51
|
Hegsted A, Yingling CV, Pruyne D. Inverted formins: A subfamily of atypical formins. Cytoskeleton (Hoboken) 2017; 74:405-419. [PMID: 28921928 DOI: 10.1002/cm.21409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/22/2017] [Accepted: 08/31/2017] [Indexed: 12/25/2022]
Abstract
Formins are a family of regulators of actin and microtubule dynamics that are present in almost all eukaryotes. These proteins are involved in many cellular processes, including cytokinesis, stress fiber formation, and cell polarization. Here we review one subfamily of formins, the inverted formins. Inverted formins as a group break several formin stereotypes, having atypical biochemical properties and domain organization, and they have been linked to kidney disease and neuropathy in humans. In this review, we will explore recent research on members of the inverted formin sub-family in mammals, zebrafish, fruit flies, and worms.
Collapse
Affiliation(s)
- Anna Hegsted
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Curtis V Yingling
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - David Pruyne
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
52
|
Dahlhoff M, Gaborit N, Bultmann S, Leonhardt H, Yarden Y, Schneider MR. CRISPR-assisted receptor deletion reveals distinct roles for ERBB2 and ERBB3 in skin keratinocytes. FEBS J 2017; 284:3339-3349. [DOI: 10.1111/febs.14196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/25/2017] [Accepted: 08/10/2017] [Indexed: 01/06/2023]
Affiliation(s)
- Maik Dahlhoff
- Institute of Molecular Animal Breeding and Biotechnology; LMU Munich; Germany
| | - Nadège Gaborit
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| | - Sebastian Bultmann
- Human Biology and BioImaging; Department of Biology II; LMU Munich; Germany
| | - Heinrich Leonhardt
- Human Biology and BioImaging; Department of Biology II; LMU Munich; Germany
| | - Yosef Yarden
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| | - Marlon R. Schneider
- German Centre for the Protection of Laboratory Animals (Bf3R); German Federal Institute for Risk Assessment (BfR); Berlin Germany
| |
Collapse
|
53
|
Jaureguiberry-Bravo M, Wilson R, Carvallo L, Berman JW. Opioids and Opioid Maintenance Therapies: Their Impact on Monocyte-Mediated HIV Neuropathogenesis. Curr HIV Res 2017; 14:417-430. [PMID: 27009099 DOI: 10.2174/1570162x14666160324124132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/26/2015] [Accepted: 11/10/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND HIV-1 enters the CNS within two weeks after peripheral infection and results in chronic neuroinflammation that leads to HIV associated neurocognitive disorders (HAND) in more than 50% of infected people. HIV enters the CNS by transmigration of infected monocytes across the blood brain barrier. Intravenous drug abuse is a major risk factor for HIV-1 infection, and opioids have been shown to alter the progression and severity of HAND. Methadone and buprenorphine are opioid derivates that are used as opioid maintenance therapies. They are commonly used to treat opioid dependency in HIV infected substance abusers, but their effects on monocyte migration relevant to the development of cognitive impairment are not well characterized. CONCLUSION Here, we will discuss the effects of opioids and opioid maintenance therapies on the inflammatory functions of monocytes and macrophages that are related to the development of neuroinflammation in the context of HIV infection.
Collapse
Affiliation(s)
| | | | | | - Joan W Berman
- Department, of Pathology and Microbiology and Immunology, F727, Albert Einstein College of Medicine, 1300 Morris Park Ave. Bronx, NY, 10461, USA.
| |
Collapse
|
54
|
Ritto D, Tanasawet S, Singkhorn S, Klaypradit W, Hutamekalin P, Tipmanee V, Sukketsiri W. Astaxanthin induces migration in human skin keratinocytes via Rac1 activation and RhoA inhibition. Nutr Res Pract 2017; 11:275-280. [PMID: 28765773 PMCID: PMC5537536 DOI: 10.4162/nrp.2017.11.4.275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/20/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/OBJECTIVES Re-epithelialization has an important role in skin wound healing. Astaxanthin (ASX), a carotenoid found in crustaceans including shrimp, crab, and salmon, has been widely used for skin protection. Therefore, we investigated the effects of ASX on proliferation and migration of human skin keratinocyte cells and explored the mechanism associated with that migration. MATERIAL/METHOD HaCaT keratinocyte cells were exposed to 0.25-1 µg/mL of ASX. Proliferation of keratinocytes was analyzed by using MTT assays and flow cytometry. Keratinocyte migration was determined by using a scratch wound-healing assay. A mechanism for regulation of migration was explored via immunocytochemistry and western blot analysis. RESULTS Our results suggest that ASX produces no significant toxicity in human keratinocyte cells. Cell-cycle analysis on ASX-treated keratinocytes demonstrated a significant increase in keratinocyte cell proliferation at the S phase. In addition, ASX increased keratinocyte motility across the wound space in a time-dependent manner. The mechanism by which ASX increased keratinocyte migration was associated with induction of filopodia and formation of lamellipodia, as well as with increased Cdc42 and Rac1 activation and decreased RhoA activation. CONCLUSIONS ASX stimulates the migration of keratinocytes through Cdc42, Rac1 activation and RhoA inhibition. ASX has a positive role in the re-epithelialization of wounds. Our results may encourage further in vivo and clinical study into the development of ASX as a potential agent for wound repair.
Collapse
Affiliation(s)
- Dakanda Ritto
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, 15 Hat Yai, Songkhla 90110, Thailand
| | - Supita Tanasawet
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Sawana Singkhorn
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, 15 Hat Yai, Songkhla 90110, Thailand
| | - Wanwimol Klaypradit
- Department of Fishery Product, Faculty of Fishery, Kasetsart University, Bangkok 10900, Thailand
| | - Pilaiwanwadee Hutamekalin
- Department of Physiology, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Wanida Sukketsiri
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, 15 Hat Yai, Songkhla 90110, Thailand
| |
Collapse
|
55
|
The interplay between histone deacetylases and rho kinases is important for cancer and neurodegeneration. Cytokine Growth Factor Rev 2017; 37:29-45. [PMID: 28606734 DOI: 10.1016/j.cytogfr.2017.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/18/2017] [Accepted: 05/21/2017] [Indexed: 12/24/2022]
Abstract
Rho associated coiled-coil containing kinases (ROCKs) respond to defined extra- and intracellular stimuli to control cell migration, cell proliferation, and apoptosis. Histone deacetylases (HDACs) are epigenetic modifiers that regulate nuclear and cytoplasmic signaling through the deacetylation of histones and non-histone proteins. ROCK and HDAC functions are important compounds of basic and applied research interests. Recent evidence suggests a physiologically important interplay between HDACs and ROCKs in various cells and organisms. Here we summarize the crosstalk between these enzymatic families and its implications for cancer and neurodegeneration.
