1
|
Liu F, Medyukhina A, Olesen KM, Shirinifard A, Jin H, Li L, Mapelli M, Khairy K, Han YG. Canonical Hedgehog Signaling Controls Astral Microtubules and Mitotic Spindle Orientation in Neural Progenitors and iPSCs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639780. [PMID: 40060449 PMCID: PMC11888381 DOI: 10.1101/2025.02.23.639780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Mitotic spindle orientation is crucial for cell fate determination and tissue organization. Although the intracellular machinery governing spindle orientation is well characterized, whether and how secreted factors, such as morphogens, regulate this process remains poorly understood. This study investigated the role of Hedgehog (HH) signaling in modulating mitotic spindle orientation in neural progenitor cells and in induced pluripotent stem cells (iPSCs). Time-lapse microscopy of cerebral organoids and iPSCs revealed that HH signaling increases the angle of the mitotic spindle relative to the apical surface, prolongs mitosis, and enhances spindle rotation. Mechanistically, HH signaling reduces both the number and the length of astral microtubules, key regulators of spindle orientation. This reduction correlates with increased spindle angle in iPSCs. Furthermore, we show that canonical HH signaling, involving GLI-dependent transcriptional regulation, contributes to these effects. RNA sequencing and gene set enrichment analysis (GSEA) revealed that HH signaling upregulates genes associated with microtubule depolymerization, suggesting a transcriptional mechanism by which HH signaling influences astral microtubule dynamics and, consequently, mitotic spindle orientation. These findings highlight a novel link between a morphogen, transcriptional regulation, and the control of mitotic spindle orientation, with implications for development and tissue homeostasis.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Anna Medyukhina
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Kris M Olesen
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Lei Li
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Marina Mapelli
- European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Khaled Khairy
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
2
|
Wu S, Cai Y, Zhang L, Li X, Liu X, Zhou G, Luo H, Li R, Huo Y, Zhang Z, Chen S, Huang J, Shi J, Ding S, Sun Z, Zhou Z, Wang P, Wang G. Noncoding RNA Terc-53 and hyaluronan receptor Hmmr regulate aging in mice. Protein Cell 2025; 16:28-48. [PMID: 38721690 DOI: 10.1093/procel/pwae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/11/2024] [Indexed: 01/07/2025] Open
Abstract
One of the basic questions in the aging field is whether there is a fundamental difference between the aging of lower invertebrates and mammals. A major difference between the lower invertebrates and mammals is the abundancy of noncoding RNAs, most of which are not conserved. We have previously identified a noncoding RNA Terc-53 that is derived from the RNA component of telomerase Terc. To study its physiological functions, we generated two transgenic mouse models overexpressing the RNA in wild-type and early-aging Terc-/- backgrounds. Terc-53 mice showed age-related cognition decline and shortened life span, even though no developmental defects or physiological abnormality at an early age was observed, indicating its involvement in normal aging of mammals. Subsequent mechanistic study identified hyaluronan-mediated motility receptor (Hmmr) as the main effector of Terc-53. Terc-53 mediates the degradation of Hmmr, leading to an increase of inflammation in the affected tissues, accelerating organismal aging. adeno-associated virus delivered supplementation of Hmmr in the hippocampus reversed the cognition decline in Terc-53 transgenic mice. Neither Terc-53 nor Hmmr has homologs in C. elegans. Neither do arthropods express hyaluronan. These findings demonstrate the complexity of aging in mammals and open new paths for exploring noncoding RNA and Hmmr as means of treating age-related physical debilities and improving healthspan.
