony is the dominant morphotype during P. globosa bloom. However, the underlying mechanism of colony formation is poorly understood. Here, we comprehensively compared global transcriptomes of P. globosa cells at four distinctive colony formation stages: free-living solitary cells, two cell-, four cell- and multi-cell colonies, under low (20 °C) and high (32 °C) temperatures, and characterized the genes involved in colony formation. Glycosaminoglycan (GAG) synthesis was enhanced while its degradation was decreased during colony formation, resulting in the accumulation of GAGs that are an essential substrate of the colony matrix. Nitrogen metabolism and glutamine synthesis were remarkably increased in the colonial cells, which provided precursors for GAG synthesis. Furthermore, cell defense and motility were down-regulated in the colonial cells, thereby conserving energy for GAG synthesis. Notably, high temperature led to decreased synthesis and increased degradation of GAGs, resulting in insufficient substrates to form the colony. Our study indicates that GAGs accumulation is critical for colony formation of P. globosa, but high temperature inhibits GAGs' accumulation and colony formation.
Collapse
Affiliation(s)
- Shu-Feng Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Kun Zhang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Hua-Min Cheng
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Lin Lin
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China.
| | - Da-Zhi Wang
- State Key Laboratory of Marine Environmental Science/College of the Environment and Ecology, Xiamen University, Xiamen 361102, China; Key Laboratory of Marine Ecology & Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
114
|
Baek JH, Yun HS, Kim JY, Lee J, Lee YJ, Lee CW, Song JY, Ahn J, Park JK, Kim JS, Lee KH, Kim EH, Hwang SG. Kinesin light chain 4 as a new target for lung cancer chemoresistance via targeted inhibition of checkpoint kinases in the DNA repair network. Cell Death Dis 2020; 11:398. [PMID: 32457423 PMCID: PMC7250887 DOI: 10.1038/s41419-020-2592-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
The poor therapeutic efficacy of non-small cell lung cancer (NSCLC) is partly attributed to the acquisition of chemoresistance. To investigate the mechanism underlying this resistance, we examined the potential link between kinesin light chain 4 (KLC4), which we have previously reported to be associated with radioresistance in NSCLC, and sensitivity to chemotherapy in human lung cancer cell lines. KLC4 protein levels in lung cancer cells correlated with the degree of chemoresistance to cisplatin treatment. Furthermore, KLC4 silencing enhanced the cytotoxic effect of cisplatin by promoting DNA double-strand breaks and apoptosis. These effects were mediated by interaction with the checkpoint kinase CHK2, as KLC4 knockdown increased CHK2 activation, which was further enhanced in combination with cisplatin treatment. In addition, KLC4 and CHEK2 expression levels showed negative correlation in lung tumor samples from patients, and KLC4 overexpression correlated negatively with survival. Our results indicate a novel link between the KLC4 and CHK2 pathways regulating DNA damage response in chemoresistance, and highlight KLC4 as a candidate for developing lung cancer-specific drugs and customized targeted molecular therapy.
Collapse
Affiliation(s)
- Jeong-Hwa Baek
- Radiation Biology Research Team, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, Republic of Korea
| | - Hong Shik Yun
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ju-Young Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Janet Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Yeon-Joo Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 440-746, Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jong Kuk Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Kee-Ho Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, 33, 17-gil, Duryugongwon-ro, Nam-gu, Daegu, Korea.
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Korea.
| |
Collapse
|
115
|
Mutations in the KIF21B kinesin gene cause neurodevelopmental disorders through imbalanced canonical motor activity. Nat Commun 2020; 11:2441. [PMID: 32415109 PMCID: PMC7229210 DOI: 10.1038/s41467-020-16294-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 04/26/2020] [Indexed: 01/08/2023] Open
Abstract
KIF21B is a kinesin protein that promotes intracellular transport and controls microtubule dynamics. We report three missense variants and one duplication in KIF21B in individuals with neurodevelopmental disorders associated with brain malformations, including corpus callosum agenesis (ACC) and microcephaly. We demonstrate, in vivo, that the expression of KIF21B missense variants specifically recapitulates patients’ neurodevelopmental abnormalities, including microcephaly and reduced intra- and inter-hemispheric connectivity. We establish that missense KIF21B variants impede neuronal migration through attenuation of kinesin autoinhibition leading to aberrant KIF21B motility activity. We also show that the ACC-related KIF21B variant independently perturbs axonal growth and ipsilateral axon branching through two distinct mechanisms, both leading to deregulation of canonical kinesin motor activity. The duplication introduces a premature termination codon leading to nonsense-mediated mRNA decay. Although we demonstrate that Kif21b haploinsufficiency leads to an impaired neuronal positioning, the duplication variant might not be pathogenic. Altogether, our data indicate that impaired KIF21B autoregulation and function play a critical role in the pathogenicity of human neurodevelopmental disorder. Kinesins regulate intracellular transport and microtubule dynamics. Here, the authors show that KIF21B variants in humans associate with corpus callosum agenesis and microcephaly. Using mice and zebrafish, they showed the cellular mechanisms altered by the missense KIF21B variants.
Collapse
|
116
|
Sinawang G, Osaki M, Takashima Y, Yamaguchi H, Harada A. Biofunctional hydrogels based on host–guest interactions. Polym J 2020. [DOI: 10.1038/s41428-020-0352-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
117
|
Graybill PM, Davalos RV. Cytoskeletal Disruption after Electroporation and Its Significance to Pulsed Electric Field Therapies. Cancers (Basel) 2020; 12:E1132. [PMID: 32366043 PMCID: PMC7281591 DOI: 10.3390/cancers12051132] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Pulsed electric fields (PEFs) have become clinically important through the success of Irreversible Electroporation (IRE), Electrochemotherapy (ECT), and nanosecond PEFs (nsPEFs) for the treatment of tumors. PEFs increase the permeability of cell membranes, a phenomenon known as electroporation. In addition to well-known membrane effects, PEFs can cause profound cytoskeletal disruption. In this review, we summarize the current understanding of cytoskeletal disruption after PEFs. Compiling available studies, we describe PEF-induced cytoskeletal disruption and possible mechanisms of disruption. Additionally, we consider how cytoskeletal alterations contribute to cell-cell and cell-substrate disruption. We conclude with a discussion of cytoskeletal disruption-induced anti-vascular effects of PEFs and consider how a better understanding of cytoskeletal disruption after PEFs may lead to more effective therapies.
Collapse
Affiliation(s)
- Philip M. Graybill
- BEMS Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA;
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Rafael V. Davalos
- BEMS Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA;
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
- Virginia Tech–Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, VA 24061, USA
| |
Collapse
|
118
|
Belabed M, Mauvais FX, Maschalidi S, Kurowska M, Goudin N, Huang JD, Fischer A, de Saint Basile G, van Endert P, Sepulveda FE, Ménasché G. Kinesin-1 regulates antigen cross-presentation through the scission of tubulations from early endosomes in dendritic cells. Nat Commun 2020; 11:1817. [PMID: 32286311 PMCID: PMC7156633 DOI: 10.1038/s41467-020-15692-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 03/25/2020] [Indexed: 11/09/2022] Open
Abstract
Dendritic cells (DCs) constitute a specialized population of immune cells that present exogenous antigen (Ag) on major histocompatibility complex (MHC) class I molecules to initiate CD8 + T cell responses against pathogens and tumours. Although cross-presentation depends critically on the trafficking of Ag-containing intracellular vesicular compartments, the molecular machinery that regulates vesicular transport is incompletely understood. Here, we demonstrate that mice lacking Kif5b (the heavy chain of kinesin-1) in their DCs exhibit a major impairment in cross-presentation and thus a poor in vivo anti-tumour response. We find that kinesin-1 critically regulates antigen cross-presentation in DCs, by controlling Ag degradation, the endosomal pH, and MHC-I recycling. Mechanistically, kinesin-1 appears to regulate early endosome maturation by allowing the scission of endosomal tubulations. Our results highlight kinesin-1’s role as a molecular checkpoint that modulates the balance between antigen degradation and cross-presentation. Kinesin-1 is a motor protein transporting cargo along microtubules. Here the authors show that kinesin-1 is required for antigen cross-presentation and coordinates endosome scission from early endosomes to allow sorting internalized cargoes towards the recycling endosomal or lysosomal compartments.
