1
|
Ebrahimi AM, Gawlik W, Wojciechowski AM, Rajfur Z. Cell-particles interaction - selective uptake and transport of microdiamonds. Commun Biol 2024; 7:318. [PMID: 38480800 PMCID: PMC10937934 DOI: 10.1038/s42003-024-05974-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 02/26/2024] [Indexed: 03/17/2024] Open
Abstract
Diamond particles have recently emerged as novel agents in cellular studies because of their superb biocompatibility. Their unique characteristics, including small size and the presence of fluorescent color centers, stimulate many important applications. However, the mechanism of interaction between cells and diamond particles-uptake, transport, and final localization within cells-is not yet fully understood. Herein, we show a novel, to the best of our knowledge, cell behavior wherein cells actively target and uptake diamond particles rather than latex beads from their surroundings, followed by their active transport within cells. Furthermore, we demonstrate that myosin-X is involved in cell-particle interaction, while myosin-II does not participate in particle uptake and transport. These results can have important implications for drug delivery and improve sensing methods that use diamond particles.
Collapse
Affiliation(s)
- Armin M Ebrahimi
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348, Kraków, Poland
| | - Wojciech Gawlik
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland
| | - Adam M Wojciechowski
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland.
| | - Zenon Rajfur
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland.
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland.
| |
Collapse
|
2
|
Liu Q, Cheng C, Huang J, Yan W, Wen Y, Liu Z, Zhou B, Guo S, Fang W. MYH9: A key protein involved in tumor progression and virus-related diseases. Biomed Pharmacother 2024; 171:116118. [PMID: 38181716 DOI: 10.1016/j.biopha.2023.116118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of non-muscle myosin IIA (NMIIA), which belongs to the myosin II subfamily of actin-based molecular motors. Previous studies have demonstrated that abnormal expression and mutations of MYH9 were correlated with MYH9-related diseases and tumors. Furthermore, earlier investigations identified MYH9 as a tumor suppressor. However, subsequent research revealed that MYH9 promoted tumorigenesis, progression and chemoradiotherapy resistance. Note-worthily, MYH9 has also been linked to viral infections, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Epstein-Barr virus, and hepatitis B virus, as a receptor or co-receptor. In addition, MYH9 promotes the development of hepatocellular carcinoma by interacting with the hepatitis B virus-encoding X protein. Finally, various findings highlighted the role of MYH9 in the development of these illnesses, especially in tumors. This review summarizes the involvement of the MYH9-regulated signaling network in tumors and virus-related diseases and presents possible drug interventions on MYH9, providing insights for the use of MYH9 as a therapeutic target for tumors and virus-mediated diseases.
Collapse
Affiliation(s)
- Qing Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Chao Cheng
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology hospital, Shenzhen 518000, China
| | - Jiyu Huang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Weiwei Yan
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Yinhao Wen
- Department of Oncology, Pingxiang People's Hospital, Pingxiang 337000, China
| | - Zhen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; Key Laboratory of Protein Modification and Degradation, Basic School of Guangzhou Medical University, Guangzhou 510315, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou 525200, China.
| | - Suiqun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; The People's Hospital of Gaozhou, Gaozhou 525200, China; Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
3
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Mechanisms underlying Myosin 10's contribution to the maintenance of mitotic spindle bipolarity. Mol Biol Cell 2024; 35:ar14. [PMID: 38019611 PMCID: PMC10881153 DOI: 10.1091/mbc.e23-07-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Myosin 10 (Myo10) couples microtubules and integrin-based adhesions to movement along actin filaments via its microtubule-binding MyTH4 domain and integrin-binding FERM domain, respectively. Here we show that Myo10-depleted HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in Myo10-depleted MEFs and in Myo10-depleted HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates y-tubulin-positive acentriolar foci that serve as extra spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both microtubules and integrins to promote PCM/pole integrity. Conversely, Myo10 only needs interact with integrins to promote supernumerary centrosome clustering. Importantly, images of metaphase Halo-Myo10 knockin cells show that the myosin localizes exclusively to the spindle and the tips of adhesive retraction fibers. We conclude that Myo10 promotes PCM/pole integrity in part by interacting with spindle microtubules, and that it promotes supernumerary centrosome clustering by supporting retraction fiber-based cell adhesion, which likely serves to anchor the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
4
|
Szabó-Meleg E. Intercellular Highways in Transport Processes. Results Probl Cell Differ 2024; 73:173-201. [PMID: 39242380 DOI: 10.1007/978-3-031-62036-2_9] [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: 09/09/2024]
Abstract
Communication among cells is vital in multicellular organisms. Various structures and mechanisms have evolved over time to achieve the intricate flow of material and information during this process. One such way of communication is through tunnelling membrane nanotubes (TNTs), which were initially described in 2004. These TNTs are membrane-bounded actin-rich cellular extensions, facilitating direct communication between distant cells. They exhibit remarkable diversity in terms of structure, morphology, and function, in which cytoskeletal proteins play an essential role. Biologically, TNTs play a crucial role in transporting membrane components, cell organelles, and nucleic acids, and they also present opportunities for the efficient transmission of bacteria and viruses, furthermore, may contribute to the dissemination of misfolded proteins in certain neurodegenerative diseases. Convincing results of studies conducted both in vitro and in vivo indicate that TNTs play roles in various biomedical processes, including cell differentiation, tissue regeneration, neurodegenerative diseases, immune response and function, as well as tumorigenesis.
Collapse
Affiliation(s)
- Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
5
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Myosin 10 uses its MyTH4 and FERM domains differentially to support two aspects of spindle pole biology required for mitotic spindle bipolarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545002. [PMID: 37398378 PMCID: PMC10312724 DOI: 10.1101/2023.06.15.545002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myosin 10 (Myo10) has the ability to link actin filaments to integrin-based adhesions and to microtubules by virtue of its integrin-binding FERM domain and microtubule-binding MyTH4 domain, respectively. Here we used Myo10 knockout cells to define Myo10's contribution to the maintenance of spindle bipolarity, and complementation to quantitate the relative contributions of its MyTH4 and FERM domains. Myo10 knockout HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in knockout MEFs and knockout HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates γ-tubulin-positive acentriolar foci that serve as additional spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both integrins and microtubules to promote PCM/pole integrity. Conversely, Myo10's ability to promote the clustering of supernumerary centrosomes only requires that it interact with integrins. Importantly, images of Halo-Myo10 knock-in cells show that the myosin localizes exclusively within adhesive retraction fibers during mitosis. Based on these and other results, we conclude that Myo10 promotes PCM/pole integrity at a distance, and that it facilitates supernumerary centrosome clustering by promoting retraction fiber-based cell adhesion, which likely provides an anchor for the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Tolue Ghasaban F, Maharati A, Akhlaghipour I, Moghbeli M. MicroRNAs as the critical regulators of autophagy-mediated cisplatin response in tumor cells. Cancer Cell Int 2023; 23:80. [PMID: 37098542 PMCID: PMC10127417 DOI: 10.1186/s12935-023-02925-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 04/27/2023] Open
Abstract
Chemotherapy is one of the most common therapeutic methods in advanced and metastatic tumors. Cisplatin (CDDP) is considered as one of the main first-line chemotherapy drugs in solid tumors. However, there is a high rate of CDDP resistance in cancer patients. Multi-drug resistance (MDR) as one of the main therapeutic challenges in cancer patients is associated with various cellular processes such as drug efflux, DNA repair, and autophagy. Autophagy is a cellular mechanism that protects the tumor cells toward the chemotherapeutic drugs. Therefore, autophagy regulatory factors can increase or decrease the chemotherapy response in tumor cells. MicroRNAs (miRNAs) have a pivotal role in regulation of autophagy in normal and tumor cells. Therefore, in the present review, we discussed the role of miRNAs in CDDP response through the regulation of autophagy. It has been reported that miRNAs mainly increased the CDDP sensitivity in tumor cells by inhibition of autophagy. PI3K/AKT signaling pathway and autophagy-related genes (ATGs) were the main targets of miRNAs in the regulation of autophagy-mediated CDDP response in tumor cells. This review can be an effective step to introduce the miRNAs as efficient therapeutic options to increase autophagy-mediated CDDP sensitivity in tumor cells.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Fuchs J, Bareesel S, Kroon C, Polyzou A, Eickholt BJ, Leondaritis G. Plasma membrane phospholipid phosphatase-related proteins as pleiotropic regulators of neuron growth and excitability. Front Mol Neurosci 2022; 15:984655. [PMID: 36187351 PMCID: PMC9520309 DOI: 10.3389/fnmol.2022.984655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neuronal plasma membrane proteins are essential for integrating cell extrinsic and cell intrinsic signals to orchestrate neuronal differentiation, growth and plasticity in the developing and adult nervous system. Here, we shed light on the family of plasma membrane proteins phospholipid phosphatase-related proteins (PLPPRs) (alternative name, PRGs; plasticity-related genes) that fine-tune neuronal growth and synaptic transmission in the central nervous system. Several studies uncovered essential functions of PLPPRs in filopodia formation, axon guidance and branching during nervous system development and regeneration, as well as in the control of dendritic spine number and excitability. Loss of PLPPR expression in knockout mice increases susceptibility to seizures, and results in defects in sensory information processing, development of psychiatric disorders, stress-related behaviors and abnormal social interaction. However, the exact function of PLPPRs in the context of neurological diseases is largely unclear. Although initially described as active lysophosphatidic acid (LPA) ecto-phosphatases that regulate the levels of this extracellular bioactive lipid, PLPPRs lack catalytic activity against LPA. Nevertheless, they emerge as atypical LPA modulators, by regulating LPA mediated signaling processes. In this review, we summarize the effects of this protein family on cellular morphology, generation and maintenance of cellular protrusions as well as highlight their known neuronal functions and phenotypes of KO mice. We discuss the molecular mechanisms of PLPPRs including the deployment of phospholipids, actin-cytoskeleton and small GTPase signaling pathways, with a focus on identifying gaps in our knowledge to stimulate interest in this understudied protein family.