Collapse
Affiliation(s)
- Sipeng Wu
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yiqi Cai
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Lixiao Zhang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiang Li
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xu Liu
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guangkeng Zhou
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Hongdi Luo
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Renjian Li
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yujia Huo
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhirong Zhang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Siyi Chen
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jinliang Huang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiahao Shi
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Shanwei Ding
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhe Sun
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zizhuo Zhou
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Pengcheng Wang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Geng Wang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
3
|
Chen YJ, Tseng SC, Chen PT, Hwang E. The non-mitotic role of HMMR in regulating the localization of TPX2 and the dynamics of microtubules in neurons. eLife 2024; 13:RP94547. [PMID: 38904660 PMCID: PMC11192530 DOI: 10.7554/elife.94547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
A functional nervous system is built upon the proper morphogenesis of neurons to establish the intricate connection between them. The microtubule cytoskeleton is known to play various essential roles in this morphogenetic process. While many microtubule-associated proteins (MAPs) have been demonstrated to participate in neuronal morphogenesis, the function of many more remains to be determined. This study focuses on a MAP called HMMR in mice, which was originally identified as a hyaluronan binding protein and later found to possess microtubule and centrosome binding capacity. HMMR exhibits high abundance on neuronal microtubules and altering the level of HMMR significantly affects the morphology of neurons. Instead of confining to the centrosome(s) like cells in mitosis, HMMR localizes to microtubules along axons and dendrites. Furthermore, transiently expressing HMMR enhances the stability of neuronal microtubules and increases the formation frequency of growing microtubules along the neurites. HMMR regulates the microtubule localization of a non-centrosomal microtubule nucleator TPX2 along the neurite, offering an explanation for how HMMR contributes to the promotion of growing microtubules. This study sheds light on how cells utilize proteins involved in mitosis for non-mitotic functions.
Collapse
Affiliation(s)
- Yi-Ju Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Shun-Cheng Tseng
- Department of Orthopedic Surgery, Changhua Christian HospitalChanghuaTaiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Peng-Tzu Chen
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| | - Eric Hwang
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung UniversityHsinchuTaiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung UniversityHsinchuTaiwan
| |
Collapse
|
4
|
Shang J, Zhang X, Hou G, Qi Y. HMMR potential as a diagnostic and prognostic biomarker of cancer-speculation based on a pan-cancer analysis. Front Surg 2023; 9:998598. [PMID: 36704516 PMCID: PMC9873350 DOI: 10.3389/fsurg.2022.998598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Background Although the status of universal upregulation for the Hyaluronan-Mediated Motility Receptor (HMMR) in pan-cancer is still unknown, HMMR is upregulated and associated with poor prognosis for some tumors. Methods Exploring HMMR expression in different tumor types using The Cancer Genome Atlas (TCGA) or other public databases for a pan-cancer analysis, exploring the relationship between HMMR and tumor prognosis, and exploring the role of HMMR in tumor immunity. Results No matter the pairing or unpairing of data, HMMR expression generally increased compared to corresponding normal tissue. Based on a CCLE study, our results indicated that HMMR is widely expressed in various tumor cells. For most tumor types, high HMMR expression was associated with reduced Overall Survival (OS), Return to Functional Status (RFS), and Platinum Free Interval (PFI). ROC curves indicated that HMMR displays high prediction potential for most tumor types. In pan-cancer, HMMR is correlated with some clinical staging, immune cells, and immune checkpoints for some tumors. The GO/KEGG enrichment analysis results for proteins most closely related to HMMR indicate that the most highly enriched pathways are all related to tumor development. Conclusions Our pan-cancer analysis of HMMR suggests that HMMR can be used as a potential diagnostic and prognostic indicator of pan-cancer and that HMMR may be involved in tumor development.
Collapse
Affiliation(s)
- Junyi Shang
- Department of Respiratory and Critical Care Medicine; Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine; Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangjie Hou
- Department of Thoracic Surgery; Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine; Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
De la Cruz G, Nikolaishvili Feinberg N, Williams SE. Automated Immunofluorescence Staining for Analysis of Mitotic Stages and Division Orientation in Brain Sections. Methods Mol Biol 2023; 2583:63-79. [PMID: 36418726 DOI: 10.1007/978-1-0716-2752-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Microcephaly often results from mitotic defects in neuronal progenitors, frequently by decreasing proliferation rates or shifting cell fates. During neurogenesis, oriented cell division-the molecular control of mitotic spindle positioning to control the axis of division-represents an important mechanism to balance expansion of the progenitor pool with generating cellular diversity. While mostly studied in the context of cortical development, more recently, spindle orientation has emerged as a key player in the formation of other brain regions such as the cerebellum. Here we describe methods to perform automated dual-color fluorescent immunohistochemistry on murine cerebellar sections using the mitotic markers phospho-Histone H3 and Survivin, and detail analytical and statistical approaches to display and compare division orientation datasets.