Collapse
Affiliation(s)
- Meriem Belabed
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France
| | - François-Xavier Mauvais
- Université de Paris, INSERM, U1151, Institut Necker Enfants Malades; Université de Paris; CNRS, UMR8253, F-75015, Paris, France
| | - Sophia Maschalidi
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France
| | - Mathieu Kurowska
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France
| | - Nicolas Goudin
- Cell Imaging Facility, Université de Paris, Imagine Institute, F-75015, Paris, France
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Alain Fischer
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France.,Immunology and Pediatric Hematology Department, Necker Children's Hospital, AP-HP, F-75015, Paris, France.,Collège de France, F-75005, Paris, France
| | - Geneviève de Saint Basile
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France
| | - Peter van Endert
- Université de Paris, INSERM, U1151, Institut Necker Enfants Malades; Université de Paris; CNRS, UMR8253, F-75015, Paris, France
| | - Fernando E Sepulveda
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France.,Centre national de la recherche scientifique (CNRS), F-75015, Paris, France
| | - Gaël Ménasché
- Université de Paris, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, INSERM UMR1163, F-75015, Paris, France.
| |
Collapse
|
119
|
Cho YB, Hong S, Kang KW, Kang JH, Lee SM, Seo YJ. Selective and ATP-competitive kinesin KIF18A inhibitor suppresses the replication of influenza A virus. J Cell Mol Med 2020; 24:5463-5475. [PMID: 32253833 PMCID: PMC7214149 DOI: 10.1111/jcmm.15200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/13/2020] [Accepted: 03/06/2020] [Indexed: 12/22/2022] Open
Abstract
The influenza virus is one of the major public health threats. However, the development of efficient vaccines and therapeutic drugs to combat this virus is greatly limited by its frequent genetic mutations. Because of this, targeting the host factors required for influenza virus replication may be a more effective strategy for inhibiting a broader spectrum of variants. Here, we demonstrated that inhibition of a motor protein kinesin family member 18A (KIF18A) suppresses the replication of the influenza A virus (IAV). The expression of KIF18A in host cells was increased following IAV infection. Intriguingly, treatment with the selective and ATP‐competitive mitotic kinesin KIF18A inhibitor BTB‐1 substantially decreased the expression of viral RNAs and proteins, and the production of infectious viral particles, while overexpression of KIF18A enhanced the replication of IAV. Importantly, BTB‐1 treatment attenuated the activation of AKT, p38 MAPK, SAPK and Ran‐binding protein 3 (RanBP3), which led to the prevention of the nuclear export of viral ribonucleoprotein complexes. Notably, administration of BTB‐1 greatly improved the viability of IAV‐infected mice. Collectively, our results unveiled a beneficial role of KIF18A in IAV replication, and thus, KIF18A could be a potential therapeutic target for the control of IAV infection.
Collapse
Affiliation(s)
- Yong-Bin Cho
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| | - Sungguan Hong
- Department of Chemistry, Chung-Ang University, Seoul, South Korea
| | - Kyung-Won Kang
- Division of Biotechnology, College of Environmental and Bioresources, Jeonbuk National University, Iksan, South Korea
| | - Ji-Hun Kang
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, College of Environmental and Bioresources, Jeonbuk National University, Iksan, South Korea
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| |
Collapse
|
120
|
Wiegand T. A solid-state NMR tool box for the investigation of ATP-fueled protein engines. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2020; 117:1-32. [PMID: 32471533 DOI: 10.1016/j.pnmrs.2020.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 06/11/2023]
Abstract
Motor proteins are involved in a variety of cellular processes. Their main purpose is to convert the chemical energy released during adenosine triphosphate (ATP) hydrolysis into mechanical work. In this review, solid-state Nuclear Magnetic Resonance (NMR) approaches are discussed allowing studies of structures, conformational events and dynamic features of motor proteins during a variety of enzymatic reactions. Solid-state NMR benefits from straightforward sample preparation based on sedimentation of the proteins directly into the Magic-Angle Spinning (MAS) rotor. Protein resonance assignment is the crucial and often time-limiting step in interpreting the wealth of information encoded in the NMR spectra. Herein, potentials, challenges and limitations in resonance assignment for large motor proteins are presented, focussing on both biochemical and spectroscopic approaches. This work highlights NMR tools available to study the action of the motor domain and its coupling to functional processes, as well as to identify protein-nucleotide interactions during events such as DNA replication. Arrested protein states of reaction coordinates such as ATP hydrolysis can be trapped for NMR studies by using stable, non-hydrolysable ATP analogues that mimic the physiological relevant states as accurately as possible. Recent advances in solid-state NMR techniques ranging from Dynamic Nuclear Polarization (DNP), 31P-based heteronuclear correlation experiments, 1H-detected spectra at fast MAS frequencies >100 kHz to paramagnetic NMR are summarized and their applications to the bacterial DnaB helicase from Helicobacter pylori are discussed.
Collapse
Affiliation(s)
- Thomas Wiegand
- Physical Chemistry, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
121
|
Theocharopoulou G. The ubiquitous role of mitochondria in Parkinson and other neurodegenerative diseases. AIMS Neurosci 2020; 7:43-65. [PMID: 32455165 PMCID: PMC7242057 DOI: 10.3934/neuroscience.2020004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Orderly mitochondrial life cycle, plays a key role in the pathology of neurodegenerative diseases. Mitochondria are ubiquitous in neurons as they respond to an ever-changing demand for energy supply. Mitochondria constantly change in shape and location, feature of their dynamic nature, which facilitates a quality control mechanism. Biological studies in mitochondria dynamics are unveiling the mechanisms of fission and fusion, which essentially arrange morphology and motility of these organelles. Control of mitochondrial network homeostasis is a critical factor for the proper function of neurons. Disease-related genes have been reported to be implicated in mitochondrial dysfunction. Increasing evidence implicate mitochondrial perturbation in neuronal diseases, such as AD, PD, HD, and ALS. The intricacy involved in neurodegenerative diseases and the dynamic nature of mitochondria point to the idea that, despite progress toward detecting the biology underlying mitochondrial disorders, its link to these diseases is difficult to be identified in the laboratory. Considering the need to model signaling pathways, both in spatial and temporal level, there is a challenge to use a multiscale modeling framework, which is essential for understanding the dynamics of a complex biological system. The use of computational models in order to represent both a qualitative and a quantitative structure of mitochondrial homeostasis, allows to perform simulation experiments so as to monitor the conformational changes, as well as the intersection of form and function.
Collapse
|
122
|
Xie P. Theoretical Analysis of Dynamics of Kinesin Molecular Motors. ACS OMEGA 2020; 5:5721-5730. [PMID: 32226850 PMCID: PMC7097908 DOI: 10.1021/acsomega.9b03738] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/23/2020] [Indexed: 05/07/2023]
Abstract
Kinesin is a typical molecular motor that can step processively on microtubules powered by hydrolysis of adenosine triphosphate (ATP) molecules, playing a critical role in intracellular transports. Its dynamical properties such as its velocity, stepping ratio, run length, dissociation rate, etc. as well as the load dependencies of these quantities have been well documented through single-molecule experimental methods. In particular, the run length shows a dramatic asymmetry with respect to the direction of the load, and the dissociation rate exhibits a slip-catch-slip bond behavior under the backward load. Here, an analytic theory was provided for the dynamics of kinesin motors under both forward and backward loads, explaining consistently and quantitatively the diverse available experimental results.