Collapse
Affiliation(s)
- Joachim Fuchs
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shannon Bareesel
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Cristina Kroon
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexandra Polyzou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Britta J. Eickholt
- Institute of Molecular Biology and Biochemistry, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- *Correspondence: Britta J. Eickholt,
| | - George Leondaritis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Institute of Biosciences, University Research Center Ioannina, University of Ioannina, Ioannina, Greece
- George Leondaritis,
| |
Collapse
|
8
|
Kim SJ, Sun EG, Bae JA, Park S, Hong C, Park Z, Kim H, Kim KK. A peptide interfering with the dimerization of oncogenic KITENIN protein and its stability suppresses colorectal tumour progression. Clin Transl Med 2022; 12:e871. [PMID: 35853101 PMCID: PMC9296036 DOI: 10.1002/ctm2.871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 12/22/2022] Open
Abstract
The stability of a protein, as well as its function and versatility, can be enhanced through oligomerization. KITENIN (KAI1 C-terminal interacting tetraspanin) is known to promote the malignant progression of colorectal cancer (CRC). How KITENIN maintains its structural integrity and stability are largely unknown, however. Here we investigated the mechanisms regulating the stability of KITENIN with the aim of developing therapeutics blocking its oncogenic functions. We found that KITENIN formed a homo-oligomeric complex and that the intracellular C-terminal domain (KITENIN-CTD) was needed for this oligomerization. Expression of the KITENIN-CTD alone interfered with the formation of the KITENIN homodimer, and the amino acid sequence from 463 to 471 within the KITENIN-CTD was the most effective. This sequence coupled with a cell-penetrating peptide was named a KITENIN dimerization-interfering peptide (KDIP). We next studied the mechanisms by which KDIP affected the stability of KITENIN. The KITENIN-interacting protein myosin-X (Myo10), which has oncogenic activity in several cancers, functioned as an effector to stabilize the KITENIN homodimer in the cis formation. Treatment with KDIP resulted in the disintegration of the homodimer via downregulation of Myo10, which led to increased binding of RACK1 to the exposed RACK1-interacting motif (463-471 aa), and subsequent autophagy-dependent degradation of KITENIN and reduced CRC cell invasion. Intravenous injection of KDIP significantly reduced the tumour burden in a syngeneic mouse tumour model and colorectal liver metastasis in an intrasplenic hepatic metastasis model. Collectively, our present results provide a new cancer therapeutic peptide for blocking colorectal liver metastasis, which acts by inducing the downregulation of Myo10 and specifically targeting the stability of the oncogenic KITENIN protein.
Collapse
Affiliation(s)
- Sung Jin Kim
- Department of PharmacologyChonnam National University Medical SchoolGwangjuRepublic of Korea
- College of PharmacySunchon National UniversitySuncheonRepublic of Korea
| | - Eun Gene Sun
- Department of PharmacologyChonnam National University Medical SchoolGwangjuRepublic of Korea
| | - Jeong A Bae
- Department of PharmacologyChonnam National University Medical SchoolGwangjuRepublic of Korea
| | - Sehoon Park
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Chang‐Soo Hong
- Department of PharmacologyChonnam National University Medical SchoolGwangjuRepublic of Korea
| | - Zee‐Yong Park
- School of Life SciencesGwangju Institute of Science and TechnologyGwangjuRepublic of Korea
| | - Hangun Kim
- College of PharmacySunchon National UniversitySuncheonRepublic of Korea
| | - Kyung Keun Kim
- Department of PharmacologyChonnam National University Medical SchoolGwangjuRepublic of Korea
| |
Collapse
|
9
|
Application of piconewton forces to individual filopodia reveals mechanosensory role of L-type Ca 2+ channels. Biomaterials 2022; 284:121477. [PMID: 35395455 DOI: 10.1016/j.biomaterials.2022.121477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/18/2022] [Indexed: 11/02/2022]
Abstract
Filopodia are ubiquitous membrane projections that play crucial role in guiding cell migration on rigid substrates and through extracellular matrix by utilizing yet unknown mechanosensing molecular pathways. As recent studies show that Ca2+ channels localized to filopodia play an important role in regulation of their formation and since some Ca2+ channels are known to be mechanosensitive, force-dependent activity of filopodial Ca2+ channels might be linked to filopodia's mechanosensing function. We tested this hypothesis by monitoring changes in the intra-filopodial Ca2+ level in response to application of stretching force to individual filopodia of several cell types using optical tweezers. Results show that stretching forces of tens of pN strongly promote Ca2+ influx into filopodia, causing persistent Ca2+ oscillations that last for minutes even after the force is released. Several known mechanosensitive Ca2+ channels, such as Piezo 1, Piezo 2 and TRPV4, were found to be dispensable for the observed force-dependent Ca2+ influx, while L-type Ca2+ channels appear to be a key player in the discovered phenomenon. As previous studies have shown that intra-filopodial transient Ca2+ signals play an important role in guidance of cell migration, our results suggest that the force-dependent activation of L-type Ca2+ channels may contribute to this process. Overall, our study reveals an intricate interplay between mechanical forces and Ca2+ signaling in filopodia, providing novel mechanistic insights for the force-dependent filopodia functions in guidance of cell migration.
Collapse
|
10
|
Tyrmi JS, Arffman RK, Pujol-Gualdo N, Kurra V, Morin-Papunen L, Sliz E, Piltonen TT, Laisk T, Kettunen J, Laivuori H. Leveraging Northern European population history: novel low-frequency variants for polycystic ovary syndrome. Hum Reprod 2022; 37:352-365. [PMID: 34791234 PMCID: PMC8804330 DOI: 10.1093/humrep/deab250] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/07/2021] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION Can we identify novel variants associated with polycystic ovary syndrome (PCOS) by leveraging the unique population history of Northern Europe? SUMMARY ANSWER We identified three novel genome-wide significant associations with PCOS, with two putative independent causal variants in the checkpoint kinase 2 (CHEK2) gene and a third in myosin X (MYO10). WHAT IS KNOWN ALREADY PCOS is a common, complex disorder with unknown aetiology. While previous genome-wide association studies (GWAS) have mapped several loci associated with PCOS, the analysis of populations with unique population history and genetic makeup has the potential to uncover new low-frequency variants with larger effects. STUDY DESIGN, SIZE, DURATION A population-based case-control GWAS was carried out. PARTICIPANTS/MATERIALS, SETTING, METHODS We identified PCOS cases from national registers by ICD codes (ICD-10 E28.2, ICD-9 256.4, or ICD-8 256.90), and all remaining women were considered controls. We then conducted a three-stage case-control GWAS: in the discovery phase, we had a total of 797 cases and 140 558 controls from the FinnGen study. For validation, we used an independent dataset from the Estonian Biobank, including 2812 cases and 89 230 controls. Finally, we performed a joint meta-analysis of 3609 cases and 229 788 controls from both cohorts. Additionally, we reran the association analyses including BMI as a covariate, with 2169 cases and 160 321 controls from both cohorts. MAIN RESULTS AND THE ROLE OF CHANCE Two out of the three novel genome-wide significant variants associating with PCOS, rs145598156 (P = 3.6×10-8, odds ratio (OR) = 3.01 [2.02-4.50] minor allele frequency (MAF) = 0.005) and rs182075939 (P = 1.9×10-16, OR = 1.69 [1.49-1.91], MAF = 0.04), were found to be enriched in the Finnish and Estonian populations and are tightly linked to a deletion c.1100delC (r2 = 0.95) and a missense I157T (r2 = 0.83) in CHEK2. The third novel association is a common variant near MYO10 (rs9312937, P = 1.7 × 10-8, OR = 1.16 [1.10-1.23], MAF = 0.44). We also replicated four previous reported associations near the genes Erb-B2 Receptor Tyrosine Kinase 4 (ERBB4), DENN Domain Containing 1A (DENND1A), FSH Subunit Beta (FSHB) and Zinc Finger And BTB Domain Containing 16 (ZBTB16). When adding BMI as a covariate only one of the novel variants remained genome-wide significant in the meta-analysis (the EstBB lead signal in CHEK2 rs182075939, P = 1.9×10-16, OR = 1.74 [1.5-2.01]) possibly owing to reduced sample size. LARGE SCALE DATA The age- and BMI-adjusted GWAS meta-analysis summary statistics are available for download from the GWAS Catalog with accession numbers GCST90044902 and GCST90044903. LIMITATIONS, REASONS FOR CAUTION The main limitation was the low prevalence of PCOS in registers; however, the ones with the diagnosis most likely represent the most severe cases. Also, BMI data were not available for all (63% for FinnGen, 76% for EstBB), and the biobank setting limited the accessibility of PCOS phenotypes and laboratory values. WIDER IMPLICATIONS OF THE FINDINGS This study encourages the use of isolated populations to perform genetic association studies for the identification of rare variants contributing to the genetic landscape of complex diseases such as PCOS. STUDY FUNDING/COMPETING INTEREST(S) This work has received funding from the European Union's Horizon 2020 research and innovation programme under the MATER Marie Skłodowska-Curie grant agreement No. 813707 (N.P.-G., T.L., T.P.), the Estonian Research Council grant (PRG687, T.L.), the Academy of Finland grants 315921 (T.P.), 321763 (T.P.), 297338 (J.K.), 307247 (J.K.), 344695 (H.L.), Novo Nordisk Foundation grant NNF17OC0026062 (J.K.), the Sigrid Juselius Foundation project grants (T.L., J.K., T.P.), Finska Läkaresällskapet (H.L.) and Jane and Aatos Erkko Foundation (H.L.). The funders had no role in study design, data collection and analysis, publishing or preparation of the manuscript. The authors declare no conflicts of interest.
Collapse
Affiliation(s)
- Jaakko S Tyrmi
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Natàlia Pujol-Gualdo
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Venla Kurra
- Department of Clinical Genetics, Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Eeva Sliz
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Hannele Laivuori
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
11
|
Aliyu IA, Kumurya AS, Bala JA, Yahaya H, Saidu H. Proteomes, kinases and signalling pathways in virus-induced filopodia, as potential antiviral therapeutics targets. Rev Med Virol 2021; 31:1-9. [PMID: 33314425 PMCID: PMC7883202 DOI: 10.1002/rmv.2202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Filopodia are thin finger-like protrusions at the surface of cells that are internally occupied with bundles of tightly parallel actin filaments. They play significant roles in cellular physiological processes, such as adhesion to extracellular matrix, guidance towards chemo-attractants and in wound healing. Filopodia were recently reported to play important roles in viral infection including initial viral attachment to host cells, cell surfing, viral trafficking, internalization, budding, virus release and spread to other cells in a form that would avoid the host immune system. The detailed virus-host protein interactions underlying most of these processes remain to be elucidated. This review will describe some reported virus-host protein interactions on filopodia with the aim of identifying potential new anti-virus therapeutic targets. Exploring this research area may lead to the development of novel classes of anti-viral therapeutics that can block signalling pathways used by the virus to trigger filopodia formation. Successful compounds would inhibit initial virus attachment, formation of filopodia, expression of putative virus binding protein, extracellular virus trafficking, and budding.