Collapse
Affiliation(s)
- Gabriela De la Cruz
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Pathology Services Core, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nana Nikolaishvili Feinberg
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Pathology Services Core, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott E Williams
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
6
|
Zhong T, Gongye X, Wang M, Yu J. Understanding the underlying mechanisms governing spindle orientation: How far are we from there? J Cell Mol Med 2022; 26:4904-4910. [PMID: 36029193 PMCID: PMC9549511 DOI: 10.1111/jcmm.17526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Proper spindle orientation is essential for cell fate determination and tissue morphogenesis. Recently, accumulating studies have elucidated several factors that regulate spindle orientation, including geometric, internal and external cues. Abnormality in these factors generally leads to defects in the physiological functions of various organs and the development of severe diseases. Herein, we first review models that are commonly used for studying spindle orientation. We then review a conservative heterotrimeric complex critically involved in spindle orientation regulation in different models. Finally, we summarize some cues that affect spindle orientation and explore whether we can establish a model that precisely elucidates the effects of spindle orientation without interfusing other spindle functions. We aim to summarize current models used in spindle orientation studies and discuss whether we can build a model that disturbs spindle orientation alone. This can substantially improve our understanding of how spindle orientation is regulated and provide insights to investigate this complex event.
Collapse
Affiliation(s)
- Tao Zhong
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Xiaoxiao Gongye
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Minglei Wang
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Jinming Yu
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
7
|
Hinneh JA, Gillis JL, Moore NL, Butler LM, Centenera MM. The role of RHAMM in cancer: Exposing novel therapeutic vulnerabilities. Front Oncol 2022; 12:982231. [PMID: 36033439 PMCID: PMC9400171 DOI: 10.3389/fonc.2022.982231] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Receptor for hyaluronic acid-mediated motility (RHAMM) is a cell surface receptor for hyaluronic acid that is critical for cell migration and a cell cycle protein involved in microtubule assembly and stability. These functions of RHAMM are required for cellular stress responses and cell cycle progression but are also exploited by tumor cells for malignant progression and metastasis. RHAMM is often overexpressed in tumors and is an independent adverse prognostic factor for a number of cancers such as breast and prostate. Interestingly, pharmacological or genetic inhibition of RHAMM in vitro and in vivo ablates tumor invasiveness and metastatic spread, implicating RHAMM as a potential therapeutic target to restrict tumor growth and improve patient survival. However, RHAMM’s pro-tumor activity is dependent on its subcellular distribution, which complicates the design of RHAMM-directed therapies. An alternative approach is to identify downstream signaling pathways that mediate RHAMM-promoted tumor aggressiveness. Herein, we discuss the pro-tumoral roles of RHAMM and elucidate the corresponding regulators and signaling pathways mediating RHAMM downstream events, with a specific focus on strategies to target the RHAMM signaling network in cancer cells.
Collapse
Affiliation(s)
- Josephine A. Hinneh
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Joanna L. Gillis
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Nicole L. Moore
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Lisa M. Butler
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Lisa M. Butler, ; Margaret M. Centenera,
| | - Margaret M. Centenera
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Lisa M. Butler, ; Margaret M. Centenera,
| |
Collapse
|
8
|
Wilsch-Bräuninger M, Huttner WB. Primary Cilia and Centrosomes in Neocortex Development. Front Neurosci 2021; 15:755867. [PMID: 34744618 PMCID: PMC8566538 DOI: 10.3389/fnins.2021.755867] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
During mammalian brain development, neural stem and progenitor cells generate the neurons for the six-layered neocortex. The proliferative capacity of the different types of progenitor cells within the germinal zones of the developing neocortex is a major determinant for the number of neurons generated. Furthermore, the various modes of progenitor cell divisions, for which the orientation of the mitotic spindle of progenitor cells has a pivotal role, are a key parameter to ensure the appropriate size and proper cytoarchitecture of the neocortex. Here, we review the roles of primary cilia and centrosomes of progenitor cells in these processes during neocortical development. We specifically focus on the apical progenitor cells in the ventricular zone. In particular, we address the alternating, dual role of the mother centriole (i) as a component of one of the spindle poles during mitosis, and (ii) as the basal body of the primary cilium in interphase, which is pivotal for the fate of apical progenitor cells and their proliferative capacity. We also discuss the interactions of these organelles with the microtubule and actin cytoskeleton, and with junctional complexes. Centriolar appendages have a specific role in this interaction with the cell cortex and the plasma membrane. Another topic of this review is the specific molecular composition of the ciliary membrane and the membrane vesicle traffic to the primary cilium of apical progenitors, which underlie the ciliary signaling during neocortical development; this signaling itself, however, is not covered in depth here. We also discuss the recently emerging evidence regarding the composition and roles of primary cilia and centrosomes in basal progenitors, a class of progenitors thought to be of particular importance for neocortex expansion in development and evolution. While the tight interplay between primary cilia and centrosomes makes it difficult to allocate independent roles to either organelle, mutations in genes encoding ciliary and/or centrosome proteins indicate that both are necessary for the formation of a properly sized and functioning neocortex during development. Human neocortical malformations, like microcephaly, underpin the importance of primary cilia/centrosome-related processes in neocortical development and provide fundamental insight into the underlying mechanisms involved.