Collapse
|
56
|
Dong DD, Zhou H, Li G. GPR78 promotes lung cancer cell migration and metastasis by activation of Gαq-Rho GTPase pathway. BMB Rep 2017; 49:623-628. [PMID: 27697106 PMCID: PMC5346323 DOI: 10.5483/bmbrep.2016.49.11.133] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Indexed: 12/31/2022] Open
Abstract
GPR78 is an orphan G-protein coupled receptor (GPCR) that is predominantly expressed in human brain tissues. Currently, the function of GPR78 is unknown. This study revealed that GPR78 was expressed in lung cancer cells and functioned as a novel regulator of lung cancer cell migration and metastasis. We found that knockdown of GPR78 in lung cancer cells suppressed cell migration. Moreover, GPR78 modulated the formation of actin stress fibers in A549 cells, in a RhoA- and Rac1-dependent manner. At the molecular level, GPR78 regulated cell motility through the activation of Gαq-RhoA/Rac1 pathway. We further demonstrated that in vivo, the knockdown of GPR78 inhibited lung cancer cell metastasis. These findings suggest that GPR78 is a novel regulator for lung cancer metastasis and may serve as a potential drug target against metastatic human lung cancer. [BMB Reports 2016; 49(11): 623-628].
Collapse
Affiliation(s)
- Dan-Dan Dong
- Department of Pathology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan, China
| | - Hui Zhou
- Department of Thoracic Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Gao Li
- Thoracic Surgery , Hainan General Hospital, Haikou 570311, Hainan, PR China
| |
Collapse
|
57
|
Li C, Imanishi A, Komatsu N, Terai K, Amano M, Kaibuchi K, Matsuda M. A FRET Biosensor for ROCK Based on a Consensus Substrate Sequence Identified by KISS Technology. Cell Struct Funct 2017; 42:1-13. [DOI: 10.1247/csf.16016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Chunjie Li
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University
| | - Ayako Imanishi
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University
| | - Naoki Komatsu
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University
| | - Kenta Terai
- Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University
| |
Collapse
|
58
|
Wang W, Tao K, Wang J, Yang G, Ouyang Q, Wang Y, Zhang L, Liu F. Exploring the inhibitory effect of membrane tension on cell polarization. PLoS Comput Biol 2017; 13:e1005354. [PMID: 28135277 PMCID: PMC5305267 DOI: 10.1371/journal.pcbi.1005354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 02/13/2017] [Accepted: 01/11/2017] [Indexed: 01/02/2023] Open
Abstract
Cell polarization toward an attractant is influenced by both physical and chemical factors. Most existing mathematical models are based on reaction-diffusion systems and only focus on the chemical process occurring during cell polarization. However, membrane tension has been shown to act as a long-range inhibitor of cell polarization. Here, we present a cell polarization model incorporating the interplay between Rac GTPase, filamentous actin (F-actin), and cell membrane tension. We further test the predictions of this model by performing single cell measurements of the spontaneous polarization of cancer stem cells (CSCs) and non-stem cancer cells (NSCCs), as the former have lower cell membrane tension. Based on both our model and the experimental results, cell polarization is more sensitive to stimuli under low membrane tension, and high membrane tension improves the robustness and stability of cell polarization such that polarization persists under random perturbations. Furthermore, our simulations are the first to recapitulate the experimental results described by Houk et al., revealing that aspiration (elevation of tension) and release (reduction of tension) result in a decrease in and recovery of the activity of Rac-GTP, respectively, and that the relaxation of tension induces new polarity of the cell body when a cell with the pseudopod-neck-body morphology is severed.
Collapse
Affiliation(s)
- Weikang Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, People’s Republic of China
| | - Kuan Tao
- Center for Quantitative Biology, Peking University, Beijing, People’s Republic of China
| | - Jing Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, People’s Republic of China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, People’s Republic of China
| | - Qi Ouyang
- Center for Quantitative Biology, Peking University, Beijing, People’s Republic of China
| | - Yugang Wang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, People’s Republic of China
| | - Lei Zhang
- Center for Quantitative Biology, Peking University, Beijing, People’s Republic of China
- Beijing International Center for Mathematical Research, Peking University, Beijing, People’s Republic of China
| | - Feng Liu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing, People’s Republic of China
- Center for Quantitative Biology, Peking University, Beijing, People’s Republic of China
| |
Collapse
|
59
|
Kamiya K, Takeuchi S. Giant liposome formation toward the synthesis of well-defined artificial cells. J Mater Chem B 2017; 5:5911-5923. [DOI: 10.1039/c7tb01322a] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review focuses on microfluidic technologies for giant liposome formations which emulate environments of biological cells.
Collapse
Affiliation(s)
- Koki Kamiya
- Artificial Cell Membrane Systems Group
- Kanagawa Institute of Industrial Science and Technology
- Kawasaki
- Japan
| | - Shoji Takeuchi
- Artificial Cell Membrane Systems Group
- Kanagawa Institute of Industrial Science and Technology
- Kawasaki
- Japan
- Institute of Industrial Science
| |
Collapse
|
60
|
Feng G, Zhu Z, Li WJ, Lin Q, Chai Y, Dong MQ, Ou G. Hippo kinases maintain polarity during directional cell migration in Caenorhabditis elegans. EMBO J 2016; 36:334-345. [PMID: 28011581 DOI: 10.15252/embj.201695734] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/07/2016] [Accepted: 11/16/2016] [Indexed: 01/21/2023] Open
Abstract
Precise positioning of cells is crucial for metazoan development. Despite immense progress in the elucidation of the attractive cues of cell migration, the repulsive mechanisms that prevent the formation of secondary leading edges remain less investigated. Here, we demonstrate that Caenorhabditis elegans Hippo kinases promote cell migration along the anterior-posterior body axis via the inhibition of dorsal-ventral (DV) migration. Ectopic DV polarization was also demonstrated in gain-of-function mutant animals for C. elegans RhoG MIG-2. We identified serine 139 of MIG-2 as a novel conserved Hippo kinase phosphorylation site and demonstrated that purified Hippo kinases directly phosphorylate MIG-2S139 Live imaging analysis of genome-edited animals indicates that MIG-2S139 phosphorylation impedes actin assembly in migrating cells. Intriguingly, Hippo kinases are excluded from the leading edge in wild-type cells, while MIG-2 loss induces uniform distribution of Hippo kinases. We provide evidence that Hippo kinases inhibit RhoG activity locally and are in turn restricted to the cell body by RhoG-mediated polarization. Therefore, we propose that the Hippo-RhoG feedback regulation maintains cell polarity during directional cell motility.