Collapse
Affiliation(s)
- Isah Abubakar Aliyu
- Department of Medical Laboratory ScienceFaculty of Allied Health SciencesCollege of Health SciencesBayero University KanoKano StateNigeria
| | - Abdulhadi Sale Kumurya
- Department of Medical Laboratory ScienceFaculty of Allied Health SciencesCollege of Health SciencesBayero University KanoKano StateNigeria
| | - Jamilu Abubakar Bala
- Department of Medical Laboratory ScienceFaculty of Allied Health SciencesCollege of Health SciencesBayero University KanoKano StateNigeria
- Virology UnitDepartment of Pathology and MicrobiologyFaculty of Veterinary MedicineUniversity Putra MalaysiaSelangorMalaysia
| | - Hassan Yahaya
- Department of Medical Microbiology and ParasitologyFaculty of Medicine and Health ScienceUniversity Putra MalaysiaSelangorMalaysia
- Department of Medical Laboratory ScienceFaculty of Allied Health SciencesCollege of Health SciencesBayero University KanoKano StateNigeria
| | - Hayatu Saidu
- Department of Medical Laboratory ScienceFaculty of Allied Health SciencesCollege of Health SciencesBayero University KanoKano StateNigeria
| |
Collapse
|
12
|
Arthur AL, Crawford A, Houdusse A, Titus MA. VASP-mediated actin dynamics activate and recruit a filopodia myosin. eLife 2021; 10:68082. [PMID: 34042588 PMCID: PMC8352590 DOI: 10.7554/elife.68082] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Filopodia are thin, actin-based structures that cells use to interact with their environments. Filopodia initiation requires a suite of conserved proteins but the mechanism remains poorly understood. The actin polymerase VASP and a MyTH-FERM (MF) myosin, DdMyo7 in amoeba, are essential for filopodia initiation. DdMyo7 is localized to dynamic regions of the actin-rich cortex. Analysis of VASP mutants and treatment of cells with anti-actin drugs shows that myosin recruitment and activation in Dictyostelium requires localized VASP-dependent actin polymerization. Targeting of DdMyo7 to the cortex alone is not sufficient for filopodia initiation; VASP activity is also required. The actin regulator locally produces a cortical actin network that activates myosin and together they shape the actin network to promote extension of parallel bundles of actin during filopodia formation. This work reveals how filopodia initiation requires close collaboration between an actin-binding protein, the state of the actin cytoskeleton and MF myosin activity.
Collapse
Affiliation(s)
- Ashley L Arthur
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Amy Crawford
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, Paris, France
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| |
Collapse
|
13
|
Cordero Cervantes D, Zurzolo C. Peering into tunneling nanotubes-The path forward. EMBO J 2021; 40:e105789. [PMID: 33646572 PMCID: PMC8047439 DOI: 10.15252/embj.2020105789] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/21/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
The identification of Tunneling Nanotubes (TNTs) and TNT-like structures signified a critical turning point in the field of cell-cell communication. With hypothesized roles in development and disease progression, TNTs' ability to transport biological cargo between distant cells has elevated these structures to a unique and privileged position among other mechanisms of intercellular communication. However, the field faces numerous challenges-some of the most pressing issues being the demonstration of TNTs in vivo and understanding how they form and function. Another stumbling block is represented by the vast disparity in structures classified as TNTs. In order to address this ambiguity, we propose a clear nomenclature and provide a comprehensive overview of the existing knowledge concerning TNTs. We also discuss their structure, formation-related pathways, biological function, as well as their proposed role in disease. Furthermore, we pinpoint gaps and dichotomies found across the field and highlight unexplored research avenues. Lastly, we review the methods employed to date and suggest the application of new technologies to better understand these elusive biological structures.
Collapse
Affiliation(s)
| | - Chiara Zurzolo
- Institut PasteurMembrane Traffic and PathogenesisParisFrance
| |
Collapse
|
14
|
Ramirez I, Gholkar AA, Velasquez EF, Guo X, Tofig B, Damoiseaux R, Torres JZ. The myosin regulatory light chain Myl5 localizes to mitotic spindle poles and is required for proper cell division. Cytoskeleton (Hoboken) 2021; 78:23-35. [PMID: 33641240 DOI: 10.1002/cm.21654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/18/2022]
Abstract
Myosins are ATP-dependent actin-based molecular motors critical for diverse cellular processes like intracellular trafficking, cell motility, and cell invasion. During cell division, myosin MYO10 is important for proper mitotic spindle assembly, the anchoring of the spindle to the cortex, and positioning of the spindle to the cell mid-plane. However, myosins are regulated by myosin regulatory light chains (RLCs), and whether RLCs are important for cell division has remained unexplored. Here, we have determined that the previously uncharacterized myosin RLC Myl5 associates with the mitotic spindle and is required for cell division. We show that Myl5 localizes to the leading edge and filopodia during interphase and to mitotic spindle poles and spindle microtubules during early mitosis. Importantly, depletion of Myl5 led to defects in mitotic spindle assembly, chromosome congression, and chromosome segregation and to a slower transition through mitosis. Furthermore, Myl5 bound to MYO10 in vitro and co-localized with MYO10 at the spindle poles. These results suggest that Myl5 is important for cell division and that it may be performing its function through MYO10.
Collapse
Affiliation(s)
- Ivan Ramirez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Ankur A Gholkar
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Erick F Velasquez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Xiao Guo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Bobby Tofig
- California NanoSystems Institute, Los Angeles, California, USA
| | - Robert Damoiseaux
- California NanoSystems Institute, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, Los Angeles, California, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
15
|
The Expressions and Mechanisms of Sarcomeric Proteins in Cancers. DISEASE MARKERS 2020; 2020:8885286. [PMID: 32670437 PMCID: PMC7346232 DOI: 10.1155/2020/8885286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
The sarcomeric proteins control the movement of cells in diverse species, whereas the deregulation can induce tumours in model organisms and occurs in human carcinomas. Sarcomeric proteins are recognized as oncogene and related to tumor cell metastasis. Recent insights into their expressions and functions have led to new cancer therapeutic opportunities. In this review, we appraise the evidence for the sarcomeric proteins as cancer genes and discuss cancer-relevant biological functions, potential mechanisms by which sarcomeric proteins activity is altered in cancer.
Collapse
|
16
|
Wolf KJ, Shukla P, Springer K, Lee S, Coombes JD, Choy CJ, Kenny SJ, Xu K, Kumar S. A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles. Proc Natl Acad Sci U S A 2020; 117:11432-11443. [PMID: 32381732 PMCID: PMC7261014 DOI: 10.1073/pnas.1914294117] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The structure and mechanics of many connective tissues are dictated by a collagen-rich extracellular matrix (ECM), where collagen fibers provide topological cues that direct cell migration. However, comparatively little is known about how cells navigate the hyaluronic acid (HA)-rich, nanoporous ECM of the brain, a problem with fundamental implications for development, inflammation, and tumor invasion. Here, we demonstrate that glioblastoma cells adhere to and invade HA-rich matrix using microtentacles (McTNs), which extend tens of micrometers from the cell body and are distinct from filopodia. We observe these structures in continuous culture models and primary patient-derived tumor cells, as well as in synthetic HA matrix and organotypic brain slices. High-magnification and superresolution imaging reveals McTNs are dynamic, CD44-coated tubular protrusions containing microtubules and actin filaments, which respectively drive McTN extension and retraction. Molecular mechanistic studies reveal that McTNs are stabilized by an interplay between microtubule-driven protrusion, actomyosin-driven retraction, and CD44-mediated adhesion, where adhesive and cytoskeletal components are mechanistically coupled by an IQGAP1-CLIP170 complex. McTNs represent a previously unappreciated mechanism through which cells engage nanoporous HA matrix and may represent an important molecular target in physiology and disease.
Collapse
Affiliation(s)
- Kayla J Wolf
- University of California, Berkeley-University of California San Francisco Graduate Program in Bioengineering, Department of Bioengineering, University of California, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Poojan Shukla
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Kelsey Springer
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Stacey Lee
- University of California, Berkeley-University of California San Francisco Graduate Program in Bioengineering, Department of Bioengineering, University of California, Berkeley, CA 94720
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Jason D Coombes
- Department of Bioengineering, University of California, Berkeley, CA, 94720
- Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom, SE5 9NU
| | - Caleb J Choy
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Samuel J Kenny
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, CA 94720
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Sanjay Kumar
- University of California, Berkeley-University of California San Francisco Graduate Program in Bioengineering, Department of Bioengineering, University of California, Berkeley, CA 94720;
- Department of Bioengineering, University of California, Berkeley, CA, 94720
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720
| |
Collapse
|
17
|
Barger SR, Gauthier NC, Krendel M. Squeezing in a Meal: Myosin Functions in Phagocytosis. Trends Cell Biol 2019; 30:157-167. [PMID: 31836280 DOI: 10.1016/j.tcb.2019.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
Phagocytosis is a receptor-mediated, actin-dependent process of internalization of large extracellular particles, such as pathogens or apoptotic cells. Engulfment of phagocytic targets requires the activity of myosins, actin-dependent molecular motors, which perform a variety of functions at distinct steps during phagocytosis. By applying force to actin filaments, the plasma membrane, and intracellular proteins and organelles, myosins can generate contractility, directly regulate actin assembly to ensure proper phagocytic internalization, and translocate phagosomes or other cargo to appropriate cellular locations. Recent studies using engineered microenvironments and phagocytic targets have demonstrated how altering the actomyosin cytoskeleton affects phagocytic behavior. Here, we discuss how studies using genetic and biochemical manipulation of myosins, force measurement techniques, and live-cell imaging have advanced our understanding of how specific myosins function at individual steps of phagocytosis.