Collapse
Affiliation(s)
| | - Wieland B Huttner
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
9
|
Tian S, Fu L, Zhang J, Xu J, Yuan L, Qin J, Zhang W. Identification of a DNA Methylation-Driven Genes-Based Prognostic Model and Drug Targets in Breast Cancer: In silico Screening of Therapeutic Compounds and in vitro Characterization. Front Immunol 2021; 12:761326. [PMID: 34745136 PMCID: PMC8567755 DOI: 10.3389/fimmu.2021.761326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
DNA methylation is a vital epigenetic change that regulates gene transcription and helps to keep the genome stable. The deregulation hallmark of human cancer is often defined by aberrant DNA methylation which is critical for tumor formation and controls the expression of several tumor-associated genes. In various cancers, methylation changes such as tumor suppressor gene hypermethylation and oncogene hypomethylation are critical in tumor occurrences, especially in breast cancer. Detecting DNA methylation-driven genes and understanding the molecular features of such genes could thus help to enhance our understanding of pathogenesis and molecular mechanisms of breast cancer, facilitating the development of precision medicine and drug discovery. In the present study, we retrospectively analyzed over one thousand breast cancer patients and established a robust prognostic signature based on DNA methylation-driven genes. Then, we calculated immune cells abundance in each patient and lower immune activity existed in high-risk patients. The expression of leukocyte antigen (HLA) family genes and immune checkpoints genes were consistent with the above results. In addition, more mutated genes were observed in the high-risk group. Furthermore, a in silico screening of druggable targets and compounds from CTRP and PRISM databases was performed, resulting in the identification of five target genes (HMMR, CCNB1, CDC25C, AURKA, and CENPE) and five agents (oligomycin A, panobinostat, (+)-JQ1, voxtalisib, and arcyriaflavin A), which might have therapeutic potential in treating high-risk breast cancer patients. Further in vitro evaluation confirmed that (+)-JQ1 had the best cancer cell selectivity and exerted its anti-breast cancer activity through CENPE. In conclusion, our study provided new insights into personalized prognostication and may inspire the integration of risk stratification and precision therapy.
Collapse
Affiliation(s)
- Saisai Tian
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Lu Fu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jinbo Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Department of Pharmacy, Tianjin Rehabilitation Center of Joint Logistics Support Force, Tianjin, China
| | - Jia Xu
- School of Pharmacy, Henan University, Kaifeng, China
| | - Li Yuan
- Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiangjiang Qin
- Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Innovation Center of Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
RHAMM Is a Multifunctional Protein That Regulates Cancer Progression. Int J Mol Sci 2021; 22:ijms221910313. [PMID: 34638654 PMCID: PMC8508827 DOI: 10.3390/ijms221910313] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/09/2023] Open
Abstract
The functional complexity of higher organisms is not easily accounted for by the size of their genomes. Rather, complexity appears to be generated by transcriptional, translational, and post-translational mechanisms and tissue organization that produces a context-dependent response of cells to specific stimuli. One property of gene products that likely increases the ability of cells to respond to stimuli with complexity is the multifunctionality of expressed proteins. Receptor for hyaluronan-mediated motility (RHAMM) is an example of a multifunctional protein that controls differential responses of cells in response-to-injury contexts. Here, we trace its evolution into a sensor-transducer of tissue injury signals in higher organisms through the detection of hyaluronan (HA) that accumulates in injured microenvironments. Our goal is to highlight the domain and isoform structures that generate RHAMM's function complexity and model approaches for targeting its key functions to control cancer progression.