Collapse
Affiliation(s)
- Guoxin Feng
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Zhiwen Zhu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Wen-Jun Li
- National Institute of Biological Science, Beijing, China
| | - Qirong Lin
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Yongping Chai
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Meng-Qiu Dong
- National Institute of Biological Science, Beijing, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| |
Collapse
|
61
|
Raza Q, Jacobs JR. Guidance signalling regulates leading edge behaviour during collective cell migration of cardiac cells in Drosophila. Dev Biol 2016; 419:285-297. [PMID: 27618756 DOI: 10.1016/j.ydbio.2016.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/04/2016] [Accepted: 09/05/2016] [Indexed: 01/31/2023]
Abstract
Collective cell migration is the coordinated movement of cells, which organize tissues during morphogenesis, repair and some cancers. The motile cell membrane of the advancing front in collective cell migration is termed the Leading Edge. The embryonic development of the vertebrate and Drosophila hearts are both characterized by the coordinated medial migration of a bilateral cluster of mesodermal cells. In Drosophila, the cardioblasts form cohesive bilateral rows that migrate collectively as a unit towards the dorsal midline to form the dorsal vessel. We have characterized the collective cell migration of cardioblasts as an in vivo quantitative model to study the behaviour of the Leading Edge. We investigated whether guidance signalling through Slit and Netrin pathways plays a role in cell migration during heart development. Through time-lapse imaging and quantitative assessment of migratory behaviour of the cardioblasts in loss-of-function mutants, we demonstrate that both Slit and Netrin mediated signals are autonomously and concomitantly required to maximize migration velocity, filopodial and lamellipodial activities. Additionally, we show that another Slit and Netrin receptor, Dscam1, the role of which during heart development was previously unknown, is required for both normal migration of cardioblasts and luminal expansion. Leading edge behaviour analysis revealed a dosage dependent genetic interaction between Slit and Netrin receptors suggesting that downstream signalling through these receptors converge on a common output that increases leading edge activity of the cardioblasts. Finally, we found that guidance signalling maintains the balance between epithelial and mesenchymal characteristics of the migrating cardioblasts.
Collapse
Affiliation(s)
- Qanber Raza
- Department of Biology, McMaster University, 1280 Main St W, Hamilton, ON, Canada L8S 4L8
| | - J Roger Jacobs
- Department of Biology, McMaster University, 1280 Main St W, Hamilton, ON, Canada L8S 4L8.
| |
Collapse
|
62
|
Goncharenko AV, Malyuchenko NV, Moisenovich AM, Kotlyarova MS, Arkhipova AY, Kon’kov AS, Agapov II, Molochkov AV, Moisenovich MM, Kirpichnikov MP. Changes in morphology of actin filaments and expression of alkaline phosphatase at 3D cultivation of MG-63 osteoblast-like cells on mineralized fibroin scaffolds. DOKL BIOCHEM BIOPHYS 2016; 470:368-370. [DOI: 10.1134/s1607672916050197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Indexed: 11/23/2022]
|
63
|
Makihara M, Watanabe T, Usukura E, Kaibuchi K, Narita A, Tanaka N, Usukura J. A new approach for the direct visualization of the membrane cytoskeleton in cryo-electron microscopy: a comparative study with freeze-etching electron microscopy. Microscopy (Oxf) 2016; 65:488-498. [PMID: 27587510 DOI: 10.1093/jmicro/dfw037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 08/10/2016] [Indexed: 12/17/2022] Open
Abstract
An improved unroofing method consisting of tearing off the cell membrane using an adhesive electron microscopy (EM) grid instead of vitreous ice sectioning (cryo-sectioning) has enabled us to panoramically view the membrane cytoskeleton in its native state with extremely high contrast. Grids pre-treated with Alcian blue were placed on cells, and a portion of the dorsal plasma membrane was transferred onto the grid, which was then floated in buffer solution. These membrane fragments contained sufficient cytoskeleton and were of suitable thickness for observation by cryo-EM. Many actin filaments and microtubules were clearly observed on the cytoplasmic surface of the plasma membrane with extremely high contrast because the soluble components of the cytoplasm flowed out and broke away from the cells. Actin filaments extended in all directions in a smooth contour with little branching. Microtubules spread out as far as 3 µm or more while winding gently in their native state. Upon fixation with 1% glutaraldehyde, however, the microtubules became straight and fragmented. Cryo-EM revealed for the first time a smooth endoplasmic reticulum network beneath the cell membrane in native cells. Clathrin coats and caveolae were also observed on the cytoplasmic surface of the plasma membrane, similar to those seen using freeze-etching replica EM (freeze-etching EM). Unroofing was also useful for immuno-labelling in cryo-EM. Antibody-labelled IQGAP1, one of the effector proteins facilitating the formation of actin filament networks, was localized alongside actin filaments. Freeze-etching EM confirmed the morphological findings of cryo-EM.