Collapse
Affiliation(s)
- Sarah R Barger
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | - Mira Krendel
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
18
|
Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20163913. [PMID: 31408934 PMCID: PMC6719161 DOI: 10.3390/ijms20163913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recent findings have revealed the role of membrane traffic in the signaling of transforming growth factor-β (TGF-β). These findings originate from the pivotal function of TGF-β in development, cell proliferation, tumor metastasis, and many other processes essential in malignancy. Actin and unconventional myosin have crucial roles in subcellular trafficking of receptors; research has also revealed a growing number of unconventional myosins that have crucial roles in TGF-β signaling. Unconventional myosins modulate the spatial organization of endocytic trafficking and tether membranes or transport them along the actin cytoskeletons. Current models do not fully explain how membrane traffic forms a bridge between TGF-β and the downstream effectors that produce its functional responsiveness, such as cell migration. In this review, we present a brief overview of the current knowledge of the TGF-β signaling pathway and the molecular components that comprise the core pathway as follows: ligands, receptors, and Smad mediators. Second, we highlight key role(s) of myosin motor-mediated protein trafficking and membrane domain segregation in the modulation of the TGF-β signaling pathway. Finally, we review future challenges and provide future prospects in this field.
Collapse
|
19
|
Sun YY, Yang YF, Keller KE. Myosin-X Silencing in the Trabecular Meshwork Suggests a Role for Tunneling Nanotubes in Outflow Regulation. Invest Ophthalmol Vis Sci 2019; 60:843-851. [PMID: 30807639 PMCID: PMC6390986 DOI: 10.1167/iovs.18-26055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose The actin cytoskeleton plays a key role in outflow regulation through the trabecular meshwork (TM). Although actin stress fibers are a target of glaucoma therapies, the role of other actin cellular structures is unclear. Myosin-X (Myo10) is an actin-binding protein that is involved in tunneling nanotube (TNT) and filopodia formation. Here, we inhibited Myo10 pharmacologically or by gene silencing to investigate the role of filopodia/TNTs in the TM. Methods Short hairpin RNA interference (RNAi) silencing lentivirus targeting myosin-X (shMyo10) was generated. Human anterior segments were perfused with shMyo10 or CK-666, an Arp2/3 inhibitor. Confocal microscopy investigated the colocalization of Myo10 with matrix metalloproteinase (MMPs). Western immunoblotting investigated the protein levels of MMPs and extracellular matrix (ECM) proteins. MMP activity and phagocytosis assays were performed. Results CK-666 and shMyo10-silencing lentivirus caused a significant reduction in outflow rates in anterior segment perfusion culture, an ex vivo method to study intraocular pressure regulation. In human TM cells, Myo10 colocalized with MMP2, MMP14, and cortactin in podosome-like structures, which function as regions of focal ECM degradation. Furthermore, MMP activity, thrombospondin-1 and SPARC protein levels were significantly reduced in the media of CK-666-treated and shMyo10-silenced TM cells. However, neither Myo10 silencing or CK-666 treatment significantly affected phagocytic uptake. Conclusions Inhibiting filopodia/TNTs caused opposite effects on outflow compared with inhibiting stress fibers. Moreover, Myo10 may also play a role in focal ECM degradation in TM cells. Our results provide additional insight into the function of actin supramolecular assemblies and actin-binding proteins in outflow regulation.
Collapse
Affiliation(s)
- Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
20
|
Caporizzo MA, Fishman CE, Sato O, Jamiolkowski RM, Ikebe M, Goldman YE. The Antiparallel Dimerization of Myosin X Imparts Bundle Selectivity for Processive Motility. Biophys J 2019; 114:1400-1410. [PMID: 29590597 DOI: 10.1016/j.bpj.2018.01.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Myosin X is an unconventional actin-based molecular motor involved in filopodial formation, microtubule-actin filament interaction, and cell migration. Myosin X is an important component of filopodia regulation, localizing to tips of growing filopodia by an unclear targeting mechanism. The native α-helical dimerization domain of myosin X is thought to associate with antiparallel polarity of the two amino acid chains, making myosin X the only myosin that is currently considered to form antiparallel dimers. This study aims to determine if antiparallel dimerization of myosin X imparts selectivity toward actin bundles by comparing the motility of parallel and antiparallel dimers of myosin X on single and fascin-bundled actin filaments. Antiparallel myosin X dimers exhibit selective processivity on fascin-bundled actin and are only weakly processive on single actin filaments below saturating [ATP]. Artificial forced parallel dimers of myosin X are robustly processive on both single and bundled actin, exhibiting no selectivity. To determine the relationship between gating of the reaction steps and observed differences in motility, a mathematical model was developed to correlate the parameters of motility with the biochemical and mechanical kinetics of the dimer. Results from the model, constrained by experimental data, suggest that the probability of binding forward, toward the barbed end of the actin filament, is lower in antiparallel myosin X on single actin filaments compared to fascin-actin bundles and compared to constructs of myosin X with parallel dimerization.
Collapse
Affiliation(s)
- Matthew A Caporizzo
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Claire E Fishman
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Osamu Sato
- Department of Cellular and Molecular Biology, University of Texas Science Center, Tyler, Texas
| | - Ryan M Jamiolkowski
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas Science Center, Tyler, Texas
| | - Yale E Goldman
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
21
|
Bachg AC, Horsthemke M, Skryabin BV, Klasen T, Nagelmann N, Faber C, Woodham E, Machesky LM, Bachg S, Stange R, Jeong HW, Adams RH, Bähler M, Hanley PJ. Phenotypic analysis of Myo10 knockout (Myo10 tm2/tm2) mice lacking full-length (motorized) but not brain-specific headless myosin X. Sci Rep 2019; 9:597. [PMID: 30679680 PMCID: PMC6345916 DOI: 10.1038/s41598-018-37160-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023] Open
Abstract
We investigated the physiological functions of Myo10 (myosin X) using Myo10 reporter knockout (Myo10tm2) mice. Full-length (motorized) Myo10 protein was deleted, but the brain-specific headless (Hdl) isoform (Hdl-Myo10) was still expressed in homozygous mutants. In vitro, we confirmed that Hdl-Myo10 does not induce filopodia, but it strongly localized to the plasma membrane independent of the MyTH4-FERM domain. Filopodia-inducing Myo10 is implicated in axon guidance and mice lacking the Myo10 cargo protein DCC (deleted in colorectal cancer) have severe commissural defects, whereas MRI (magnetic resonance imaging) of isolated brains revealed intact commissures in Myo10tm2/tm2 mice. However, reminiscent of Waardenburg syndrome, a neural crest disorder, Myo10tm2/tm2 mice exhibited pigmentation defects (white belly spots) and simple syndactyly with high penetrance (>95%), and 24% of mutant embryos developed exencephalus, a neural tube closure defect. Furthermore, Myo10tm2/tm2 mice consistently displayed bilateral persistence of the hyaloid vasculature, revealed by MRI and retinal whole-mount preparations. In principle, impaired tissue clearance could contribute to persistence of hyaloid vasculature and syndactyly. However, Myo10-deficient macrophages exhibited no defects in the phagocytosis of apoptotic or IgG-opsonized cells. RNA sequence analysis showed that Myo10 was the most strongly expressed unconventional myosin in retinal vascular endothelial cells and expression levels increased 4-fold between P6 and P15, when vertical sprouting angiogenesis gives rise to deeper layers. Nevertheless, imaging of isolated adult mutant retinas did not reveal vascularization defects. In summary, Myo10 is important for both prenatal (neural tube closure and digit formation) and postnatal development (hyaloid regression, but not retinal vascularization).
Collapse
Affiliation(s)
- Anne C Bachg
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Markus Horsthemke
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Boris V Skryabin
- Department of Medicine, Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Tim Klasen
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Nina Nagelmann
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Emma Woodham
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Laura M Machesky
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Sandra Bachg
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Martin Bähler
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Peter J Hanley
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany.
| |
Collapse
|
22
|
He K, Sakai T, Tsukasaki Y, Watanabe TM, Ikebe M. Myosin X is recruited to nascent focal adhesions at the leading edge and induces multi-cycle filopodial elongation. Sci Rep 2017; 7:13685. [PMID: 29057977 PMCID: PMC5651867 DOI: 10.1038/s41598-017-06147-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
Filopodia protrude from the leading edge of cells and play important roles in cell motility. Here we report the mechanism of myosin X (encoded by Myo10)-induced multi-cycle filopodia extension. We found that actin, Arp2/3, vinculin and integrin-β first accumulated at the cell's leading edge. Myosin X was then gathered at these sites, gradually clustered by lateral movement, and subsequently initiated filopodia formation. During filopodia extension, we found the translocation of Arp2/3 and integrin-β along filopodia. Arp2/3 and integrin-β then became localized at the tip of filopodia, from where myosin X initiated the second extension of filopodia with a change in extension direction, thus producing long filopodia. Elimination of integrin-β, Arp2/3 and vinculin by siRNA significantly attenuated the myosin-X-induced long filopodia formation. We propose the following mechanism. Myosin X accumulates at nascent focal adhesions at the cell's leading edge, where myosin X promotes actin convergence to create the base of filopodia. Then myosin X moves to the filopodia tip and attracts integrin-β and Arp2/3 for further actin nucleation. The tip-located myosin X then initiates the second cycle of filopodia elongation to produce the long filopodia.
Collapse
Affiliation(s)
- Kangmin He
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.,Graduate School of Frontier Biosciences, Osaka University, Osaka, 5650871, Japan.,Department of Cell Biology, Harvard Medical School, and Cellular and Molecular Medicine Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA
| | - Yoshikazu Tsukasaki
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA.,Laboratory for Comprehensive Bioimaging, RIKEN Quantitative Biology Center (QBiC), Osaka, 5650874, Japan.,Department of Pharmacology, University of Illinois Chicago College of Medicine, Boston Children's Hospital, Chicago, Illinois 60612, USA
| | - Tomonobu M Watanabe
- Graduate School of Frontier Biosciences, Osaka University, Osaka, 5650871, Japan. .,Laboratory for Comprehensive Bioimaging, RIKEN Quantitative Biology Center (QBiC), Osaka, 5650874, Japan. .,World Premier International Research Center Initiative, iFReC, Osaka University, Osaka, 5650871, Japan.