Collapse
|
123
|
Microtubule Minus-End Binding Protein CAMSAP2 and Kinesin-14 Motor KIFC3 Control Dendritic Microtubule Organization. Curr Biol 2020; 30:899-908.e6. [PMID: 32084403 PMCID: PMC7063570 DOI: 10.1016/j.cub.2019.12.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/28/2019] [Accepted: 12/18/2019] [Indexed: 12/21/2022]
Abstract
Neuronal dendrites are characterized by an anti-parallel microtubule organization. The mixed oriented microtubules promote dendrite development and facilitate polarized cargo trafficking; however, the mechanism that regulates dendritic microtubule organization is still unclear. Here, we found that the kinesin-14 motor KIFC3 is important for organizing dendritic microtubules and to control dendrite development. The kinesin-14 motor proteins (Drosophila melanogaster Ncd, Saccharomyces cerevisiae Kar3, Saccharomyces pombe Pkl1, and Xenopus laevis XCTK2) are characterized by a C-terminal motor domain and are well described to organize the spindle microtubule during mitosis using an additional microtubule binding site in the N terminus [1-4]. In mammals, there are three kinesin-14 members, KIFC1, KIFC2, and KIFC3. It was recently shown that KIFC1 is important for organizing axonal microtubules in neurons, a process that depends on the two microtubule-interacting domains [5]. Unlike KIFC1, KIFC2 and KIFC3 lack the N-terminal microtubule binding domain and only have one microtubule-interacting domain, the motor domain [6, 7]. Thus, in order to regulate microtubule-microtubule crosslinking or sliding, KIFC2 and KIFC3 need to interact with additional microtubule binding proteins to connect two microtubules. We found that KIFC3 has a dendrite-specific distribution and interacts with microtubule minus-end binding protein CAMSAP2. Depletion of KIFC3 or CAMSAP2 results in increased microtubule dynamics during dendritic development. We propose a model in which CAMSAP2 anchors KIFC3 at microtubule minus ends and immobilizes microtubule arrays in dendrites.
Collapse
|
124
|
Wu Z, Zhang H, Sun Z, Wang C, Chen Y, Luo P, Yan W. Knockdown of Kinesin Family 15 Inhibits Osteosarcoma through Suppressing Cell Proliferation and Promoting Cell Apoptosis. Chemotherapy 2020; 64:187-196. [PMID: 32074608 DOI: 10.1159/000505014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/25/2019] [Indexed: 11/19/2022]
Abstract
Kinesin family (KIF) members have vital roles in mitosis, meiosis, and transport of macromolecules in eukaryotic cells. In this study, we aimed to investigate the role of KIF15 in osteosarcoma. Immunohistochemical staining was performed to determine expression levels of KIF15 in osteosarcoma tissues and adjacent normal tissues. Tissue microarray analysis showed a correlation between the expression of KIF15 and pathological features of patients. Subsequently, lentivirus was used to inhibit the expression of KIF15 in osteosarcoma cells. An MTT assay was performed to examine cell proliferation. Transwell and wound healing assays were used to estimate the invasion and migration of osteosarcoma cells, respectively. Flow cytometric analysis was employed to define the apoptosis of osteosarcoma cells. Our results showed that KIF15 expression was significantly upregulated in osteosarcoma tissues compared with adjacent normal tissues. The Mann-Whitney U test and Spearman correlation analysis showed that the expression levels of KIF15 in osteosarcoma tissues were positively correlated with tumor infiltrate, a pathological characteristic of patients. The expression of KIF15 was successfully suppressed by shKIF15, and the knockdown efficiency reached 80 and 69% in MNNG/HOS and U2OS cells, respectively. Subsequently, knockdown of KIF15 significantly inhibited the capacity of cell proliferation, colony formation, invasion, and migration, but enhanced G2 phase arrest and partially enhanced cell apoptosis. This study preliminarily showed KIF15 to be a critical regulatory molecule involved in osteosarcoma cell proliferation. Consequently, KIF15 can be a potential diagnostic and therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Zhiqiang Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao Zhang
- Department of Orthopaedic Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Zhengwang Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chunmeng Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yong Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Peng Luo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wangjun Yan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China, .,Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China,
| |
Collapse
|
125
|
KIF3C is associated with favorable prognosis in glioma patients and may be regulated by PI3K/AKT/mTOR pathway. J Neurooncol 2020; 146:513-521. [DOI: 10.1007/s11060-020-03399-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/03/2023]
|
126
|
Arata T. Myosin and Other Energy-Transducing ATPases: Structural Dynamics Studied by Electron Paramagnetic Resonance. Int J Mol Sci 2020; 21:E672. [PMID: 31968570 PMCID: PMC7014194 DOI: 10.3390/ijms21020672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/05/2020] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
The objective of this article was to document the energy-transducing and regulatory interactions in supramolecular complexes such as motor, pump, and clock ATPases. The dynamics and structural features were characterized by motion and distance measurements using spin-labeling electron paramagnetic resonance (EPR) spectroscopy. In particular, we focused on myosin ATPase with actin-troponin-tropomyosin, neural kinesin ATPase with microtubule, P-type ion-motive ATPase, and cyanobacterial clock ATPase. Finally, we have described the relationships or common principles among the molecular mechanisms of various energy-transducing systems and how the large-scale thermal structural transition of flexible elements from one state to the other precedes the subsequent irreversible chemical reactions.
Collapse
Affiliation(s)
- Toshiaki Arata
- Department of Biology, Graduate School of Science, Osaka City University, Osaka 558-8585, Japan
| |
Collapse
|
127
|
Dutta R, Sarkar SR. Role of Dynein and Dynactin (DCTN-1) in Neurodegenerative Diseases. ACTA ACUST UNITED AC 2019. [DOI: 10.33805/2641-8991.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathophysiology and concept of degeneration in central nervous system is very complex and overwhelming at times. There is a complex mechanism which exists among different molecules in the cytoplasm of cell bodies of neurons, antegrade and retrograde axonal transport of cargoes and accumulation of certain substances and proteins which can influence the excitatory neurotransmitter like glutamate initiating the process of neurodegeneration. Neurons have extensive processes and communication between those processes and the cell body is crucial to neuronal function, viability and survival over time with progression of age. Researchers believe neurons are uniquely dependent on microtubule-based cargo transport. There is enough evidence to support that deficits in retrograde axonal transport contribute to pathogenesis in multiple neurodegenerative diseases. Cytoplasmic dynein and its regulation by Dynactin (DCTN1) is the major molecular motor cargo involved in autophagy, mitosis and neuronal cell survival. Mutation in dynactin gene located in 2p13.1,is indeed studied very extensively and is considered to be involved directly or indirectly to various conditions like Perry syndrome, familial and sporadic Amyotrophic lateral sclerosis, Hereditary spastic paraplegia, Spinocerebellar Ataxia (SCA-5), Huntingtons disease, Alzheimers disease, Charcot marie tooth disease, Hereditary motor neuropathy 7B, prion disease, parkinsons disease, malformation of cortical development, polymicrogyria to name a few with exception of Multiple Sclerosis (MS).