Collapse
Affiliation(s)
| | - Takashi Watanabe
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550
| | - Eiji Usukura
- Structural Biology Research Center, Nagoya University, Nagoya 464-8603
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550
| | - Akihiro Narita
- Structural Biology Research Center, Nagoya University, Nagoya 464-8603
| | - Nobuo Tanaka
- Institute of Materials and Systems for Sustainability, Nagoya University, Nagoya 464-8603, Japan
| | - Jiro Usukura
- Structural Biology Research Center, Nagoya University, Nagoya 464-8603
| |
Collapse
|
64
|
Zoheir KM, Abd-Rabou AA, Harisa GI, Kumar A, Ahmad SF, Ansari MA, Abd-Allah AR. IQGAP1 gene silencing induces apoptosis and decreases the invasive capacity of human hepatocellular carcinoma cells. Tumour Biol 2016; 37:13927-13939. [PMID: 27488117 DOI: 10.1007/s13277-016-5283-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/15/2016] [Indexed: 12/24/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) belong to a conserved family, and they are involved in various intracellular processes. IQGAP1 is expressed in all cells, while IQGAP2 and IQGAP3 are mainly expressed in hepatic cells. IQGAP1 has been suggested to be an oncogene, while IQGAP2 is considered a tumor-suppressor gene. However, the relationship between RAS family genes and IQGAP genes remains unclear. We recently demonstrated this interaction in a chemically induced mouse liver cancer. In this study, IQGAP1 expression was partially silenced in human hepatocellular carcinoma (HepG2) cells. We investigated the impact of IQGAP1 silencing on the interactions of IQGAP and RAS with several apoptotic proteins, including caspase-3 (CASP3), BCL2-associated X protein (BAX), and B-cell leukemia/lymphoma 2 (BCL2). Additionally, we investigated the effects of the interactions of these genes on cell viability, proliferation, apoptosis, and invasive capacity. IQGAP1 siRNA-treated HepG2 cells showed lower invasive capacity than the control cells, and this reduction was time- and vector concentration-dependent. In addition, IQGAP1 silencing resulted in significantly lower IQGAP1 level and subsequently higher IQGAP2 and IQGAP3 expression in HepG2 cells than in the control. Flow cytometry analyses indicated that the silencing of IQGAP1 can induce early and late apoptosis in HepG2 cells. Additionally, IQGAP2, IQGAP3, CASP3, and BAX were upregulated whereas IQGAP1 and BCL2 were downregulated in the siRNA-treated cells. Furthermore, we observed that the mRNA levels of HRAS, KRAS, NRAS, and MRAS decreased upon IQGAP1 silencing. These findings indicate that IQGAP1 potentially regulates the expression of IQGAP and RAS gene families and demonstrate its regulatory role in the apoptotic network. Taken together, our findings suggest that IQGAP1 silencing plays crucial roles in the apoptosis of HepG2 cells and lowers their proliferative and invasive capacities.
Collapse
Affiliation(s)
- Khairy Ma Zoheir
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia. .,Cell Biology Department, National Research Centre, Cairo, 12622, Egypt.
| | - Ahmed A Abd-Rabou
- Hormones Department, Medical Research Division, National Research Centre, Cairo, 12622, Egypt
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, PO Box 11451, Riyadh, Saudi Arabia
| | - Ashok Kumar
- Vitiligo Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh Fayaz Ahmad
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq Ahmad Ansari
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Adel R Abd-Allah
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
65
|
Zhang G, Gu Y, Begum R, Chen H, Gao X, McGrath JA, Parsons M, Song B. Kindlin-1 Regulates Keratinocyte Electrotaxis. J Invest Dermatol 2016; 136:2229-2239. [PMID: 27427485 PMCID: PMC5756539 DOI: 10.1016/j.jid.2016.05.129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/18/2016] [Accepted: 05/10/2016] [Indexed: 12/05/2022]
Abstract
Kindler syndrome (KS) is an autosomal recessive blistering skin disease resulting from pathogenic mutations in FERMT1. This gene encodes kindlin-1, a focal adhesion protein involved in activation of the integrin family of extracellular matrix receptors. Most cases of KS show a marked reduction or complete absence of the kindlin-1 protein in keratinocytes, resulting in defective cell adhesion and migration. Electric fields also act as intrinsic regulators of adhesion and migration in the skin, but the molecular mechanisms by which this occurs are poorly understood. Here we show that keratinocytes derived from KS patients are unable to undergo electrotaxis, and this defect is restored by overexpression of wild-type kindlin-1 but not a W612A mutation that prevents kindlin-integrin binding. Moreover, deletion of the pleckstrin homology domain of kindlin-1 also failed to rescue electrotaxis in KS cells, indicating that both integrin and lipid binding are required for this function. Kindlin-1 was also required for the maintenance of lamellipodial protrusions during electrotaxis via electric field-activated β1 integrin. Indeed, inhibition of β1 integrins also leads to loss of electrotaxis in keratinocytes. Our data suggest that loss of kindlin-1 function may therefore result in epithelial insensitivity to electric fields and contribute to KS disease pathology.
Collapse
Affiliation(s)
- Gaofeng Zhang
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang, China; School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Yu Gu
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Rumena Begum
- Randall Division of Cell and Molecular Biophysics, Kings College London, London, UK
| | - Hongduo Chen
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang, China
| | - Xinghua Gao
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang, China
| | - John A McGrath
- St. Johns Institute of Dermatology, King's College London, Guys Campus, London, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, Kings College London, London, UK.
| | - Bing Song
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang, China; School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
66
|
Kundu S. Stochastic modelling suggests that an elevated superoxide anion - hydrogen peroxide ratio can drive extravascular phagocyte transmigration by lamellipodium formation. J Theor Biol 2016; 407:143-154. [PMID: 27380944 DOI: 10.1016/j.jtbi.2016.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/01/2016] [Indexed: 11/24/2022]
Abstract
Chemotaxis, integrates diverse intra- and inter-cellular molecular processes into a purposeful patho-physiological response; the operatic rules of which, remain speculative. Here, I surmise, that superoxide anion induced directional motility, in a responding cell, results from a quasi pathway between the stimulus, surrounding interstitium, and its biochemical repertoire. The epochal event in the mounting of an inflammatory response, is the extravascular transmigration of a phagocyte competent cell towards the site of injury, secondary to the development of a lamellipodium. This stochastic-to-markovian process conversion, is initiated by the cytosolic-ROS of the damaged cell, but is maintained by the inverse association of a de novo generated pool of self-sustaining superoxide anions and sub-critical hydrogen peroxide levels. Whilst, the exponential rise of O2(.-) is secondary to the focal accumulation of higher order lipid raft-Rac1/2-actin oligomers; O2(.-) mediated inactivation and redistribution of ECSOD, accounts for the minimal concentration of H2O2 that the phagocyte experiences. The net result of this reciprocal association between ROS/ RNS members, is the prolonged perturbation and remodeling of the cytoskeleton and plasma membrane, a prelude to chemotactic migration. The manuscript also describes the significance of stochastic modeling, in the testing of plausible molecular hypotheses of observable phenomena in complex biological systems.