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA.
| |
Collapse
|
23
|
Ušaj M, Henn A. Kinetic adaptation of human Myo19 for active mitochondrial transport to growing filopodia tips. Sci Rep 2017; 7:11596. [PMID: 28912602 PMCID: PMC5599584 DOI: 10.1038/s41598-017-11984-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/29/2017] [Indexed: 11/09/2022] Open
Abstract
Myosins are actin-based molecular motors which are enzymatically adapted for their cellular functions such as transportation and membrane tethering. Human Myo19 affects mitochondrial motility, and promotes their localization to stress-induced filopodia. Therefore, studying Myo19 enzymology is essential to understand how this motor may facilitate mitochondrial motility. Towards this goal, we have purified Myo19 motor domain (Myo19-3IQ) from a human-cell expression system and utilized transient kinetics to study the Myo19-3IQ ATPase cycle. We found that Myo19-3IQ exhibits noticeable conformational changes (isomerization steps) preceding both ATP and ADP binding, which may contribute to nucleotide binding regulation. Notably, the ADP isomerization step and subsequent ADP release contribute significantly to the rate-limiting step of the Myo19-3IQ ATPase cycle. Both the slow ADP isomerization and ADP release prolong the time Myo19-3IQ spend in the strong actin binding state and hence contribute to its relatively high duty ratio. However, the predicted duty ratio is lower than required to support motility as a monomer. Therefore, it may be that several Myo19 motors are required to propel mitochondria movement on actin filaments efficiently. Finally, we provide a model explaining how Myo19 translocation may be regulated by the local ATP/ADP ratio, coupled to the mitochondria presence in the filopodia.
Collapse
Affiliation(s)
- Marko Ušaj
- Faculty of Biology, Technion- Israel Institute of Technology, Haifa, 3200003, Israel
| | - Arnon Henn
- Faculty of Biology, Technion- Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
24
|
ROS induced distribution of mitochondria to filopodia by Myo19 depends on a class specific tryptophan in the motor domain. Sci Rep 2017; 7:11577. [PMID: 28912530 PMCID: PMC5599611 DOI: 10.1038/s41598-017-11002-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/17/2017] [Indexed: 01/08/2023] Open
Abstract
The role of the actin cytoskeleton in relation to mitochondria function and dynamics is only recently beginning to be recognized. Myo19 is an actin-based motor that is bound to the outer mitochondrial membrane and promotes the localization of mitochondria to filopodia in response to glucose starvation. However, how glucose starvation induces mitochondria localization to filopodia, what are the dynamics of this process and which enzymatic adaptation allows the translocation of mitochondria to filopodia are not known. Here we show that reactive oxygen species (ROS) mimic and mediate the glucose starvation induced phenotype. In addition, time-lapse fluorescent microscopy reveals that ROS-induced Myo19 motility is a highly dynamic process which is coupled to filopodia elongation and retraction. Interestingly, Myo19 motility is inhibited by back-to-consensus-mutation of a unique residue of class XIX myosins in the motor domain. Kinetic analysis of the purified mutant Myo19 motor domain reveals that the duty ratio (time spent strongly bound to actin) is highly compromised in comparison to that of the WT motor domain, indicating that Myo19 unique motor properties are necessary to propel mitochondria to filopodia tips. In summary, our study demonstrates the contribution of actin-based motility to the mitochondrial localization to filopodia by specific cellular cues.
Collapse
|
25
|
Vavra KC, Xia Y, Rock RS. Competition between Coiled-Coil Structures and the Impact on Myosin-10 Bundle Selection. Biophys J 2017; 110:2517-2527. [PMID: 27276269 DOI: 10.1016/j.bpj.2016.04.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/27/2022] Open
Abstract
Coiled-coil fusions are a useful approach to enforce dimerization in protein engineering. However, the final structures of coiled-coil fusion proteins have received relatively little attention. Here, we determine the structural outcome of adjacent parallel and antiparallel coiled coils. The targets are coiled coils that stabilize myosin-10 in single-molecule biophysical studies. We reveal the solution structure of a short, antiparallel, myosin-10 coiled-coil fused to the parallel GCN4-p1 coiled coil. Surprisingly, this structure is a continuous, antiparallel coiled coil where GCN4-p1 pairs with myosin-10 rather than itself. We also show that longer myosin-10 segments in these parallel/antiparallel fusions are dynamic and do not fold cooperatively. Our data resolve conflicting results on myosin-10 selection of actin filament bundles, demonstrating the importance of understanding coiled-coil orientation and stability.
Collapse
Affiliation(s)
- Kevin C Vavra
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - Youlin Xia
- Minnesota NMR Center, University of Minnesota, Minneapolis, Minnesota
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
26
|
Defective Gpsm2/Gα i3 signalling disrupts stereocilia development and growth cone actin dynamics in Chudley-McCullough syndrome. Nat Commun 2017; 8:14907. [PMID: 28387217 PMCID: PMC5385604 DOI: 10.1038/ncomms14907] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 02/13/2017] [Indexed: 01/14/2023] Open
Abstract
Mutations in GPSM2 cause Chudley-McCullough syndrome (CMCS), an autosomal recessive neurological disorder characterized by early-onset sensorineural deafness and brain anomalies. Here, we show that mutation of the mouse orthologue of GPSM2 affects actin-rich stereocilia elongation in auditory and vestibular hair cells, causing deafness and balance defects. The G-protein subunit Gαi3, a well-documented partner of Gpsm2, participates in the elongation process, and its absence also causes hearing deficits. We show that Gpsm2 defines an ∼200 nm nanodomain at the tips of stereocilia and this localization requires the presence of Gαi3, myosin 15 and whirlin. Using single-molecule tracking, we report that loss of Gpsm2 leads to decreased outgrowth and a disruption of actin dynamics in neuronal growth cones. Our results elucidate the aetiology of CMCS and highlight a new molecular role for Gpsm2/Gαi3 in the regulation of actin dynamics in epithelial and neuronal tissues. Mutations in GPSM2 cause a rare disease characterized by deafness and brain abnormalities. Here the authors show that Gpsm2 forms a molecular complex with a heterotrimeric G-protein subunit, whirlin and a myosin motor to regulate actin dynamics in neurons and auditory hair cell stereocilia.
Collapse
|
27
|
Activated full-length myosin-X moves processively on filopodia with large steps toward diverse two-dimensional directions. Sci Rep 2017; 7:44237. [PMID: 28287133 PMCID: PMC5346999 DOI: 10.1038/srep44237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Myosin-X, (Myo 10), is an unconventional myosin that transports the specific cargos to filopodial tips, and is associated with the mechanism underlying filopodia formation and extension. To clarify the innate motor characteristic, we studied the single molecule movement of a full-length myosin-X construct with leucine zipper at the C-terminal end of the tail (M10FullLZ) and the tail-truncated myosin-X without artificial dimerization motif (BAP-M101–979HMM). M10FullLZ localizes at the tip of filopodia like myosin-X full-length (M10Full). M10FullLZ moves on actin filaments in the presence of PI(3,4,5)P3, an activator of myosin-X. Single molecule motility analysis revealed that the step sizes of both M10FullLZ and BAP-M101–979HMM are widely distributed on single actin filaments that is consistent with electron microscopy observation. M10FullLZ moves on filopodial actin bundles of cells with a mean step size (~36 nm), similar to the step size on single actin filaments (~38 nm). Cartesian plot analysis revealed that M10FullLZ meandered on filopodial actin bundles to both x- and y- directions. These results suggest that the lever-arm of full-length myosin-X is flexible enough to processively steps on different actin filaments within the actin bundles of filopodia. This characteristic of myosin-X may facilitate actin filament convergence for filopodia production.
Collapse
|
28
|
Tripathi KP, Evangelista D, Zuccaro A, Guarracino MR. Transcriptator: An Automated Computational Pipeline to Annotate Assembled Reads and Identify Non Coding RNA. PLoS One 2015; 10:e0140268. [PMID: 26581084 PMCID: PMC4651556 DOI: 10.1371/journal.pone.0140268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022] Open
Abstract
RNA-seq is a new tool to measure RNA transcript counts, using high-throughput sequencing at an extraordinary accuracy. It provides quantitative means to explore the transcriptome of an organism of interest. However, interpreting this extremely large data into biological knowledge is a problem, and biologist-friendly tools are lacking. In our lab, we developed Transcriptator, a web application based on a computational Python pipeline with a user-friendly Java interface. This pipeline uses the web services available for BLAST (Basis Local Search Alignment Tool), QuickGO and DAVID (Database for Annotation, Visualization and Integrated Discovery) tools. It offers a report on statistical analysis of functional and Gene Ontology (GO) annotation's enrichment. It helps users to identify enriched biological themes, particularly GO terms, pathways, domains, gene/proteins features and protein-protein interactions related informations. It clusters the transcripts based on functional annotations and generates a tabular report for functional and gene ontology annotations for each submitted transcript to the web server. The implementation of QuickGo web-services in our pipeline enable the users to carry out GO-Slim analysis, whereas the integration of PORTRAIT (Prediction of transcriptomic non coding RNA (ncRNA) by ab initio methods) helps to identify the non coding RNAs and their regulatory role in transcriptome. In summary, Transcriptator is a useful software for both NGS and array data. It helps the users to characterize the de-novo assembled reads, obtained from NGS experiments for non-referenced organisms, while it also performs the functional enrichment analysis of differentially expressed transcripts/genes for both RNA-seq and micro-array experiments. It generates easy to read tables and interactive charts for better understanding of the data. The pipeline is modular in nature, and provides an opportunity to add new plugins in the future. Web application is freely available at: http://www-labgtp.na.icar.cnr.it/Transcriptator.
Collapse
Affiliation(s)
- Kumar Parijat Tripathi
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council of Italy (CNR), Via Pietro Castellino, 111, Napoli, Italy
- * E-mail:
| | - Daniela Evangelista
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council of Italy (CNR), Via Pietro Castellino, 111, Napoli, Italy
| | - Antonio Zuccaro
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council of Italy (CNR), Via Pietro Castellino, 111, Napoli, Italy
| | - Mario Rosario Guarracino
- Laboratory for Genomics, Transcriptomics and Proteomics (LAB-GTP), High Performance Computing and Networking Institute (ICAR), National Research Council of Italy (CNR), Via Pietro Castellino, 111, Napoli, Italy
| |
Collapse
|
29
|
Li YR, Yang WX. Myosin superfamily: The multi-functional and irreplaceable factors in spermatogenesis and testicular tumors. Gene 2015; 576:195-207. [PMID: 26478466 DOI: 10.1016/j.gene.2015.10.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/21/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Spermatogenesis is a fundamental process in sexual development and reproduction, in which the diploid spermatogonia transform into haploid mature spermatozoa. This process is under the regulation of multiple factors and pathway. Myosin has been implicated in various aspects during spermatogenesis. Myosins constitute a diverse superfamily of actin-based molecular motors that translocate along microfilament in an ATP-dependent manner, and six kinds of myosins have been proved that function during spermatogenesis. In mitosis and meiosis, myosins play an important role in spindle assembly and positioning, karyokinesis and cytokinesis. During spermiogenesis, myosins participate in acrosomal formation, nuclear morphogenesis, mitochondrial translocation and spermatid individualization. In this review, we summarize current understanding of the functions of myosin in spermatogenesis and some reproductive system diseases such as testicular tumors and prostate cancer, and discuss the roles of possible upstream molecules which regulate myosin in these processes.