Collapse
|
11
|
Centrosome regulation and function in mammalian cortical neurogenesis. Curr Opin Neurobiol 2021; 69:256-266. [PMID: 34303132 DOI: 10.1016/j.conb.2021.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023]
Abstract
As the primary microtubule-organizing center in animal cells, centrosomes regulate microtubule cytoskeleton to support various cellular behaviors. They also serve as the base for nucleating primary cilia, the hub of diverse signaling pathways. Cells typically possess one centrosome that contains two inequal centrioles and undergoes semi-conservative duplication during cell division, resulting in two centrosomes with an inherent asymmetry in age and properties. While the centrosome is ubiquitously present, mutations of centrosome proteins are strongly associated with human microcephaly characterized by a small cerebral cortex, underscoring the importance of an intact centrosome in supporting cortical neurogenesis. Here we review recent advances on centrosome regulation and function in mammalian cortical neural progenitors and discuss the implications for a better understanding of cortical neurogenesis and related disease mechanisms.
Collapse
|
12
|
Doi N, Togari H, Minagi K, Iwaoka Y, Tai A, Nakaoji K, Hamada K, Tatsuka M. 2-O-Octadecylascorbic acid represses RhoGDIβ expression and ameliorates DNA damage-induced abnormal spindle orientations. J Cell Biochem 2021; 122:739-751. [PMID: 33586155 DOI: 10.1002/jcb.29908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 01/08/2023]
Abstract
The appropriate regulation of spindle orientation maintains proper tissue homeostasis and avoids aberrant tissue repair or regeneration. Spindle misorientation due to imbalance or improper functioning leads to a loss of tissue integrity and aberrant growth, such as tissue loss or overgrowth. Pharmacological manipulation to prevent spindle misorientation will enable a better understanding of how spindle orientation is involved in physiological and pathological conditions and will provide therapeutic possibilities to treat patients associated with abnormal tissue function caused by spindle misorientation. N-terminal-deleted Rho guanine nucleotide dissociation inhibitor β (RhoGDIβ/RhoGDI2/LyGDI) produced by caspase-3 activation perturbs spindle orientation in surviving cells following exposure to either ionizing radiation or UVC. Thus, presumably, RhoGDIβ cleaved by caspase-3 activation acts as a determinant of radiation-induced spindle misorientation that promote aberrant tissue repair due to deregulation of directional organization of cell population and therefore becomes a potential target of drugs to prevent such response. The objective of this study was to screen and identify chemicals that suppress RhoGDIβ expression. We focused our attention on ascorbic acid (AA) derivatives because of their impact on the maintenance of skin tissue homeostasis. Here, we screened for AA derivatives that suppress RhoGDIβ expression in HeLa cells and identified a lipophilic derivative, 2-O-octadecylascorbic acid (2-OctadecylAA), as a novel RhoGDIβ inhibitor that ameliorated ionizing radiation-induced abnormal spindle orientations. Among all examined AA derivatives, which were also antioxidative, the inhibition activity was specific to 2-OctadecylAA. Therefore, this activity was not due to simple antioxidant properties. 2-OctadecylAA was previously shown to prevent hepatocellular carcinoma development. Our findings suggest that the anticarcinogenic effects of 2-OctadecylAA are partly due to RhoGDIβ inhibition mechanisms by which spindle orientation perturbations are attenuated. Thus, the molecular targeting features of RhoGDIβ warrant its further development for the treatment or control of spindle orientation abnormalities that affect epithelial homeostasis.