Collapse
Affiliation(s)
- Siddhartha Kundu
- Department of Biochemistry, Dr. Baba Saheb Ambedkar Medical College & Hospital, Government of NCT Delhi, Sector - 6, Rohini, Delhi 110085, India; Mathematical and Computational Biology, Information Technology Research Academy (ITRA), Media Lab Asia, 2nd Floor, Block 2, C-DOT Campus, Mehrauli, New Delhi 110030, India; School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| |
Collapse
|
67
|
Duan B, Cui J, Sun S, Zheng J, Zhang Y, Ye B, Chen Y, Deng W, Du J, Zhu Y, Chen Y, Gu L. EGF-stimulated activation of Rab35 regulates RUSC2-GIT2 complex formation to stabilize GIT2 during directional lung cancer cell migration. Cancer Lett 2016; 379:70-83. [PMID: 27238570 DOI: 10.1016/j.canlet.2016.05.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/20/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
Abstract
Non-small cell lung cancer (NSCLC) remains one of the most metastasizing tumors, and directional cell migration is critical for targeting tumor metastasis. GIT2 has been known to bind to Paxillin to control cell polarization and directional migration. However, the molecular mechanisms underlying roles of GIT2 in controlling cell polarization and directional migration remain elusive. Here we demonstrated GIT2 control cell polarization and direction dependent on the regulation of Golgi through RUSC2. RUSC2 interacts with SHD of GIT2 in various lung cancer cells, and stabilizes GIT2 (Mazaki et al., 2006; Yu et al., 2009) by decreasing degradation and increasing its phosphorylation. Silencing of RUSC2 showed reduced stability of GIT2, defective Golgi reorientation toward the wound edge and decreased directional migration. Moreover, short-term EGF stimulation can increase the interaction between RUSC2 and GIT2, prolonged stimulation leads to a decrease of their interaction through activating Rab35. Silencing of Rab35 also reduced stability and phosphorylation of GIT2 and decreased cell migration. Taken together, our study indicated that RUSC2 participates in EGFR signaling and regulates lung cancer progression, and may be a new therapeutic target against lung cancer metastasis.
Collapse
Affiliation(s)
- Biao Duan
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jie Cui
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Shixiu Sun
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jianchao Zheng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yujie Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bixing Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yan Chen
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wenjie Deng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
68
|
Yamaoka M, Ishizaki T, Kimura T. GTP- and GDP-Dependent Rab27a Effectors in Pancreatic Beta-Cells. Biol Pharm Bull 2016; 38:663-8. [PMID: 25947911 DOI: 10.1248/bpb.b14-00886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Small guanosine triphosphatases (GTPases) participate in a wide variety of cellular functions including proliferation, differentiation, adhesion, and intracellular transport. Conventionally, only the guanosine 5'-triphosphate (GTP)-bound small GTPase interacts with effector proteins, and the resulting downstream signals control specific cellular functions. Therefore, the GTP-bound form is regarded as active, and the focus has been on searching for proteins that bind the GTP form to look for their effectors. The Rab family small GTPase Rab27a is highly expressed in some secretory cells and is involved in the control of membrane traffic. The present study reviews recent progress in our understanding of the roles of Rab27a and its effectors in pancreatic beta-cells. In the basal state, GTP-bound Rab27a controls insulin secretion at pre-exocytic stages via its GTP-dependent effectors. We previously identified novel guanosine 5'-diphosphate (GDP)-bound Rab27-interacting proteins. Interestingly, GDP-bound Rab27a controls endocytosis of the secretory membrane via its interaction with these proteins. We also demonstrated that the insulin secretagogue glucose converts Rab27a from its GTP- to GDP-bound forms. Thus, GTP- and GDP-bound Rab27a regulate pre-exocytic and endocytic stages in membrane traffic, respectively. Since the physiological importance of GDP-bound GTPases has been largely overlooked, we consider that the investigation of GDP-dependent effectors for other GTPases is necessary for further understanding of cellular function.
Collapse
Affiliation(s)
- Mami Yamaoka
- Department of Pharmacology, Oita University Faculty of Medicine
| | | | | |
Collapse
|
69
|
Gandalovičová A, Vomastek T, Rosel D, Brábek J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget 2016; 7:25022-49. [PMID: 26872368 PMCID: PMC5041887 DOI: 10.18632/oncotarget.7214] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Apico-basal polarity is typical of cells present in differentiated epithelium while front-rear polarity develops in motile cells. In cancer development, the transition from epithelial to migratory polarity may be seen as the hallmark of cancer progression to an invasive and metastatic disease. Despite the morphological and functional dissimilarity, both epithelial and migratory polarity are controlled by a common set of polarity complexes Par, Scribble and Crumbs, phosphoinositides, and small Rho GTPases Rac, Rho and Cdc42. In epithelial tissues, their mutual interplay ensures apico-basal and planar cell polarity. Accordingly, altered functions of these polarity determinants lead to disrupted cell-cell adhesions, cytoskeleton rearrangements and overall loss of epithelial homeostasis. Polarity proteins are further engaged in diverse interactions that promote the establishment of front-rear polarity, and they help cancer cells to adopt different invasion modes. Invading cancer cells can employ either the collective, mesenchymal or amoeboid invasion modes or actively switch between them and gain intermediate phenotypes. Elucidation of the role of polarity proteins during these invasion modes and the associated transitions is a necessary step towards understanding the complex problem of metastasis. In this review we summarize the current knowledge of the role of cell polarity signaling in the plasticity of cancer cell invasiveness.