Collapse
Affiliation(s)
- Yan-Ruide Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
30
|
Yochelis A, Ebrahim S, Millis B, Cui R, Kachar B, Naoz M, Gov NS. Self-organization of waves and pulse trains by molecular motors in cellular protrusions. Sci Rep 2015; 5:13521. [PMID: 26335545 PMCID: PMC4558574 DOI: 10.1038/srep13521] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/29/2015] [Indexed: 11/13/2022] Open
Abstract
Actin-based cellular protrusions are an ubiquitous feature of cells, performing a variety of critical functions ranging from cell-cell communication to cell motility. The formation and maintenance of these protrusions relies on the transport of proteins via myosin motors, to the protrusion tip. While tip-directed motion leads to accumulation of motors (and their molecular cargo) at the protrusion tip, it is observed that motors also form rearward moving, periodic and isolated aggregates. The origins and mechanisms of these aggregates, and whether they are important for the recycling of motors, remain open puzzles. Motivated by novel myosin-XV experiments, a mass conserving reaction-diffusion-advection model is proposed. The model incorporates a non-linear cooperative interaction between motors, which converts them between an active and an inactive state. Specifically, the type of aggregate formed (traveling waves or pulse-trains) is linked to the kinetics of motors at the protrusion tip which is introduced by a boundary condition. These pattern selection mechanisms are found not only to qualitatively agree with empirical observations but open new vistas to the transport phenomena by molecular motors in general.
Collapse
Affiliation(s)
- A Yochelis
- Department of Solar Energy and Environmental Physics, Swiss Institute for Dryland Environmental and Energy Research, Blaustein Institutes for Desert Research (BIDR), Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion 84990, Israel
| | - S Ebrahim
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - B Millis
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - R Cui
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - B Kachar
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - M Naoz
- Department of Chemical Physics, Weizmann Institute of Science, P.O.B. 26, Rehovot, Israel 76100
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, P.O.B. 26, Rehovot, Israel 76100
| |
Collapse
|
31
|
Orly G, Naoz M, Gov NS. Physical model for the geometry of actin-based cellular protrusions. Biophys J 2015; 107:576-587. [PMID: 25099797 DOI: 10.1016/j.bpj.2014.05.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 11/17/2022] Open
Abstract
Actin-based cellular protrusions are a ubiquitous feature of cell morphology, e.g., filopodia and microvilli, serving a huge variety of functions. Despite this, there is still no comprehensive model for the mechanisms that determine the geometry of these protrusions. We present here a detailed computational model that addresses a combination of multiple biochemical and physical processes involved in the dynamic regulation of the shape of these protrusions. We specifically explore the role of actin polymerization in determining both the height and width of the protrusions. Furthermore, we show that our generalized model can explain multiple morphological features of these systems, and account for the effects of specific proteins and mutations.
Collapse
Affiliation(s)
- G Orly
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - M Naoz
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
32
|
Abstract
Morphogenesis is the developmental process by which tissues and organs acquire the shape that is critical to their function. Here, we review recent advances in our understanding of the mechanisms that drive morphogenesis in the developing eye. These investigations have shown that regulation of the actin cytoskeleton is central to shaping the presumptive lens and retinal epithelia that are the major components of the eye. Regulation of the actin cytoskeleton is mediated by Rho family GTPases, by signaling pathways and indirectly, by transcription factors that govern the expression of critical genes. Changes in the actin cytoskeleton can shape cells through the generation of filopodia (that, in the eye, connect adjacent epithelia) or through apical constriction, a process that produces a wedge-shaped cell. We have also learned that one tissue can influence the shape of an adjacent one, probably by direct force transmission, in a process we term inductive morphogenesis. Though these mechanisms of morphogenesis have been identified using the eye as a model system, they are likely to apply broadly where epithelia influence the shape of organs during development.
Collapse
|
33
|
Linder S, Wiesner C. Tools of the trade: podosomes as multipurpose organelles of monocytic cells. Cell Mol Life Sci 2015; 72:121-35. [PMID: 25300510 PMCID: PMC11113205 DOI: 10.1007/s00018-014-1731-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 01/07/2023]
Abstract
Podosomes are adhesion and invasion structures that are particularly prominent in cells of the monocytic lineage such as macrophages, dendritic cells, and osteoclasts. They are multifunctional organelles that combine several key abilities required for cell migration and invasion. The podosome repertoire includes well-established functions such as cell-substrate adhesion, and extracellular matrix degradation, recently discovered abilities such as rigidity and topology sensing as well as antigen sampling, and also more speculative functions such as cell protrusion stabilization and transmigration. Collectively, podosomes not only enable dynamic interactions of cells with their surroundings, they also gather information about the pericellular environment, and are actively involved in its reshaping. This review presents an overview of the current knowledge on podosome composition, architecture, and regulation. We focus in particular on the growing list of podosome functions and discuss the specific properties of podosomes in macrophages, dendritic cells, and osteoclasts. Moreover, this article highlights podosome-related intracellular transport processes, the formation of podosomes in 3D environments as well as potentially podosome-associated diseases involving monocytic cells.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Martinistr. 52, 20246, Hamburg, Germany,
| | | |
Collapse
|
34
|
Ouderkirk JL, Krendel M. Non-muscle myosins in tumor progression, cancer cell invasion, and metastasis. Cytoskeleton (Hoboken) 2014; 71:447-63. [PMID: 25087729 DOI: 10.1002/cm.21187] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 02/06/2023]
Abstract
The actin cytoskeleton, which regulates cell polarity, adhesion, and migration, can influence cancer progression, including initial acquisition of malignant properties by normal cells, invasion of adjacent tissues, and metastasis to distant sites. Actin-dependent molecular motors, myosins, play key roles in regulating tumor progression and metastasis. In this review, we examine how non-muscle myosins regulate neoplastic transformation and cancer cell migration and invasion. Members of the myosin superfamily can act as either enhancers or suppressors of tumor progression. This review summarizes the current state of knowledge on how mutations or epigenetic changes in myosin genes and changes in myosin expression may affect tumor progression and patient outcomes and discusses the proposed mechanisms linking myosin inactivation or upregulation to malignant phenotype, cancer cell migration, and metastasis.
Collapse
Affiliation(s)
- Jessica L Ouderkirk
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 E. Adams St., Syracuse, New York
| | | |
Collapse
|
35
|
Elevated expression of myosin X in tumours contributes to breast cancer aggressiveness and metastasis. Br J Cancer 2014; 111:539-50. [PMID: 24921915 PMCID: PMC4119973 DOI: 10.1038/bjc.2014.298] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/01/2014] [Accepted: 05/09/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Myosin X (MYO10) was recently reported to promote tumour invasion by transporting integrins to filopodial tips in breast cancer. However, the role of MYO10 in tumours remains poorly defined. Here, we report that MYO10 is required in invadopodia to mediate invasive growth and extracellular matrix degradation, which depends on the binding of MYO10's pleckstrin homology domain to PtdIns(3,4,5)P3. METHODS The expression of MYO10 and its associations with clinicopathological and biological factors were examined in breast cancer cells and breast cancer specimens (n=120). Cell migration and invasion were investigated after the silencing of MYO10. The ability of cells to form invadopodia was studied using a fluorescein isothiocyanate-conjugated gelatin degradation assay. A mouse model was established to study tumour invasive growth and metastasis in vivo. RESULTS Elevated MYO10 levels were correlated with oestrogen receptor status, progesterone receptor status, poor differentiation, and lymph node metastasis. Silencing MYO10 reduced cell migration and invasion. Invadopodia were responsible for MYO10's role in promoting invasion. Furthermore, decreased invasive growth and lung metastasis were observed in the MYO10-silenced nude mouse model. CONCLUSIONS Our findings suggest that elevated MYO10 expression increases the aggressiveness of breast cancer; this effect is dependent on the involvement of MYO10 in invadopodial formation.
Collapse
|
36
|
Abstract
Several bacterial pathogens, including Listeria monocytogenes, Shigella flexneri and Rickettsia spp., have evolved mechanisms to actively spread within human tissues. Spreading is initiated by the pathogen-induced recruitment of host filamentous (F)-actin. F-actin forms a tail behind the microbe, propelling it through the cytoplasm. The motile pathogen then encounters the host plasma membrane, forming a bacterium-containing protrusion that is engulfed by an adjacent cell. Over the past two decades, much progress has been made in elucidating mechanisms of F-actin tail formation. Listeria and Shigella produce tails of branched actin filaments by subverting the host Arp2/3 complex. By contrast, Rickettsia forms tails with linear actin filaments through a bacterial mimic of eukaryotic formins. Compared with F-actin tail formation, mechanisms controlling bacterial protrusions are less well understood. However, recent findings have highlighted the importance of pathogen manipulation of host cell–cell junctions in spread. Listeria produces a soluble protein that enhances bacterial protrusions by perturbing tight junctions. Shigella protrusions are engulfed through a clathrin-mediated pathway at ‘tricellular junctions’—specialized membrane regions at the intersection of three epithelial cells. This review summarizes key past findings in pathogen spread, and focuses on recent developments in actin-based motility and the formation and internalization of bacterial protrusions.