Collapse
|
128
|
Manna RN, Dutta M, Jana B. Mechanistic study of the ATP hydrolysis reaction in dynein motor protein. Phys Chem Chem Phys 2019; 22:1534-1542. [PMID: 31872818 DOI: 10.1039/c9cp02194a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dynein, a large and complex motor protein, harnesses energy from adenosine triphosphate (ATP) hydrolysis to regulate essential cellular activities. The ATP hydrolysis mechanism for the dynein motor is still shrouded in mystery. Herein, molecular dynamics simulations of a dynein motor disclosed that two water molecules are present close to the γ-phosphate of ATP and Glu1742 at the AAA1 site of dynein. We have proposed three possible mechanisms for the ATP hydrolysis. We divulge by using a quantum mechanics/molecular mechanics (QM/MM) study that two water molecules and Glu1742 are crucial for facilitating the ATP hydrolysis reaction in dynein. Moreover, the ATP hydrolysis step is initiated by the activation of lytic water (W1) by Glu1742 through relay proton transfers with the help of auxiliary water (W2) yielding HPO42- and ADP, as a product. In the next step, a proton is shifted back from Glu1742 to generate inorganic phosphate (H2PO4-) via another relay proton transfer event. The overall activation barrier for the Glu1742 assisted ATP hydrolysis is found to be the most favourable pathway compared to other plausible pathways. We also unearthed that ATP hydrolysis in dynein follows a so-called associative-like pathway in its rate-limiting step. Our study ascertained the important indirect roles of the two amino acids (such as Arg2109, Asn1792) and Mg2+ ion in the ATP hydrolysis of dynein. Additionally, multiple sequence alignment of the different organisms of dynein motors has conveyed the evolutionary importance of the Glu1742, Asn1742, and Arg2109 residues, respectively. As similar mechanisms are also prevalent in other motors, and GTPase and ATPase enzymes, the present finding spells out the definitive requirement of a proton relay process through an extended water-chain as one of the key components in an enzymatic ATP hydrolysis reaction.
Collapse
Affiliation(s)
- Rabindra Nath Manna
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| | - Mandira Dutta
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| | - Biman Jana
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| |
Collapse
|
129
|
Jia Y, Xuan M, Feng X, Duan L, Li J, Li J. Reconstitution of Motor Proteins through Molecular Assembly. CHINESE J CHEM 2019. [DOI: 10.1002/cjoc.201900382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yi Jia
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Xiyun Feng
- Yunnan Normal University Kunming Yunnan 650500 China
| | - Li Duan
- Northwest Institute of Nuclear Technology Xi'an Shaanxi 710024 China
| | - Jieling Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
130
|
Lee S, Higuchi H. 3D rotational motion of an endocytic vesicle on a complex microtubule network in a living cell. BIOMEDICAL OPTICS EXPRESS 2019; 10:6611-6624. [PMID: 31853420 PMCID: PMC6913383 DOI: 10.1364/boe.10.006611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 05/24/2023]
Abstract
The transport dynamics of endocytic vesicles in a living cell contains essential biomedical information. Although the movement mechanism of a vesicle by motor proteins has been revealed, understanding the precise movement of vesicles on the cytoskeleton in a living cell has been considered challenging, due to the complex 3D network of cytoskeletons. Here, we specify the shape of the 3D interaction between the vesicle and microtubule, based on the theoretically estimated location of the microtubule and the vesicle trajectory data acquired at high spatial and temporal precision. We detected that vesicles showed more frequent direction changes with either in very acute or in obtuse angles than right angles, on similar time scales in a microtubule network. Interestingly, when a vesicle interacted with a relatively longer (> 400 nm) microtubule filament, rotational movement along the axis of the microtubule was frequently observed. Our results are expected to give in-depth insight into understanding the actual 3D interactions between the intracellular molecule and complex cytoskeletal network.
Collapse
|
131
|
Goswami A, Saha S, Biswas PK, Schmittel M. (Nano)mechanical Motion Triggered by Metal Coordination: from Functional Devices to Networked Multicomponent Catalytic Machinery. Chem Rev 2019; 120:125-199. [DOI: 10.1021/acs.chemrev.9b00159] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Abir Goswami
- Center of Micro- and Nanochemistry and Engineering, Organische Chemie I, Universität Siegen, Adolf-Reichwein-Strase 2, D-57068 Siegen, Germany
| | - Suchismita Saha
- Center of Micro- and Nanochemistry and Engineering, Organische Chemie I, Universität Siegen, Adolf-Reichwein-Strase 2, D-57068 Siegen, Germany
| | - Pronay Kumar Biswas
- Center of Micro- and Nanochemistry and Engineering, Organische Chemie I, Universität Siegen, Adolf-Reichwein-Strase 2, D-57068 Siegen, Germany
| | - Michael Schmittel
- Center of Micro- and Nanochemistry and Engineering, Organische Chemie I, Universität Siegen, Adolf-Reichwein-Strase 2, D-57068 Siegen, Germany
| |
Collapse
|
132
|
Hao QY, Jiang R, Hu MB, Zhang Y, Wu CY, Guo N. Theoretical analysis and simulation of phase separation in a driven bidirectional two-lane system. Phys Rev E 2019; 100:032133. [PMID: 31640021 DOI: 10.1103/physreve.100.032133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Indexed: 11/07/2022]
Abstract
The two-lane driven system is a type of important model to research some transport systems, and also a powerful tool to investigate properties of nonequilibrium state systems. This paper presents a driven bidirectional two-lane model. The dynamic characteristics of the model with periodic boundary are investigated by Monte Carlo simulation, simple mean field, and cluster mean field methods, respectively. By simulations, phase separations are observed in the system with some values of model parameters. When the phase separation does not occur, cluster mean field results are in good agreement with simulation results. According to the cluster mean field analysis and simulations, a conjecture about the condition that the phase separation happens is proposed. Based on the conjecture, the phase boundary distinguishing phase separation state and homogeneous state is determined, and a corresponding phase diagram is drawn. The conjecture is validated through observing directly the spatiotemporal diagram and investigating the coarsening process of the system by simulation, and a possible mechanism causing the phase separation is also discussed. These outcomes maybe contribute to understand deeply transport systems including the congestion and efficiency of the transport, and enrich explorations of nonequilibrium state systems.
Collapse
Affiliation(s)
- Qing-Yi Hao
- Key Laboratory of Modeling, Simulation and Control of Complex Ecosystem in Dabie Mountains of Anhui Higher Education Institutes, School of Mathematics and Computational Science, Anqing Normal University, Anqing 246133, China.,School of Mathematical Sciences, Fudan University, Shanghai 200433, China
| | - Rui Jiang
- MOE Key Laboratory for Urban Transportation Complex Systems Theory and Technology, Beijing Jiaotong University, Beijing 100044, China
| | - Mao-Bin Hu
- School of Engineering Science, University of Science and Technology of China, Hefei 230026, China
| | - Yunxin Zhang
- School of Mathematical Sciences, Fudan University, Shanghai 200433, China
| | - Chao-Yun Wu
- Key Laboratory of Modeling, Simulation and Control of Complex Ecosystem in Dabie Mountains of Anhui Higher Education Institutes, School of Mathematics and Computational Science, Anqing Normal University, Anqing 246133, China.,School of Engineering Science, University of Science and Technology of China, Hefei 230026, China
| | - Ning Guo
- School of Automotive and Transportation Engineering, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|
133
|
Directional Stepping Model for Yeast Dynein: Longitudinal- and Side-Step Distributions. Biophys J 2019; 117:1892-1899. [PMID: 31676137 DOI: 10.1016/j.bpj.2019.09.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/08/2019] [Accepted: 09/30/2019] [Indexed: 12/19/2022] Open
Abstract
Motor proteins are biological machines that convert chemical energy stored in ATP to mechanical work. Kinesin and dynein are microtubule (MT)-associated motor proteins that, among other functions, facilitate intracellular transport. Here, we focus on dynein motility. We deduce the directional step distribution of yeast dynein motor protein on the MT surface by combing intrinsic features of the dynein and MTs. These include the probability distribution of the separation vector between the two microtubule-binding domains, the angular probability distribution of a single microtubule-binding domain translation, the existence of an MT seam defect, MT-binding sites, and theoretical extension that accounts for a load force on the motor. Our predictions are in excellent accord with the measured longitudinal step size distributions at various load forces. Moreover, we predict the side-step distribution and its dependence on longitudinal load forces, which shows a few surprising features. First, the distribution is broad. Second, in the absence of load, we find a small right-handed bias. Third, the side-step bias is susceptible to the longitudinal load force; it vanishes at a load equal to the motor stalling force and changes to a left-hand bias above that value. Fourth, our results are sensitive to the ability of the motor to explore the seam several times during its walk. Although available measurements of side-way distribution are limited, our findings are amenable to experimental check and, moreover, suggest a diversity of results depending on whether the MT seam is viable to motor sampling.