Collapse
Affiliation(s)
- Natsumi Doi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Hiro Togari
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Kenji Minagi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Yuji Iwaoka
- Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan
| | - Akihiro Tai
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Koichi Nakaoji
- Research & Development Division, Pias Corporation, Kobe, Japan
| | - Kazuhiko Hamada
- Research & Development Division, Pias Corporation, Kobe, Japan
| | - Masaaki Tatsuka
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| |
Collapse
|
13
|
Pibuel MA, Poodts D, Díaz M, Hajos SE, Lompardía SL. The scrambled story between hyaluronan and glioblastoma. J Biol Chem 2021; 296:100549. [PMID: 33744285 PMCID: PMC8050860 DOI: 10.1016/j.jbc.2021.100549] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in cancer biology are revealing the importance of the cancer cell microenvironment on tumorigenesis and cancer progression. Hyaluronan (HA), the main glycosaminoglycan in the extracellular matrix, has been associated with the progression of glioblastoma (GBM), the most frequent and lethal primary tumor in the central nervous system, for several decades. However, the mechanisms by which HA impacts GBM properties and processes have been difficult to elucidate. In this review, we provide a comprehensive assessment of the current knowledge on HA's effects on GBM biology, introducing its primary receptors CD44 and RHAMM and the plethora of relevant downstream signaling pathways that can scramble efforts to directly link HA activity to biological outcomes. We consider the complexities of studying an extracellular polymer and the different strategies used to try to capture its function, including 2D and 3D in vitro studies, patient samples, and in vivo models. Given that HA affects not only migration and invasion, but also cell proliferation, adherence, and chemoresistance, we highlight the potential role of HA as a therapeutic target. Finally, we review the different existing approaches to diminish its protumor effects, such as the use of 4-methylumbelliferone, HA oligomers, and hyaluronidases and encourage further research along these lines in order to improve the survival and quality of life of GBM patients.
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| |
Collapse
|
14
|
He Z, Mei L, Connell M, Maxwell CA. Hyaluronan Mediated Motility Receptor (HMMR) Encodes an Evolutionarily Conserved Homeostasis, Mitosis, and Meiosis Regulator Rather than a Hyaluronan Receptor. Cells 2020; 9:cells9040819. [PMID: 32231069 PMCID: PMC7226759 DOI: 10.3390/cells9040819] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
Hyaluronan is an extracellular matrix component that absorbs water in tissues and engages cell surface receptors, like Cluster of Differentiation 44 (CD44), to promote cellular growth and movement. Consequently, CD44 demarks stem cells in normal tissues and tumor-initiating cells isolated from neoplastic tissues. Hyaluronan mediated motility receptor (HMMR, also known as RHAMM) is another one of few defined hyaluronan receptors. HMMR is also associated with neoplastic processes and its role in cancer progression is often attributed to hyaluronan-mediated signaling. But, HMMR is an intracellular, microtubule-associated, spindle assembly factor that localizes protein complexes to augment the activities of mitotic kinases, like polo-like kinase 1 and Aurora kinase A, and control dynein and kinesin motor activities. Expression of HMMR is elevated in cells prior to and during mitosis and tissues with detectable HMMR expression tend to be highly proliferative, including neoplastic tissues. Moreover, HMMR is a breast cancer susceptibility gene product. Here, we briefly review the associations between HMMR and tumorigenesis as well as the structure and evolution of HMMR, which identifies Hmmr-like gene products in several insect species that do not produce hyaluronan. This review supports the designation of HMMR as a homeostasis, mitosis, and meiosis regulator, and clarifies how its dysfunction may promote the tumorigenic process and cancer progression.
Collapse
Affiliation(s)
- Zhengcheng He
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Lin Mei
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
| | - Christopher A. Maxwell
- Department of Pediatrics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (Z.H.); (L.M.); (M.C.)
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital, Vancouver, BC V5Z 4H4, Canada
- Correspondence: ; Tel.: +1-6048752000 (ext. 4691)
| |
Collapse
|
15
|
Doi N, Kunimatsu Y, Fujiura K, Togari H, Minagi K, Nakaoji K, Hamada K, Temme A, Tatsuka M. RhoGDIβ affects HeLa cell spindle orientation following UVC irradiation. J Cell Physiol 2019; 234:15134-15146. [PMID: 30652309 DOI: 10.1002/jcp.28154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
The molecular signals that regulate mitotic spindle orientation to determine the proper division axis play a critical role in the development and maintenance of tissue homeostasis. However, deregulation of signaling events can result in spindle misorientation, which in turn can trigger developmental defects and cancer progression. Little is known about the cellular signaling pathway involved in the misorientation of proliferating cells that evade apoptosis after DNA damage. In this study, we found that perturbations to spindle orientation were induced in ultraviolet C (UVC)-irradiated surviving cells. N-terminal truncated Rho GDP-dissociation inhibitor β (RhoGDIβ), which is produced by UVC irradiation, distorted the spindle orientation of HeLa cells cultured on Matrigel. The short hairpin RNA-mediated knockdown of RhoGDIβ significantly attenuated UVC-induced misorientation. Subsequent expression of wild-type RhoGDIβ, but not a noncleavable mutant, RhoGDIβ (D19A), again led to a relative increase in spindle misorientation in response to UVC. Our findings revealed that RhoGDIβ impacts spindle orientation in response to DNA damage.