Collapse
Affiliation(s)
- Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
37
|
Sebé-Pedrós A, Burkhardt P, Sánchez-Pons N, Fairclough SR, Lang BF, King N, Ruiz-Trillo I. Insights into the origin of metazoan filopodia and microvilli. Mol Biol Evol 2013; 30:2013-23. [PMID: 23770652 PMCID: PMC3748353 DOI: 10.1093/molbev/mst110] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Filopodia are fine actin-based cellular projections used for both environmental sensing and cell motility, and they are essential organelles for metazoan cells. In this study, we reconstruct the origin of metazoan filopodia and microvilli. We first report on the evolutionary assembly of the filopodial molecular toolkit and show that homologs of many metazoan filopodial components, including fascin and myosin X, were already present in the unicellular or colonial progenitors of metazoans. Furthermore, we find that the actin crosslinking protein fascin localizes to filopodia-like structures and microvilli in the choanoflagellate Salpingoeca rosetta. In addition, homologs of filopodial genes in the holozoan Capsaspora owczarzaki are upregulated in filopodia-bearing cells relative to those that lack them. Therefore, our findings suggest that proteins essential for metazoan filopodia and microvilli are functionally conserved in unicellular and colonial holozoans and that the last common ancestor of metazoans bore a complex and specific filopodial machinery.
Collapse
Affiliation(s)
- Arnau Sebé-Pedrós
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Catalonia, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Plantman S, Zelano J, Novikova LN, Novikov LN, Cullheim S. Neuronal myosin-X is upregulated after peripheral nerve injury and mediates laminin-induced growth of neurites. Mol Cell Neurosci 2013; 56:96-101. [PMID: 23603155 DOI: 10.1016/j.mcn.2013.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 03/06/2013] [Accepted: 04/01/2013] [Indexed: 01/07/2023] Open
Abstract
The successful outcome of peripheral neuronal regeneration is attributed both to the growth permissive milieu and the intrinsic ability of the neuron to initiate appropriate cellular responses such as changes in gene expression and cytoskeletal rearrangements. Even though numerous studies have shown the importance of interactions between the neuron and the extracellular matrix (ECM) in axonal outgrowth, the molecular mechanisms underlying the contact between ECM receptors and the cellular cytoskeleton remain largely unknown. Unconventional myosins constitute an important group of cytoskeletal-associated motor proteins. One member of this family is the recently described myosin-X. This protein interacts with several members of the axon growth-associated ECM receptor family of integrins and could therefore be important in neuronal outgrowth. In this study, using radioactive in situ hybridization, we found that expression of myosin-X mRNA is upregulated in adult rat sensory neurons and spinal motoneurons after peripheral nerve injury, but not after central injury. Thus, myosin-X was upregulated after injuries that can be followed by axonal regeneration. We also found that the protein is localized to neuronal growth cones and that silencing of myosin-X using RNA interference impairs the integrin-mediated growth of neurites on laminin, but has no effect on non-integrin mediated growth on N-cadherin.
Collapse
Affiliation(s)
- Stefan Plantman
- Department of Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
39
|
Jung Y, McCarty JH. Band 4.1 proteins regulate integrin-dependent cell spreading. Biochem Biophys Res Commun 2012; 426:578-84. [PMID: 22982319 DOI: 10.1016/j.bbrc.2012.08.129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 08/25/2012] [Indexed: 11/30/2022]
Abstract
Integrins link the extracellular matrix (ECM) to the cytoskeleton to control cell behaviors including adhesion, spreading and migration. Band 4.1 proteins contain 4.1, ezrin, radixin, moesin (FERM) domains that likely mediate signaling events and cytoskeletal reorganization via integrins. However, the mechanisms by which Band 4.1 proteins and integrins are functionally interconnected remain enigmatic. Here we have investigated roles for Band 4.1 proteins in integrin-mediated cell spreading using primary astrocytes as a model system. We demonstrate that Proteins 4.1B and 4.1G show dynamic patterns of sub-cellular localization in astrocytes spreading on fibronectin. During early stages of cell spreading Proteins 4.1B and 4.1G are enriched in ECM adhesion sites but become more diffusely localized at later stages of spreading. Combinatorial inactivation of Protein 4.1B and 4.1G expression leads to impaired astrocyte spreading. Furthermore, in exogenous expression systems we show that the isolated Protein 4.1 FERM domain significantly enhances integrin-mediated cell spreading. Protein 4.1B is dispensable for reactive astrogliosis in experimental models of cortical injury, likely due to functional compensation by related Protein 4.1 family members. Collectively, these findings reveal that Band 4.1 proteins are important intracellular components for integrin-mediated cell spreading.
Collapse
Affiliation(s)
- Youngsin Jung
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston TX 77030, United States
| | | |
Collapse
|
40
|
Lu Q, Lu S, Gao X, Luo Y, Tong B, Wei Z, Lu T, Xia Y, Chou G, Wang Z, Dai Y. Norisoboldine, an alkaloid compound isolated from Radix Linderae, inhibits synovial angiogenesis in adjuvant-induced arthritis rats by moderating Notch1 pathway-related endothelial tip cell phenotype. Exp Biol Med (Maywood) 2012; 237:919-32. [PMID: 22875342 DOI: 10.1258/ebm.2012.011416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Synovial angiogenesis is well recognized as participating in the pathogenesis of rheumatoid arthritis (RA) and has been regarded as a potential target for RA therapy. Previously, we have shown that norisoboldine (NOR) can protect joints from destruction in mice with collagen II-induced arthritis (CIA). Here, we investigate the effect of NOR on synovial angiogenesis in adjuvant-induced arthritis (AA) rats, and clarify the mechanisms in vitro. NOR, administered orally, significantly reduced the number of blood vessels and expression of growth factors in the synovium of AA rats. In vitro, it markedly prevented the migration and sprouting of endothelial cells. Notably, the endothelial tip cell phenotype, which is essential for the migration of endothelial cells and subsequent angiogenesis, was significantly inhibited by NOR. This inhibitory effect was attenuated by pretreatment with N-{N-[2-(3,5-difluorophenyl) acetyl]-(S)-alanyl}-(S)-phenylglycine tert-butyl ester, a Notch1 inhibitor, suggesting that the action of NOR was related to the Notch1 pathway. A molecular docking study further confirmed that NOR was able to promote Notch1 activation by binding the Notch1 transcription complex. In conclusion, NOR was able to prevent synovial angiogenesis in AA rats, which is a putatively new mechanism responsible for its anti-rheumatoid effect. The anti-angiogenesis action of NOR was likely achieved by moderating the Notch1 pathway-related endothelial tip cell phenotype with a potential action target of the Notch1 transcription complex.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pharmacology of Chinese Materia Medica, State Key Laboratory of Natural Medicines
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Perisic L, Lal M, Hulkko J, Hultenby K, Önfelt B, Sun Y, Dunér F, Patrakka J, Betsholtz C, Uhlen M, Brismar H, Tryggvason K, Wernerson A, Pikkarainen T. Plekhh2, a novel podocyte protein downregulated in human focal segmental glomerulosclerosis, is involved in matrix adhesion and actin dynamics. Kidney Int 2012; 82:1071-83. [PMID: 22832517 DOI: 10.1038/ki.2012.252] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pleckstrin homology domain-containing, family H (with MyTH4 domain), member 2 (Plekhh2) is a 1491-residue intracellular protein highly enriched in renal glomerular podocytes for which no function has been ascribed. Analysis of renal biopsies from patients with focal segmental glomerulosclerosis revealed a significant reduction in total podocyte Plekhh2 expression compared to controls. Sequence analysis indicated a putative α-helical coiled-coil segment as the only recognizable domain within the N-terminal half of the polypeptide, while the C-terminal half contains two PH, a MyTH4, and a FERM domain. We identified a phosphatidylinositol-3-phosphate consensus-binding site in the PH1 domain required for Plekhh2 localization to peripheral regions of cell lamellipodia. The N-terminal half of Plekkh2 is not necessary for lamellipodial targeting but mediates self-association. Yeast two-hybrid screening showed that Plekhh2 directly interacts through its FERM domain with the focal adhesion protein Hic-5 and actin. Plekhh2 and Hic-5 coprecipitated and colocalized at the soles of podocyte foot processes in situ and Hic-5 partially relocated from focal adhesions to lamellipodia in Plekhh2-expressing podocytes. In addition, Plekhh2 stabilizes the cortical actin cytoskeleton by attenuating actin depolymerization. Our findings suggest a structural and functional role for Plekhh2 in the podocyte foot processes.
Collapse
Affiliation(s)
- Ljubica Perisic
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhuravlev PI, Lan Y, Minakova MS, Papoian GA. Theory of active transport in filopodia and stereocilia. Proc Natl Acad Sci U S A 2012; 109:10849-54. [PMID: 22711803 PMCID: PMC3390872 DOI: 10.1073/pnas.1200160109] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The biological processes in elongated organelles of living cells are often regulated by molecular motor transport. We determined spatial distributions of motors in such organelles, corresponding to a basic scenario when motors only walk along the substrate, bind, unbind, and diffuse. We developed a mean-field model, which quantitatively reproduces elaborate stochastic simulation results as well as provides a physical interpretation of experimentally observed distributions of Myosin IIIa in stereocilia and filopodia. The mean-field model showed that the jamming of the walking motors is conspicuous, and therefore damps the active motor flux. However, when the motor distributions are coupled to the delivery of actin monomers toward the tip, even the concentration bump of G actin that they create before they jam is enough to speed up the diffusion to allow for severalfold longer filopodia. We found that the concentration profile of G actin along the filopodium is rather nontrivial, containing a narrow minimum near the base followed by a broad maximum. For efficient enough actin transport, this nonmonotonous shape is expected to occur under a broad set of conditions. We also find that the stationary motor distribution is universal for the given set of model parameters regardless of the organelle length, which follows from the form of the kinetic equations and the boundary conditions.
Collapse
Affiliation(s)
- Pavel I. Zhuravlev
- Institute for Physical Science and Technology, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742; and
| | - Yueheng Lan
- Department of Physics, Tsinghua University, Beijing 100084, China
| | - Maria S. Minakova
- Institute for Physical Science and Technology, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742; and
| | - Garegin A. Papoian
- Institute for Physical Science and Technology, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742; and
| |
Collapse
|
43
|
Yu H, Wang N, Ju X, Yang Y, Sun D, Lai M, Cui L, Sheikh MA, Zhang J, Wang X, Zhu X. PtdIns (3,4,5) P3 recruitment of Myo10 is essential for axon development. PLoS One 2012; 7:e36988. [PMID: 22590642 PMCID: PMC3349655 DOI: 10.1371/journal.pone.0036988] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 04/11/2012] [Indexed: 02/03/2023] Open
Abstract
Myosin X (Myo10) with pleckstrin homology (PH) domains is a motor protein acting in filopodium initiation and extension. However, its potential role has not been fully understood, especially in neuronal development. In the present study the preferential accumulation of Myo10 in axon tips has been revealed in primary culture of hippocampal neurons with the aid of immunofluorescence from anti-Myo10 antibody in combination with anti-Tuj1 antibody as specific marker. Knocking down Myo10 gene transcription impaired outgrowth of axon with loss of Tau-1-positive phenotype. Interestingly, inhibition of actin polymerization by cytochalasin D rescued the defect of axon outgrowth. Furthermore, ectopic expression of Myo10 with enhanced green fluorescence protein (EGFP) labeled Myo10 mutants induced multiple axon-like neurites in a motor-independent way. Mechanism studies demonstrated that the recruitment of Myo10 through its PH domain to phosphatidylinositol (3,4,5)-trisphosphate (PtdIns (3,4,5) P3) was essential for axon formation. In addition, in vivo studies confirmed that Myo10 was required for neuronal morphological transition during radial neuronal migration in the developmental neocortex.