Collapse
Affiliation(s)
- Aneta Gandalovičová
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Tomáš Vomastek
- Institute of Microbiology, Academy of Sciences of The Czech Republic, Videňská, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| |
Collapse
|
70
|
Bartolini F, Andres-Delgado L, Qu X, Nik S, Ramalingam N, Kremer L, Alonso MA, Gundersen GG. An mDia1-INF2 formin activation cascade facilitated by IQGAP1 regulates stable microtubules in migrating cells. Mol Biol Cell 2016; 27:1797-808. [PMID: 27030671 PMCID: PMC4884070 DOI: 10.1091/mbc.e15-07-0489] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 03/25/2016] [Indexed: 01/08/2023] Open
Abstract
The formin INF2 is required for stable Glu microtubule formation and inhibition of microtubule dynamics in NIH3T3 cells downstream of mDia1 and LPA. Evidence also shows that the formation of an mDia1/INF2 complex is necessary for microtubule stabilization stimulated by LPA and is regulated by IQGAP1. Multiple formins regulate microtubule (MT) arrays, but whether they function individually or in a common pathway is unknown. Lysophosphatidic acid (LPA) stimulates the formation of stabilized detyrosinated MTs (Glu MTs) in NIH3T3 fibroblasts through RhoA and the formin mDia1. Here we show that another formin, INF2, is necessary for mDia1-mediated induction of Glu MTs and regulation of MT dynamics and that mDia1 can be bypassed by activating INF2. INF2 localized to MTs after LPA treatment in an mDia1-dependent manner, suggesting that mDia1 regulates INF2. Mutants of either formin that disrupt their interaction failed to rescue MT stability in cells depleted of the respective formin, and the mDia1-interacting protein IQGAP1 regulated INF2’s localization to MTs and the induction of Glu MTs by either formin. The N-terminus of IQGAP1 associated with the C-terminus of INF2 directly, suggesting the possibility of a tripartite complex stimulated by LPA. Supporting this, the interaction of mDia1 and INF2 was induced by LPA and dependent on IQGAP1. Our data highlight a unique mechanism of formin action in which mDia1 and INF2 function in series to stabilize MTs and point to IQGAP1 as a scaffold that facilitates the activation of one formin by another.
Collapse
Affiliation(s)
- Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Laura Andres-Delgado
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Xiaoyi Qu
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Sara Nik
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Nagendran Ramalingam
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Leonor Kremer
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Miguel A Alonso
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| |
Collapse
|
71
|
Schill EM, Lake JI, Tusheva OA, Nagy N, Bery SK, Foster L, Avetisyan M, Johnson SL, Stenson WF, Goldstein AM, Heuckeroth RO. Ibuprofen slows migration and inhibits bowel colonization by enteric nervous system precursors in zebrafish, chick and mouse. Dev Biol 2016; 409:473-88. [PMID: 26586201 PMCID: PMC4862364 DOI: 10.1016/j.ydbio.2015.09.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/31/2015] [Accepted: 09/07/2015] [Indexed: 12/17/2022]
Abstract
Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.
Collapse
Affiliation(s)
- Ellen Merrick Schill
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jonathan I Lake
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Olga A Tusheva
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA; Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Saya K Bery
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Lynne Foster
- Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Marina Avetisyan
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Stephen L Johnson
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - William F Stenson
- Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
72
|
Vilboux T, Malicdan MCV, Chang YM, Guo J, Zerfas PM, Stephen J, Cullinane AR, Bryant J, Fischer R, Brooks BP, Zein WM, Wiggs EA, Zalewski CK, Poretti A, Bryan MM, Vemulapalli M, Mullikin JC, Kirby M, Anderson SM, Huizing M, Toro C, Gahl WA, Gunay-Aygun M. Cystic cerebellar dysplasia and biallelic LAMA1 mutations: a lamininopathy associated with tics, obsessive compulsive traits and myopia due to cell adhesion and migration defects. J Med Genet 2016; 53:318-29. [PMID: 27095636 DOI: 10.1136/jmedgenet-2015-103416] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/06/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Laminins are heterotrimeric complexes, consisting of α, β and γ subunits that form a major component of basement membranes and extracellular matrix. Laminin complexes have different, but often overlapping, distributions and functions. METHODS Under our clinical protocol, NCT00068224, we have performed extensive clinical and neuropsychiatric phenotyping, neuroimaging and molecular analysis in patients with laminin α1 (LAMA1)-associated lamininopathy. We investigated the consequence of mutations in LAMA1 using patient-derived fibroblasts and neuronal cells derived from neuronal stem cells. RESULTS In this paper we describe individuals with biallelic mutations in LAMA1, all of whom had the cerebellar dysplasia, myopia and retinal dystrophy, in addition to obsessive compulsive traits, tics and anxiety. Patient-derived fibroblasts have impaired adhesion, reduced migration, abnormal morphology and increased apoptosis due to impaired activation of Cdc42, a member of the Rho family of GTPases that is involved in cytoskeletal dynamics. LAMA1 knockdown in human neuronal cells also showed abnormal morphology and filopodia formation, supporting the importance of LAMA1 in neuronal migration, and marking these cells potentially useful tools for disease modelling and therapeutic target discovery. CONCLUSION This paper broadens the phenotypes associated with LAMA1 mutations. We demonstrate that LAMA1 deficiency can lead to alteration in cytoskeletal dynamics, which may invariably lead to alteration in dendrite growth and axonal formation. Estimation of disease prevalence based on population studies in LAMA1 reveals a prevalence of 1-20 in 1 000 000. TRIAL REGISTRATION NUMBER NCT00068224.