Collapse
|
134
|
Farhana TI, Nakagawa T, Ohara S, Shintaku H, Kotera H, Yokokawa R. Spatial Patterning of Kinesin-1 and Dynein Motor Proteins in an In Vitro Assay using Aqueous Two-Phase Systems (ATPS). LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13003-13010. [PMID: 31510745 DOI: 10.1021/acs.langmuir.9b01411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cooperativity of motor proteins is essential for intracellular transport. Although their motion is unidirectional, they often cause bidirectional movement by different types of motors as seen in organelles. However, in vitro assessments of such cellular functions are still inadequate owing to the experimental limitations in precisely patterning multiple motors. Here, we present an approach to immobilize two motor proteins, kinesin-1 and dynein, using the aqueous two-phase system (ATPS) made of poly(ethylene glycol) and dextran polymers. The negligible influence of polymer solutions on the attachment and velocity of motor proteins ensures the compatibility of using ATPS as the patterning technique. The selective fixation of kinesin and dynein was assessed using polarity-marked microtubules (PMMTs). Our experimental results show that on a patterned kinesin surface, 72% of PMMTs display minus-end leading motility, while on a dynein surface, 79% of PMMTs display plus-end leading motility. This work offers a universal and biocompatible method to pattern motor proteins of different classes at the nanoscale, providing a new route to study different cellular functions performed by molecular motors such as the formation of mitotic spindles.
Collapse
Affiliation(s)
- Tamanna Ishrat Farhana
- Department of Micro Engineering , Kyoto University , Kyoto Daigaku-Katsura , Nishikyo-ku, Kyoto 615-8540 , Japan
| | - Tomohiro Nakagawa
- Department of Micro Engineering , Kyoto University , Kyoto Daigaku-Katsura , Nishikyo-ku, Kyoto 615-8540 , Japan
| | - Shumpei Ohara
- Department of Micro Engineering , Kyoto University , Kyoto Daigaku-Katsura , Nishikyo-ku, Kyoto 615-8540 , Japan
| | - Hirofumi Shintaku
- Cluster for Pioneering Research, RIKEN , 2-1, Hirosawa , Wako , Saitama 351-0198 , Japan
| | | | - Ryuji Yokokawa
- Department of Micro Engineering , Kyoto University , Kyoto Daigaku-Katsura , Nishikyo-ku, Kyoto 615-8540 , Japan
| |
Collapse
|
135
|
Toyoshima M, Jiang X, Ogawa T, Ohnishi T, Yoshihara S, Balan S, Yoshikawa T, Hirokawa N. Enhanced carbonyl stress induces irreversible multimerization of CRMP2 in schizophrenia pathogenesis. Life Sci Alliance 2019; 2:2/5/e201900478. [PMID: 31591136 PMCID: PMC6781483 DOI: 10.26508/lsa.201900478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/31/2022] Open
Abstract
Enhanced carbonyl stress results in neurodevelopmental deficits by affecting microtubule function through the formation of irreversible dysfunctional multimer of carbonylated CRMP2. Enhanced carbonyl stress underlies a subset of schizophrenia, but its causal effects remain elusive. Here, we elucidated the molecular mechanism underlying the effects of carbonyl stress in iPS cells in which the gene encoding zinc metalloenzyme glyoxalase I (GLO1), a crucial enzyme for the clearance of carbonyl stress, was disrupted. The iPS cells exhibited significant cellular and developmental deficits, and hyper-carbonylation of collapsing response mediator protein 2 (CRMP2). Structural and biochemical analyses revealed an array of multiple carbonylation sites in the functional motifs of CRMP2, particularly D-hook (for dimerization) and T-site (for tetramerization), which are critical for the activity of the CRMP2 tetramer. Interestingly, carbonylated CRMP2 was stacked in the multimer conformation by irreversible cross-linking, resulting in loss of its unique function to bundle microtubules. Thus, the present study revealed that the enhanced carbonyl stress stemmed from the genetic aberrations results in neurodevelopmental deficits through the formation of irreversible dysfunctional multimer of carbonylated CRMP2.
Collapse
Affiliation(s)
- Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Xuguang Jiang
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tadayuki Ogawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Shogo Yoshihara
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, University of Tokyo, Tokyo, Japan .,Center of Excellence in Genome Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
136
|
Xie P, Guo SK, Chen H. A Generalized Kinetic Model for Coupling between Stepping and ATP Hydrolysis of Kinesin Molecular Motors. Int J Mol Sci 2019; 20:ijms20194911. [PMID: 31623357 PMCID: PMC6801755 DOI: 10.3390/ijms20194911] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022] Open
Abstract
A general kinetic model is presented for the chemomechanical coupling of dimeric kinesin molecular motors with and without extension of their neck linkers (NLs). A peculiar feature of the model is that the rate constants of ATPase activity of a kinesin head are independent of the strain on its NL, implying that the heads of the wild-type kinesin dimer and the mutant with extension of its NLs have the same force-independent rate constants of the ATPase activity. Based on the model, an analytical theory is presented on the force dependence of the dynamics of kinesin dimers with and without extension of their NLs at saturating ATP. With only a few adjustable parameters, diverse available single molecule data on the dynamics of various kinesin dimers, such as wild-type kinesin-1, kinesin-1 with mutated residues in the NLs, kinesin-1 with extension of the NLs and wild-type kinesin-2, under varying force and ATP concentration, can be reproduced very well. Additionally, we compare the power production among different kinesin dimers, showing that the mutation in the NLs reduces the power production and the extension of the NLs further reduces the power production.
Collapse
Affiliation(s)
- Ping Xie
- School of Materials Science and Energy Engineering, FoShan University, Guangdong 528000, China.
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Si-Kao Guo
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China.
| | - Hong Chen
- School of Materials Science and Energy Engineering, FoShan University, Guangdong 528000, China.
| |
Collapse
|
137
|
Wegary D, Teklewold A, Prasanna BM, Ertiro BT, Alachiotis N, Negera D, Awas G, Abakemal D, Ogugo V, Gowda M, Semagn K. Molecular diversity and selective sweeps in maize inbred lines adapted to African highlands. Sci Rep 2019; 9:13490. [PMID: 31530852 PMCID: PMC6748982 DOI: 10.1038/s41598-019-49861-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/28/2019] [Indexed: 11/08/2022] Open
Abstract
Little is known on maize germplasm adapted to the African highland agro-ecologies. In this study, we analyzed high-density genotyping by sequencing (GBS) data of 298 African highland adapted maize inbred lines to (i) assess the extent of genetic purity, genetic relatedness, and population structure, and (ii) identify genomic regions that have undergone selection (selective sweeps) in response to adaptation to highland environments. Nearly 91% of the pairs of inbred lines differed by 30-36% of the scored alleles, but only 32% of the pairs of the inbred lines had relative kinship coefficient <0.050, which suggests the presence of substantial redundancy in allelic composition that may be due to repeated use of fewer genetic backgrounds (source germplasm) during line development. Results from different genetic relatedness and population structure analyses revealed three different groups, which generally agrees with pedigree information and breeding history, but less so by heterotic groups and endosperm modification. We identified 944 single nucleotide polymorphic (SNP) markers that fell within 22 selective sweeps that harbored 265 protein-coding candidate genes of which some of the candidate genes had known functions. Details of the candidate genes with known functions and differences in nucleotide diversity among groups predicted based on multivariate methods have been discussed.