Collapse
Affiliation(s)
- Huali Yu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Nannan Wang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xingda Ju
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Yan Yang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Dong Sun
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Mingming Lai
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Lei Cui
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Muhammad Abid Sheikh
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Jianhua Zhang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xingzhi Wang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| |
Collapse
|
44
|
Abstract
Myosin-X (Myo10) is an unconventional myosin with MyTH4-FERM domains that is best known for its striking localization to the tips of filopodia and its ability to induce filopodia. Although the head domain of Myo10 enables it to function as an actin-based motor, its tail contains binding sites for several molecules with central roles in cell biology, including phosphatidylinositol (3,4,5)-trisphosphate, microtubules and integrins. Myo10 also undergoes fascinating long-range movements within filopodia, which appear to represent a newly recognized system of transport. Myo10 is also unusual in that it is a myosin with important roles in the spindle, a microtubule-based structure. Exciting new studies have begun to reveal the structure and single-molecule properties of this intriguing myosin, as well as its mechanisms of regulation and induction of filopodia. At the cellular and organismal level, growing evidence demonstrates that Myo10 has crucial functions in numerous processes ranging from invadopodia formation to cell migration.
Collapse
Affiliation(s)
- Michael L Kerber
- Department of Cell and Molecular Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7545, USA
| | | |
Collapse
|
45
|
Liu KC, Jacobs DT, Dunn BD, Fanning AS, Cheney RE. Myosin-X functions in polarized epithelial cells. Mol Biol Cell 2012; 23:1675-87. [PMID: 22419816 PMCID: PMC3338435 DOI: 10.1091/mbc.e11-04-0358] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myosin-X, an unconventional myosin that has been studied primarily in fibroblast-like cells, has been shown to have important functions in polarized epithelial cell junction formation, regulation of paracellular permeability, and epithelial morphogenesis. Myosin-X (Myo10) is an unconventional myosin that localizes to the tips of filopodia and has critical functions in filopodia. Although Myo10 has been studied primarily in nonpolarized, fibroblast-like cells, Myo10 is expressed in vivo in many epithelia-rich tissues, such as kidney. In this study, we investigate the localization and functions of Myo10 in polarized epithelial cells, using Madin-Darby canine kidney II cells as a model system. Calcium-switch experiments demonstrate that, during junction assembly, green fluorescent protein–Myo10 localizes to lateral membrane cell–cell contacts and to filopodia-like structures imaged by total internal reflection fluorescence on the basal surface. Knockdown of Myo10 leads to delayed recruitment of E-cadherin and ZO-1 to junctions, as well as a delay in tight junction barrier formation, as indicated by a delay in the development of peak transepithelial electrical resistance (TER). Although Myo10 knockdown cells eventually mature into monolayers with normal TER, these monolayers do exhibit increased paracellular permeability to fluorescent dextrans. Importantly, knockdown of Myo10 leads to mitotic spindle misorientation, and in three-dimensional culture, Myo10 knockdown cysts exhibit defects in lumen formation. Together these results reveal that Myo10 functions in polarized epithelial cells in junction formation, regulation of paracellular permeability, and epithelial morphogenesis.
Collapse
Affiliation(s)
- Katy C Liu
- Department of Cell and Molecular Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
46
|
Liu Y, Peng Y, Dai PG, Du QS, Mei L, Xiong WC. Differential regulation of myosin X movements by its cargos, DCC and neogenin. J Cell Sci 2012; 125:751-62. [PMID: 22349703 DOI: 10.1242/jcs.094946] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myosin X (Myo X), also known as MYO10, is an unconventional actin-based motor protein that plays an important role in filopodium formation. Its intra-filopodia movement, an event tightly associated with the function of Myo X, has been extensively studied. However, how the motor activity of Myo X and the direction of its movements are regulated remains largely unknown. In our previous study, we demonstrated that DCC (for 'deleted in colorectal carcinoma') and neogenin (neogenin 1, NEO1 or NGN), a family of immunoglobin-domain-containing transmembrane receptors for netrins, interact with Myo X and that DCC is a cargo of Myo X to be delivered to the neurites of cultured neurons. Here, we provide evidence for DCC and neogenin as regulators of Myo X. DCC promotes movement of Myo X along basal actin filaments and enhances Myo-X-mediated basal filopodium elongation. By contrast, neogenin appears to suppress Myo X movement on the basal side, but increases its movement towards the apical and dorsal side of a cell, promoting dorsal filopodium formation and growth. Further studies have demonstrated that DCC, but not neogenin, enhances integrin-mediated tyrosine phosphorylation of focal adhesion kinase and basal F-actin reorganization, providing a cellular mechanism underlying their distinct effects on Myo X. These results thus demonstrate differential regulatory roles on Myo X activity by its cargo proteins, DCC and neogenin, revealing different cellular functions of DCC and neogenin.
Collapse
Affiliation(s)
- Yu Liu
- Institute of Molecular Medicine & Genetics and Department of Neurology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
47
|
Schäfer C, Faust U, Kirchgessner N, Merkel R, Hoffmann B. The filopodium: a stable structure with highly regulated repetitive cycles of elongation and persistence depending on the actin cross-linker fascin. Cell Adh Migr 2012; 5:431-8. [PMID: 21975552 DOI: 10.4161/cam.5.5.17400] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The ability of mammalian cells to adhere and to migrate is an essential prerequisite to form higher organisms. Early migratory events include substrate sensing, adhesion formation, actin bundle assembly and force generation. Latest research revealed that filopodia are important not only for sensing the substrate but for all of the aforementioned highly regulated processes. However, the exact regulatory mechanisms are still barely understood. Here, we demonstrate that filopodia of human keratinocytes exhibit distinct cycles of repetitive elongation and persistence. A single filopodium thereby is able to initiate the formation of several stable adhesions. Every single filopodial cycle is characterized by an elongation phase, followed by a stabilization time and in many cases a persistence phase. The whole process is strongly connected to the velocity of the lamellipodial leading edge, characterized by a similar phase behavior with a slight time shift compared to filopodia and a different velocity. Most importantly, re-growth of existing filopodia is induced at a sharply defined distance between the filopodial tip and the lamellipodial leading edge. On the molecular level this re-growth is preceded by a strong filopodial reduction of the actin bundling protein fascin. This reduction is achieved by a switch to actin polymerization without fascin incorporation at the filopodial tip and therefore subsequent out-transport of the cross-linker by actin retrograde flow.
Collapse
Affiliation(s)
- Claudia Schäfer
- Institute of Complex Systems, Biomechanics, Forschungszentrum Jülich, Jülich, Germany
| | | | | | | | | |
Collapse
|
48
|
Sun Y, Goldman YE. Lever-arm mechanics of processive myosins. Biophys J 2011; 101:1-11. [PMID: 21723809 DOI: 10.1016/j.bpj.2011.05.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 05/07/2011] [Accepted: 05/09/2011] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yujie Sun
- Pennsylvania Muscle Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
49
|
Lu Q, Yu J, Yan J, Wei Z, Zhang M. Structural basis of the myosin X PH1(N)-PH2-PH1(C) tandem as a specific and acute cellular PI(3,4,5)P(3) sensor. Mol Biol Cell 2011; 22:4268-78. [PMID: 21965296 PMCID: PMC3216653 DOI: 10.1091/mbc.e11-04-0354] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The first PH domain of the myosin X cargo-binding domain is split into halves by insertion of another PH domain forming a PH1N-PH2-PH1C tandem. This tandem forms a rigid supramodule with the two lipid-binding pockets juxtaposed for cooperative binding to PI(3,4,5)P3-containing lipid membranes. Myosin X (MyoX) is an unconventional myosin that is known to induce the formation and elongation of filopodia in many cell types. MyoX-induced filopodial induction requires the three PH domains in its tail region, although with unknown underlying molecular mechanisms. MyoX's first PH domain is split into halves by its second PH domain. We show here that the PH1N-PH2-PH1C tandem allows MyoX to bind to phosphatidylinositol (3,4,5)-triphosphate [PI(3,4,5)P3] with high specificity and cooperativity. We further show that PH2 is responsible for the specificity of the PI(3,4,5)P3 interaction, whereas PH1 functions to enhance the lipid membrane–binding avidity of the tandem. The structure of the MyoX PH1N-PH2-PH1C tandem reveals that the split PH1, PH2, and the highly conserved interdomain linker sequences together form a rigid supramodule with two lipid-binding pockets positioned side by side for binding to phosphoinositide membrane bilayers with cooperativity. Finally, we demonstrate that disruption of PH2-mediated binding to PI(3,4,5)P3 abolishes MyoX's function in inducing filopodial formation and elongation.
Collapse
Affiliation(s)
- Qing Lu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
50
|
Abstract
Cell migration requires sustained forward movement of the plasma membrane at the cell's front or "leading edge." To date, researchers have uncovered four distinct ways of extending the membrane at the leading edge. In lamellipodia and filopodia, actin polymerization directly pushes the plasma membrane forward, whereas in invadopodia, actin polymerization couples with the extracellular delivery of matrix-degrading metalloproteases to clear a path for cells through the extracellular matrix. Membrane blebs drive the plasma membrane forward using a combination of actomyosin-based contractility and reversible detachment of the membrane from the cortical actin cytoskeleton. Each protrusion type requires the coordination of a wide spectrum of signaling molecules and regulators of cytoskeletal dynamics. In addition, these different protrusion methods likely act in concert to move cells through complex environments in vivo.
Collapse
|