Collapse
Affiliation(s)
- Thierry Vilboux
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA Division of Medical Genomics, Inova Translational Medicine Institute, Falls Church, Virginia, USA
| | - May Christine V Malicdan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA NIH Undiagnosed Diseases Program, Common Fund, National Institutes of Health, Bethesda, Maryland, USA
| | - Yun Min Chang
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer Guo
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Patricia M Zerfas
- Diagnostic and Research Services Branch, Office of Research Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshi Stephen
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew R Cullinane
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA Department of Anatomy, College of Medicine, Howard University, Washington DC, USA
| | - Joy Bryant
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Roxanne Fischer
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian P Brooks
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wadih M Zein
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Edythe A Wiggs
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher K Zalewski
- Audiology Unit, Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrea Poretti
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Melanie M Bryan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Meghana Vemulapalli
- NIH Intramural Sequencing Center (NISC), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James C Mullikin
- NIH Intramural Sequencing Center (NISC), National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Martha Kirby
- Flow Cytometry Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stacie M Anderson
- Flow Cytometry Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Marjan Huizing
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Camilo Toro
- NIH Undiagnosed Diseases Program, Common Fund, National Institutes of Health, Bethesda, Maryland, USA
| | - William A Gahl
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA NIH Undiagnosed Diseases Program, Common Fund, National Institutes of Health, Bethesda, Maryland, USA National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Meral Gunay-Aygun
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
73
|
Dubrac A, Genet G, Ola R, Zhang F, Pibouin-Fragner L, Han J, Zhang J, Thomas JL, Chedotal A, Schwartz MA, Eichmann A. Targeting NCK-Mediated Endothelial Cell Front-Rear Polarity Inhibits Neovascularization. Circulation 2015; 133:409-21. [PMID: 26659946 DOI: 10.1161/circulationaha.115.017537] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Sprouting angiogenesis is a key process driving blood vessel growth in ischemic tissues and an important drug target in a number of diseases, including wet macular degeneration and wound healing. Endothelial cells forming the sprout must develop front-rear polarity to allow sprout extension. The adaptor proteins Nck1 and 2 are known regulators of cytoskeletal dynamics and polarity, but their function in angiogenesis is poorly understood. Here, we show that the Nck adaptors are required for endothelial cell front-rear polarity and migration downstream of the angiogenic growth factors VEGF-A and Slit2. METHODS AND RESULTS Mice carrying inducible, endothelial-specific Nck1/2 deletions fail to develop front-rear polarized vessel sprouts and exhibit severe angiogenesis defects in the postnatal retina and during embryonic development. Inactivation of NCK1 and 2 inhibits polarity by preventing Cdc42 and Pak2 activation by VEGF-A and Slit2. Mechanistically, NCK binding to ROBO1 is required for both Slit2- and VEGF-induced front-rear polarity. Selective inhibition of polarized endothelial cell migration by targeting Nck1/2 prevents hypersprouting induced by Notch or Bmp signaling inhibition, and pathological ocular neovascularization and wound healing, as well. CONCLUSIONS These data reveal a novel signal integration mechanism involving NCK1/2, ROBO1/2, and VEGFR2 that controls endothelial cell front-rear polarity during sprouting angiogenesis.
Collapse
Affiliation(s)
- Alexandre Dubrac
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Gael Genet
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Roxana Ola
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Feng Zhang
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Laurence Pibouin-Fragner
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jinah Han
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jiasheng Zhang
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Jean-Léon Thomas
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Alain Chedotal
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Martin A Schwartz
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.)
| | - Anne Eichmann
- From Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (A.D., G.G., R.O., F.Z., J.H., J.Z., J.-L.T., A.E.); INSERM U1050, Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris (L.P.-F., A.E.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); Institut du Cerveau et de la Moelle, Inserm, Université Pierre et Marie Curie, Paris, France (J.-L.T.); Sorbonne Universités, UPMC Universités Paris 06, INSERM, UMR-S968, CNRS, UMR-7210, Institut de la Vision, France (A.C.); Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT (M.A.S.); and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT (A.E.).
| |
Collapse
|
74
|
Jiang L, Wen J, Luo W. Rho‑associated kinase inhibitor, Y‑27632, inhibits the invasion and proliferation of T24 and 5367 bladder cancer cells. Mol Med Rep 2015; 12:7526-30. [PMID: 26459851 DOI: 10.3892/mmr.2015.4404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 08/17/2015] [Indexed: 11/05/2022] Open
Abstract
The serine/threonine kinases, Rho‑associated protein kinase I and II (ROCK I and II), regulate the cytoskeleton by acting downstream of the small GTPase, Rho, and have been implicated in tumorigenesis and cancer metastasis. Inhibition of ROCK signaling has been shown to suppress the invasion and migration of several types of cancer cells. In this study, the effect of the ROCK inhibitor, Y‑27632, on the proliferation and invasion of T24 and 5637 bladder cancer cells was investigated. In the proliferation assays, the cells were exposed to 0, 10, 25, 50, 75, 100, 125 or 150 µmol/l Y‑27632 and proliferation was determined using Cell Counting kit‑8 after 24, 48 and 72 h. In the invasion assays, the cells were placed in the upper chamber of transwell plates and subjected to 0, 25, 50 or 75 µmol/l Y‑27632 for 24 h, after which invasion was measured. Y‑27632 significantly suppressed the cell proliferation of T24 and 5637 cells in a concentration- and time‑dependent manner. Y‑27632 also inhibited the invasion of T24 and 5637 cells in a concentration‑dependent manner (P<0.001). In addition, Y‑27632 suppressed myosin light chain kinase (MLCK) phosphorylation in T24 and 5637 cells, confirming that it is also a downstream effector of the Rho/ROCK pathway in T24 and 5637 bladder cancer cells. In conclusion, the Rho/ROCK/P‑MLCK pathway may be important in tumor cell metastasis in bladder cancer.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Emergency, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Jiaming Wen
- Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wei Luo
- Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
75
|
The prognostic value of Tiam1 protein expression in head and neck squamous cell carcinoma: a retrospective study. CHINESE JOURNAL OF CANCER 2015; 34:614-21. [PMID: 26369827 PMCID: PMC4593337 DOI: 10.1186/s40880-015-0053-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/02/2015] [Indexed: 12/25/2022]
Abstract
Introduction Head and neck squamous cell carcinoma (HNSCC) is a common cancer worldwide and has a poor prognosis. A biomarker predicting the clinical outcome of HNSCC patients could be useful in guiding treatment planning. Overexpression of the T lymphoma invasion and metastasis 1 (Tiam1) protein has been implicated in the migration and invasion of neoplasms. However, its role in HNSCC progression needs to be further validated. We detected the expression of Tiam1 in normal and tumor tissues and determined its association with clinical outcomes in patients with HNSCC. Methods We measured the expression of Tiam1 in normal and cancerous tissue samples from the patients with HNSCC treated at Sun Yat-sen University Cancer Center between 2001 and 2008. The Tiam1 expression was scored from 0 to 12 based on the percentage of positively stained cells and the staining intensity. We then determined the diagnostic performance of this score in predicting overall survival (OS) and disease-free survival (DFS). Results Of the 194 evaluable patients, those with advanced disease, lymph node metastasis at diagnosis, and recurrence or metastasis during follow-up had a higher tendency of having high Tiam1 expression as compared with their counterparts (P < 0.05). The proportion of samples with high Tiam1 expression was also higher in cancerous tissues than in non-cancerous tissues (57.7% vs. 13.9%, P < 0.001). Cox proportional hazards regression analysis revealed that Tiam1 expression scores of 5 and greater independently predicted short OS and DFS. Conclusion The Tiam1 expression is shown as a promising biomarker of clinical outcomes in patients with HNSCC and should be evaluated in prospective trials.