Collapse
Affiliation(s)
- Dagne Wegary
- International Maize and Wheat Improvement Center (CIMMYT) - Ethiopia Office, ILRI Campus, CMC Road, Gurd Sholla, P.O. Box 5689, Addis Ababa, Ethiopia
| | - Adefris Teklewold
- International Maize and Wheat Improvement Center (CIMMYT) - Ethiopia Office, ILRI Campus, CMC Road, Gurd Sholla, P.O. Box 5689, Addis Ababa, Ethiopia.
| | - Boddupalli M Prasanna
- International Maize and Wheat Improvement Center (CIMMYT), ICRAF House, United Nations Avenue, Gigiri, P.O. Box 1041-00621, Nairobi, Kenya
| | - Berhanu T Ertiro
- Bako National Maize Research Center, Ethiopian Institute of Agricultural Research (EIAR), Addis Ababa, Ethiopia
| | - Nikolaos Alachiotis
- Institute of Computer Science, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013, Heraklion, Crete, Greece
| | - Demewez Negera
- International Maize and Wheat Improvement Center (CIMMYT) - Ethiopia Office, ILRI Campus, CMC Road, Gurd Sholla, P.O. Box 5689, Addis Ababa, Ethiopia
| | - Geremew Awas
- International Maize and Wheat Improvement Center (CIMMYT) - Ethiopia Office, ILRI Campus, CMC Road, Gurd Sholla, P.O. Box 5689, Addis Ababa, Ethiopia
| | - Demissew Abakemal
- Ambo Agricultural Research Center, P.O. Box 37, West Shoa, Ambo, Ethiopia
| | - Veronica Ogugo
- International Maize and Wheat Improvement Center (CIMMYT), ICRAF House, United Nations Avenue, Gigiri, P.O. Box 1041-00621, Nairobi, Kenya
| | - Manje Gowda
- International Maize and Wheat Improvement Center (CIMMYT), ICRAF House, United Nations Avenue, Gigiri, P.O. Box 1041-00621, Nairobi, Kenya
| | - Kassa Semagn
- International Maize and Wheat Improvement Center (CIMMYT), ICRAF House, United Nations Avenue, Gigiri, P.O. Box 1041-00621, Nairobi, Kenya.
- Africa Rice Center (AfricaRice), M'bé Research Station, 01 B.P. 2551, Bouaké 01, Côte d'Ivoire.
| |
Collapse
|
138
|
Wenzel ED, Speidell A, Flowers SA, Wu C, Avdoshina V, Mocchetti I. Histone deacetylase 6 inhibition rescues axonal transport impairments and prevents the neurotoxicity of HIV-1 envelope protein gp120. Cell Death Dis 2019; 10:674. [PMID: 31515470 PMCID: PMC6742654 DOI: 10.1038/s41419-019-1920-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/25/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
Despite successful antiretroviral drug therapy, a subset of human immunodeficiency virus-1 (HIV)-positive individuals still display synaptodendritic simplifications and functional cognitive impairments referred to as HIV-associated neurocognitive disorders (HANDs). The neurological damage observed in HAND subjects can be experimentally reproduced by the HIV envelope protein gp120. However, the complete mechanism of gp120-mediated neurotoxicity is not entirely understood. Gp120 binds to neuronal microtubules and decreases the level of tubulin acetylation, suggesting that it may impair axonal transport. In this study, we utilized molecular and pharmacological approaches, in addition to microscopy, to examine the relationship between gp120-mediated tubulin deacetylation, axonal transport, and neuronal loss. Using primary rat cortical neurons, we show that gp120 decreases acetylation of tubulin and increases histone deacetylase 6 (HDAC6), a cytoplasmic enzyme that regulates tubulin deacetylation. We also demonstrate that the selective HDAC6 inhibitors tubacin and ACY-1215, which prevented gp120-mediated deacetylation of tubulin, inhibited the ability of gp120 to promote neurite shortening and cell death. We further observed by co-immunoprecipitation and confirmed with mass spectroscopy that exposure of neurons to gp120 decreases the association between tubulin and motor proteins, a well-established consequence of tubulin deacetylation. To assess the physiological consequences of this effect, we examined the axonal transport of brain-derived neurotrophic factor (BDNF). We report that gp120 decreases the velocity of BDNF transport, which was restored to baseline levels when neurons were exposed to HDAC6 inhibitors. Overall, our data suggest that gp120-mediated tubulin deacetylation causes impairment of axonal transport through alterations to the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Erin D Wenzel
- Department of Pharmacology and Physiology, Washington, DC, 20057, USA
| | - Andrew Speidell
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Sarah A Flowers
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Valeria Avdoshina
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Italo Mocchetti
- Department of Pharmacology and Physiology, Washington, DC, 20057, USA. .,Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
139
|
Lee J, Seyler SL, Pressé S. Hydrodynamic interaction facilitates the unsteady transport of two neighboring vesicles. J Chem Phys 2019; 151:094108. [DOI: 10.1063/1.5113880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Julian Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul 06978, South Korea
| | - Sean L. Seyler
- Department of Physics, Arizona State University, Tempe, Arizona 85287, USA
| | - Steve Pressé
- Department of Physics, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
140
|
Protein Kinase CK2-A Putative Target for the Therapy of Diabetes Mellitus? Int J Mol Sci 2019; 20:ijms20184398. [PMID: 31500224 PMCID: PMC6770776 DOI: 10.3390/ijms20184398] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Since diabetes is a global epidemic, the development of novel therapeutic strategies for the treatment of this disease is of major clinical interest. Diabetes is differentiated in two types: type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). T1DM arises from an autoimmune destruction of insulin-producing β-cells whereas T2DM is characterized by an insulin resistance, an impaired insulin reaction of the target cells, and/or dysregulated insulin secretion. In the past, a growing number of studies have reported on the important role of the protein kinase CK2 in the regulation of the survival and endocrine function of pancreatic β-cells. In fact, inhibition of CK2 is capable of reducing cytokine-induced loss of β-cells and increases insulin expression as well as secretion by various pathways that are regulated by reversible phosphorylation of proteins. Moreover, CK2 inhibition modulates pathways that are involved in the development of diabetes and prevents signal transduction, leading to late complications such as diabetic retinopathy. Hence, targeting CK2 may represent a novel therapeutic strategy for the treatment of diabetes.
Collapse
|
141
|
Zhou L, Ouyang L, Chen K, Wang X. Research progress on KIF3B and related diseases. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:492. [PMID: 31700928 DOI: 10.21037/atm.2019.08.47] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Kinesins constitute a protein superfamily that belongs to the motor protein group. Kinesins move along microtubules to exert their various functions, which include intracellular transportation, mitosis, and cell formation. Kinesins are responsible for the transport of various membrane organelles, protein complexes, mRNA and other material, as well as the regulation of intracellular molecular signal pathways. Cumulative studies have also indicated that kinesins are related to the development of a variety of human diseases. At present, there are 14 subfamilies of the kinesin superfamily (KIFs), comprising 45 members. KIF3 is the most common expression in KIFs. KIF3 is a complex composed of a KIF3A/3B heterodimer and a kinesin-related protein, known as KAP3. These complexes are organelles and protein complexes involved in membrane binding in various tissues and transport within cells (nerve cells, melanocytes, epithelial cells, etc.). As a member of the KIF3 subfamily, KIF3B is an essential protein that can regulate cell migration, and proliferation and has critical biological functions. During mitosis, KIF3B is responsible for vesicle transport and membrane expansion, thus regulating cell migration. In recent years, more and more attention has been paid to the relationship between KIF3B and the occurrence and development of diseases. This article reviews the recent advances in the study of KIF3B and its related diseases.