Collapse
Affiliation(s)
- Natsumi Doi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| | - Yuuki Kunimatsu
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| | - Kouhei Fujiura
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| | - Hiro Togari
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| | - Kenji Minagi
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| | - Koichi Nakaoji
- Research & Development Division, Pias Corporation, Kobe, Japan
| | - Kazuhiko Hamada
- Research & Development Division, Pias Corporation, Kobe, Japan
| | - Achim Temme
- Department of Neurosurgery, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Masaaki Tatsuka
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima, Japan
| |
Collapse
|
16
|
O'Neill RS, Schoborg TA, Rusan NM. Same but different: pleiotropy in centrosome-related microcephaly. Mol Biol Cell 2019; 29:241-246. [PMID: 29382806 PMCID: PMC5996963 DOI: 10.1091/mbc.e17-03-0192] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/22/2017] [Accepted: 11/30/2017] [Indexed: 01/19/2023] Open
Abstract
An intimate link between centrosome function and neurogenesis is revealed by the identification of many genes with centrosome-associated functions that are mutated in microcephaly disorders. Consistent with the major role of the centrosome in mitosis, mutations in these centrosome-related microcephaly (CRM) genes are thought to affect neurogenesis by depleting the pool of neural progenitor cells, primarily through apoptosis as a consequence of mitotic failure or premature differentiation as a consequence of cell cycle delay and randomization of spindle orientation. However, as suggested by the wide range of microcephaly phenotypes and the multifunctional nature of many CRM proteins, this picture of CRM gene function is incomplete. Here, we explore several examples of CRM genes pointing to additional functions that contribute to microcephaly, including regulation of cell cycle signaling, actin cytoskeleton, and Hippo pathway proteins, as well as functions in postmitotic neurons and glia. As these examples are likely just the tip of the iceberg, further exploration of the roles of microcephaly-related genes are certain to reveal additional unforeseen functions important for neurodevelopment.
Collapse
Affiliation(s)
- Ryan S O'Neill
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Todd A Schoborg
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Hauser-Kawaguchi A, Tolg C, Peart T, Milne M, Turley EA, Luyt LG. A truncated RHAMM protein for discovering novel therapeutic peptides. Bioorg Med Chem 2018; 26:5194-5203. [PMID: 30249497 DOI: 10.1016/j.bmc.2018.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
The receptor for hyaluronan mediated motility (RHAMM, gene name HMMR) belongs to a group of proteins that bind to hyaluronan (HA), a high-molecular weight anionic polysaccharide that has pro-angiogenic and inflammatory properties when fragmented. We propose to use a chemically synthesized, truncated version of the protein (706-767), 7 kDa RHAMM, as a target receptor in the screening of novel peptide-based therapeutic agents. Chemical synthesis by Fmoc-based solid-phase peptide synthesis, and optimization using pseudoprolines, results in RHAMM protein of higher purity and yield than synthesis by recombinant protein production. 7 kDa RHAMM was evaluated for its secondary structure, ability to bind the native ligand, HA, and its bioactivity. This 62-amino acid polypeptide replicates the HA binding properties of both native and recombinant RHAMM protein. Furthermore, tubulin-derived HA peptide analogues that bind to recombinant RHAMM and were previously reported to compete with HA for interactions with RHAMM, bind with a similar affinity and specificity to the 7 kDa RHAMM. Therefore, in terms of its key binding properties, the 7 kDa RHAMM mini-protein is a suitable replacement for the full-length recombinant protein.