Collapse
Affiliation(s)
- Lihui Zhou
- Department of Orthopaedic Surgery, Xiangshan First People's Hospital, Ningbo 315700, China
| | - Lian Ouyang
- Department of Orthopaedic Surgery, Xiangshan First People's Hospital, Ningbo 315700, China
| | - Keying Chen
- Department of Orthopaedic Surgery, Xiangshan First People's Hospital, Ningbo 315700, China
| | - Xucan Wang
- Department of Orthopaedic Surgery, Xiangshan First People's Hospital, Ningbo 315700, China
| |
Collapse
|
142
|
Wang T, Wang J, Wang J, Mao L, Tang B, Vanderklish PW, Liao X, Xiong ZQ, Liao L. HAP1 is an in vivo UBE3A target that augments autophagy in a mouse model of Angelman syndrome. Neurobiol Dis 2019; 132:104585. [PMID: 31445164 DOI: 10.1016/j.nbd.2019.104585] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/25/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by maternal mutation and paternal imprinting of the gene encoding UBE3A, an E3 ubiquitin ligase. Although several potential target proteins of UBE3A have been reported, how these proteins regulate neuronal development remains unclear. We performed a large-scale quantitative proteomic analysis using stable-isotope labeling of amino acids in mammals (SILAM) in mice with maternal Ube3a mutation. We identified huntingtin (Htt)-associated protein (HAP1), a protein that is involved in Huntington's disease (HD), as a new target of UBE3A. We demonstrate that HAP1 regulates autophagy at the initiation stage by promoting PtdIns3K complex formation and enhancing its activity. HAP1 also co-localized with MAP1LC3 (LC3) and other proteins involved in autophagosome expansion. As a result, HAP1 increased autophagy flux. Strikingly, knocking down of HAP1 alleviated aberrant autophagy in primary neurons from AS mice. Concordantly, treatment of AS neurons with an autophagy inhibitor alleviated the reduction in density of dendritic spines. Furthermore, autophagy inhibition in AS mice partially alleviated a social interaction deficit as shown in open field test. Thus, our results identify HAP1 as an in vivo UBE3A target that contributes to deregulated autophagy and synaptic dysfunction in the central nervous system of AS mouse.
Collapse
Affiliation(s)
- Tingting Wang
- Shanghai Key Laboratory of Regulatory Biology, Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jingyu Wang
- Shanghai Key Laboratory of Regulatory Biology, Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Mao
- Shanghai Key Laboratory of Regulatory Biology, Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Bin Tang
- Shanghai Key Laboratory of Regulatory Biology, Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peter W Vanderklish
- Department of Molecular Medicine, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Xun Liao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Key Laboratory of Brain Functional Genomics of Ministry of Education, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
143
|
Wang Q, Kolomeisky AB. Theoretical Analysis of Run Length Distributions for Coupled Motor Proteins. J Phys Chem B 2019; 123:5805-5813. [PMID: 31246472 DOI: 10.1021/acs.jpcb.9b04710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Motor proteins, also known as biological molecular motors, play important roles in various biological processes. In recent years, properties of single-motor proteins have been intensively investigated using multiple experimental and theoretical tools. However, in real cellular systems biological motors typically function in groups, but the mechanisms of their collective dynamics remain not well understood. Here we investigate theoretically distributions of run lengths for coupled motor proteins that move along linear tracks. Our approach utilizes a method of first-passage processes, which is supplemented by Monte Carlo computer simulations. Theoretical analysis allowed us to clarify several aspects of the cooperativity mechanisms for coupled biological molecular motors. It is found that the run length distribution for two motors, in contrast to single motors, is nonmonotonic. In addition, the transport efficiency of two-motor complexes might be strongly increased. We also argue that the degree of cooperativity is influenced by several characteristics of motor proteins such as the strength of intermolecular interactions, stall forces, dissociations constants, and the detachment forces. The application of our theoretical analysis for several motor proteins is also discussed.
Collapse
|
144
|
Chen HY, Welby E, Li T, Swaroop A. Retinal disease in ciliopathies: Recent advances with a focus on stem cell-based therapies. ACTA ACUST UNITED AC 2019; 4:97-115. [PMID: 31763178 PMCID: PMC6839492 DOI: 10.3233/trd-190038] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ciliopathies display extensive genetic and clinical heterogeneity, varying in severity, age of onset, disease progression and organ systems affected. Retinal involvement, as demonstrated by photoreceptor dysfunction or death, is a highly penetrant phenotype among a vast majority of ciliopathies. Photoreceptor cells possess a specialized and modified sensory cilium with membrane discs where efficient photon capture and ensuing signaling cascade initiate the visual process. Disruptions of cilia biogenesis and protein transport lead to impairment of photoreceptor function and eventually degeneration. Despite advances in elucidation of ciliogenesis and photoreceptor cilia defects, we have limited understanding of pathogenic mechanisms underlying retinal phenotype(s) observed in human ciliopathies. Patient-derived induced pluripotent stem cell (iPSC)-based approaches offer a unique opportunity to complement studies with model organisms and examine cilia disease relevant to humans. Three-dimensional retinal organoids from iPSC lines feature laminated cytoarchitecture, apical-basal polarity and emergence of a ciliary structure, thereby permitting pathogenic modeling of human photoreceptors in vitro. Here, we review the biology of photoreceptor cilia and associated defects and discuss recent progress in evolving treatment modalities, especially using patient-derived iPSCs, for retinal ciliopathies.
Collapse
Affiliation(s)
- Holly Yu Chen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily Welby
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiansen Li
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
145
|
Lopes J, Quint DA, Chapman DE, Xu M, Gopinathan A, Hirst LS. Membrane mediated motor kinetics in microtubule gliding assays. Sci Rep 2019; 9:9584. [PMID: 31270348 PMCID: PMC6610617 DOI: 10.1038/s41598-019-45847-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/13/2019] [Indexed: 01/05/2023] Open
Abstract
Motor-based transport mechanisms are critical for a wide range of eukaryotic cell functions, including the transport of vesicle cargos over long distances. Our understanding of the factors that control and regulate motors when bound to a lipid substrate is however incomplete. We used microtubule gliding assays on a lipid bilayer substrate to investigate the role of membrane diffusion in kinesin-1 on/off binding kinetics and thereby transport velocity. Fluorescence imaging experiments demonstrate motor clustering on single microtubules due to membrane diffusion in the absence of ATP, followed by rapid ATP-induced dissociation during gliding. Our experimental data combined with analytical modeling show that the on/off binding kinetics of the motors are impacted by diffusion and, as a consequence, both the effective binding and unbinding rates for motors are much lower than the expected bare rates. Our results suggest that motor diffusion in the membrane can play a significant role in transport by impacting motor kinetics and can therefore function as a regulator of intracellular transport dynamics.