Collapse
Affiliation(s)
| | - Cornelia Tolg
- Cancer Research Laboratory Program, Lawson Health Research Institute and London Regional Cancer Program, London Health Sciences Center, London, Ontario, Canada
| | - Teresa Peart
- Cancer Research Laboratory Program, Lawson Health Research Institute and London Regional Cancer Program, London Health Sciences Center, London, Ontario, Canada
| | - Mark Milne
- Cancer Research Laboratory Program, Lawson Health Research Institute and London Regional Cancer Program, London Health Sciences Center, London, Ontario, Canada
| | - Eva A Turley
- Cancer Research Laboratory Program, Lawson Health Research Institute and London Regional Cancer Program, London Health Sciences Center, London, Ontario, Canada; Department of Biochemistry, Western University, London, Ontario, Canada; Department of Oncology, Western University, London, Ontario, Canada
| | - Leonard G Luyt
- Department of Chemistry, Western University, London, Ontario, Canada; Cancer Research Laboratory Program, Lawson Health Research Institute and London Regional Cancer Program, London Health Sciences Center, London, Ontario, Canada; Department of Oncology, Western University, London, Ontario, Canada; Department of Medical Imaging, Western University, London, Ontario, Canada.
| |
Collapse
|
18
|
Connell M, Chen H, Jiang J, Kuan CW, Fotovati A, Chu TLH, He Z, Lengyell TC, Li H, Kroll T, Li AM, Goldowitz D, Frappart L, Ploubidou A, Patel MS, Pilarski LM, Simpson EM, Lange PF, Allan DW, Maxwell CA. HMMR acts in the PLK1-dependent spindle positioning pathway and supports neural development. eLife 2017; 6:e28672. [PMID: 28994651 PMCID: PMC5681225 DOI: 10.7554/elife.28672] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/05/2017] [Indexed: 01/08/2023] Open
Abstract
Oriented cell division is one mechanism progenitor cells use during development and to maintain tissue homeostasis. Common to most cell types is the asymmetric establishment and regulation of cortical NuMA-dynein complexes that position the mitotic spindle. Here, we discover that HMMR acts at centrosomes in a PLK1-dependent pathway that locates active Ran and modulates the cortical localization of NuMA-dynein complexes to correct mispositioned spindles. This pathway was discovered through the creation and analysis of Hmmr-knockout mice, which suffer neonatal lethality with defective neural development and pleiotropic phenotypes in multiple tissues. HMMR over-expression in immortalized cancer cells induces phenotypes consistent with an increase in active Ran including defects in spindle orientation. These data identify an essential role for HMMR in the PLK1-dependent regulatory pathway that orients progenitor cell division and supports neural development.
Collapse
Affiliation(s)
- Marisa Connell
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Helen Chen
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Jihong Jiang
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Chia-Wei Kuan
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverCanada
| | - Abbas Fotovati
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Tony LH Chu
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Zhengcheng He
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Tess C Lengyell
- Centre for Molecular Medicine and TherapeuticsUniversity of British ColumbiaVancouverCanada
| | - Huaibiao Li
- Leibniz Institute on Aging—Fritz Lipmann InstituteBeutenbergstrasseGermany
| | - Torsten Kroll
- Leibniz Institute on Aging—Fritz Lipmann InstituteBeutenbergstrasseGermany
| | - Amanda M Li
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
| | - Daniel Goldowitz
- Centre for Molecular Medicine and TherapeuticsUniversity of British ColumbiaVancouverCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverCanada
| | - Lucien Frappart
- Leibniz Institute on Aging—Fritz Lipmann InstituteBeutenbergstrasseGermany
| | - Aspasia Ploubidou
- Leibniz Institute on Aging—Fritz Lipmann InstituteBeutenbergstrasseGermany
| | - Millan S Patel
- Department of Medical GeneticsUniversity of British ColumbiaVancouverCanada
| | - Linda M Pilarski
- Cross Cancer Institute, Department of OncologyUniversity of AlbertaEdmontonCanada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and TherapeuticsUniversity of British ColumbiaVancouverCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverCanada
| | - Philipp F Lange
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverCanada
- Michael Cuccione Childhood Cancer Research ProgramBC Children’s HospitalVancouverCanada
| | - Douglas W Allan
- Department of Cellular and Physiological SciencesLife Sciences Centre, University of British ColumbiaVancouverCanada
| | - Christopher A Maxwell
- Department of PaediatricsUniversity of British ColumbiaVancouverCanada
- Michael Cuccione Childhood Cancer Research ProgramBC Children’s HospitalVancouverCanada
| |
Collapse
|