Collapse
Affiliation(s)
- Joseph Lopes
- Department of Physics, University of California, Merced, CA, 95343, USA
| | - David A Quint
- Department of Physics, University of California, Merced, CA, 95343, USA.,Center for Cellular and Biomolecular Machines (CCBM), University of California, Merced, CA, 95343, USA
| | - Dail E Chapman
- Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Melissa Xu
- Department of Bioengineering, University of California, Merced, CA, 95343, USA
| | - Ajay Gopinathan
- Department of Physics, University of California, Merced, CA, 95343, USA.,Center for Cellular and Biomolecular Machines (CCBM), University of California, Merced, CA, 95343, USA
| | - Linda S Hirst
- Department of Physics, University of California, Merced, CA, 95343, USA.
| |
Collapse
|
146
|
Guo SK, Shi XX, Wang PY, Xie P. Force dependence of unbinding rate of kinesin motor during its processive movement on microtubule. Biophys Chem 2019; 253:106216. [PMID: 31288174 DOI: 10.1016/j.bpc.2019.106216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/26/2019] [Accepted: 06/30/2019] [Indexed: 12/15/2022]
Abstract
Kinesin is a biological molecular motor that can move continuously on microtubule until it unbinds. Here, we studied computationally the force dependence of the unbinding rate of the motor. Our results showed that while the unbinding rate under the forward load has the expected characteristic of "slip bond", with the unbinding rate increasing monotonically with the increase of the forward load, the unbinding rate under the backward load shows counterintuitive characteristic of "slip-catch-slip bond": as the backward load increases, the unbinding rate increases exponentially firstly, then drops rapidly and then increases again. Our calculated data are in agreement with the available single-molecule data from different research groups. The mechanism of the slip-catch-slip bond was revealed.
Collapse
Affiliation(s)
- Si-Kao Guo
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing 100190, China; School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Xuan Shi
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing 100190, China; School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng-Ye Wang
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing 100190, China; School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Xie
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing 100190, China; School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
147
|
Abstract
Molecular machines are an important and emerging frontier in research encompassing interdisciplinary subjects of chemistry, physics, biology, and nanotechnology. Although there has been major interest in creating synthetic molecular machines, research on natural molecular machines is also crucial. Biomolecular motors are natural molecular machines existing in nearly every living systems. They play a vital role in almost every essential process ranging from intracellular transport to cell division, muscle contraction and the biosynthesis of ATP that fuels life processes. The construction of biomolecular motor-based biomimetic systems can help not only to deeply understand the mechanisms of motor proteins in the biological process but also to push forward the development of bionics and biomolecular motor-based devices or nanomachines. From combination of natural biomolecular motors with supramolecular chemistry, great opportunities could emerge toward the development of intelligent molecular machines and biodevices. In this Account, we describe our efforts to design and reconstitute biomolecular motor-based active biomimetic systems, in particular, the combination of motor proteins with layer-by-layer (LbL) assembled cellular structures. They are divided into two parts: (i) reconstitution of rotary molecular motor FOF1-ATPase, which is coated on the surface of LbL assembled microcapsules or multilayers and synthesizes adenosine triphosphate (ATP) through creating a proton gradient; (ii) molecular assembly of linear molecular motors, the kinesin-based active biomimetic systems, which are coated on a planar surface or LbL assembled tubular structure and drive the movement of microtubules. LbL assembled structures offer motor proteins with an environment that resembles the natural cell. This enables high activity and optimized function of the motor proteins. The assembled biomolecular motors can mimic their functionalities from the natural system. In addition, LbL assembly provides facile integration of functional components into motor protein-based active biomimetic systems and achieves the manipulation of FOF1-ATPase and kinesin. For FOF1-ATPase, the light-driven proton gradient and controlled ATP synthesis are highlighted. For kinesin, the strategies used for the direction and velocity control of kinesin-based molecular shuttles are discussed. We hope this research can inspire new ideas and propel the actual applications of biomolecular motor-based devices in the future.
Collapse
Affiliation(s)
- Yi Jia
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
148
|
Teratogenic jervine increases the activity of doxorubicin in MCF-7/ADR cells by inhibiting ABCB1. Biomed Pharmacother 2019; 117:109059. [PMID: 31207578 DOI: 10.1016/j.biopha.2019.109059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022] Open
Abstract
Jervine is a natural teratogenic compound isolated from Veratrum californicum. In this study, for the first time, we revealed a novel activity of jervine in sensitizing the anti-proliferation effect of doxorubicin (DOX). We demonstrated that the synergistic mechanism was related to the intracellular accumulation of DOX via modulating ABCB1 transportation. Jervine did not affect the expression of ABCB1 in mRNA nor protein levels. However, jervine increased the ATPase activity of ABCB1 and possibly served as a substrate of ABCB1. The molecular docking results indicated that jervine was bound to a closed ABCB1 conformation and blocked drug entrance to the central binding site at the transmembrane domain. The present study identifies jervine acts as a substrate of ABCB1, and has potential to be developed as a novel and potent chemotherapy sensitizer used for patients developing multidrug resistance.
Collapse
|
149
|
Shin K, Song S, Song YH, Hahn S, Kim JH, Lee G, Jeong IC, Sung J, Lee KT. Anomalous Dynamics of in Vivo Cargo Delivery by Motor Protein Multiplexes. J Phys Chem Lett 2019; 10:3071-3079. [PMID: 31117686 DOI: 10.1021/acs.jpclett.9b01106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Vesicle transport conducted by motor protein multiplexes (MPMs), which is ubiquitous among eukaryotes, shows anomalous and stochastic dynamics qualitatively different from the dynamics of thermal motion and artificial active matter; the relationship between in vivo vesicle-delivery dynamics and the underlying physicochemical processes is not yet quantitatively understood. Addressing this issue, we perform accurate tracking of individual vesicles, containing upconverting nanoparticles, transported by kinesin-dynein-multiplexes along axonal microtubules. The mean-square-displacement of vesicles along the microtubule exhibits unusual dynamic phase transitions that are seemingly inconsistent with the scaling behavior of the mean-first-passage time over the travel length. These paradoxical results and the vesicle displacement distribution are quantitatively explained and predicted by a multimode MPM model, developed in the current work, where ATP-hydrolysis-coupled motion of MPM has both unidirectional and bidirectional modes.
Collapse
Affiliation(s)
- Kyujin Shin
- Department of Chemistry, School of Physics and Chemistry , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Korea
| | - Sanggeun Song
- Creative Research Initiative Center for Chemical Dynamics in Living Cells , Chung-Ang University , Seoul 06974 , Korea
- Department of Chemistry , Chung-Ang University , Seoul 06974 , Korea
- National Institute of Innovative Functional Imaging , Chung-Ang University , Seoul 06974 , Korea
| | - Yo Han Song
- Department of Chemistry, School of Physics and Chemistry , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Korea
| | - Seungsoo Hahn
- Creative Research Initiative Center for Chemical Dynamics in Living Cells , Chung-Ang University , Seoul 06974 , Korea
- Da Vinci College of General Education , Chung-Ang University , Seoul 06974 , Korea
| | - Ji-Hyun Kim
- Creative Research Initiative Center for Chemical Dynamics in Living Cells , Chung-Ang University , Seoul 06974 , Korea
| | - Gibok Lee
- Department of Chemistry, School of Physics and Chemistry , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Korea
| | - In-Chun Jeong
- Creative Research Initiative Center for Chemical Dynamics in Living Cells , Chung-Ang University , Seoul 06974 , Korea
- Department of Chemistry , Chung-Ang University , Seoul 06974 , Korea
- National Institute of Innovative Functional Imaging , Chung-Ang University , Seoul 06974 , Korea
| | - Jaeyoung Sung
- Creative Research Initiative Center for Chemical Dynamics in Living Cells , Chung-Ang University , Seoul 06974 , Korea
- Department of Chemistry , Chung-Ang University , Seoul 06974 , Korea
- National Institute of Innovative Functional Imaging , Chung-Ang University , Seoul 06974 , Korea
| | - Kang Taek Lee
- Department of Chemistry, School of Physics and Chemistry , Gwangju Institute of Science and Technology (GIST) , Gwangju 61005 , Korea
| |
Collapse
|
150
|
Wu WD, Yu KW, Zhong N, Xiao Y, She ZY. Roles and mechanisms of Kinesin-6 KIF20A in spindle organization during cell division. Eur J Cell Biol 2019; 98:74-80. [DOI: 10.1016/j.ejcb.2018.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
|