Collapse
|
76
|
Abstract
Neurons are highly polarized cells with structurally and functionally distinct processes called axons and dendrites. This polarization underlies the directional flow of information in the central nervous system, so the establishment and maintenance of neuronal polarization is crucial for correct development and function. Great progress in our understanding of how neurons establish their polarity has been made through the use of cultured hippocampal neurons, while recent technological advances have enabled in vivo analysis of axon specification and elongation. This short review and accompanying poster highlight recent advances in this fascinating field, with an emphasis on the signaling mechanisms underlying axon and dendrite specification in vitro and in vivo.
Collapse
Affiliation(s)
- Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
77
|
Arden JD, Lavik KI, Rubinic KA, Chiaia N, Khuder SA, Howard MJ, Nestor-Kalinoski AL, Alberts AS, Eisenmann KM. Small-molecule agonists of mammalian Diaphanous-related (mDia) formins reveal an effective glioblastoma anti-invasion strategy. Mol Biol Cell 2015; 26:3704-18. [PMID: 26354425 PMCID: PMC4626057 DOI: 10.1091/mbc.e14-11-1502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 09/04/2015] [Indexed: 12/26/2022] Open
Abstract
Formin agonists impede the most dangerous aspect of glioblastoma—tumor spread into surrounding healthy tissue. Formin activation impairs a novel aspect of the transformed cell and informs the development of antitumor strategies for a cancer needing a more effective therapy. The extensive invasive capacity of glioblastoma (GBM) makes it resistant to surgery, radiotherapy, and chemotherapy and thus makes it lethal. In vivo, GBM invasion is mediated by Rho GTPases through unidentified downstream effectors. Mammalian Diaphanous (mDia) family formins are Rho-directed effectors that regulate the F-actin cytoskeleton to support tumor cell motility. Historically, anti-invasion strategies focused upon mDia inhibition, whereas activation remained unexplored. The recent development of small molecules directly inhibiting or activating mDia-driven F-actin assembly that supports motility allows for exploration of their role in GBM. We used the formin inhibitor SMIFH2 and mDia agonists IMM-01/-02 and mDia2-DAD peptides, which disrupt autoinhibition, to examine the roles of mDia inactivation versus activation in GBM cell migration and invasion in vitro and in an ex vivo brain slice invasion model. Inhibiting mDia suppressed directional migration and spheroid invasion while preserving intrinsic random migration. mDia agonism abrogated both random intrinsic and directional migration and halted U87 spheroid invasion in ex vivo brain slices. Thus mDia agonism is a superior GBM anti-invasion strategy. We conclude that formin agonism impedes the most dangerous GBM component—tumor spread into surrounding healthy tissue. Formin activation impairs novel aspects of transformed cells and informs the development of anti-GBM invasion strategies.
Collapse
Affiliation(s)
- Jessica D Arden
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kari I Lavik
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kaitlin A Rubinic
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Nicolas Chiaia
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Sadik A Khuder
- Departments of Medicine and Public Health and Homeland Security, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Marthe J Howard
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | | | - Arthur S Alberts
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Kathryn M Eisenmann
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614 )
| |
Collapse
|
78
|
Controlling Redox Status for Stem Cell Survival, Expansion, and Differentiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:105135. [PMID: 26273419 PMCID: PMC4530287 DOI: 10.1155/2015/105135] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/06/2014] [Indexed: 01/07/2023]
Abstract
Reactive oxygen species (ROS) have long been considered as pathological agents inducing apoptosis under adverse culture conditions. However, recent findings have challenged this dogma and physiological levels of ROS are now considered as secondary messengers, mediating numerous cellular functions in stem cells. Stem cells represent important tools for tissue engineering, drug screening, and disease modeling. However, the safe use of stem cells for clinical applications still requires culture improvements to obtain functional cells. With the examples of mesenchymal stem cells (MSCs) and pluripotent stem cells (PSCs), this review investigates the roles of ROS in the maintenance of self-renewal, proliferation, and differentiation of stem cells. In addition, this work highlights that the tight control of stem cell microenvironment, including cell organization, and metabolic and mechanical environments, may be an effective approach to regulate endogenous ROS generation. Taken together, this paper indicates the need for better quantification of ROS towards the accurate control of stem cell fate.
Collapse
|
79
|
A phosphorylation switch controls the spatiotemporal activation of Rho GTPases in directional cell migration. Nat Commun 2015; 6:7721. [PMID: 26166433 PMCID: PMC4510974 DOI: 10.1038/ncomms8721] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/04/2015] [Indexed: 12/15/2022] Open
Abstract
Although cell migration plays a central role in development and disease, the underlying molecular mechanism is not fully understood. Here we report that a phosphorylation-mediated molecular switch comprising deleted in liver cancer 1 (DLC1), tensin-3 (TNS3), phosphatase and tensin homologue (PTEN) and phosphoinositide-3-kinase (PI3K) controls the spatiotemporal activation of the small GTPases, Rac1 and RhoA, thereby initiating directional cell migration induced by growth factors. On epidermal growth factor (EGF) or platelet-derived growth factor (PDGF) stimulation, TNS3 and PTEN are phosphorylated at specific Thr residues, which trigger the rearrangement of the TNS3–DLC1 and PTEN–PI3K complexes into the TNS3–PI3K and PTEN–DLC1 complexes. Subsequently, the TNS3–PI3K complex translocates to the leading edge of a migrating cell to promote Rac1 activation, whereas PTEN–DLC1 translocates to the posterior for localized RhoA activation. Our work identifies a core signalling mechanism by which an external motility stimulus is coupled to the spatiotemporal activation of Rac1 and RhoA to drive directional cell migration. Directed cell migration requires spatially regulated activity of GTPases Rac1 and RhoA. Here Cao et al. show that growth factor stimulation promotes phosphorylation of tensin-3 and phosphatase and tensin homologue (PTEN) and their association with PI 3-kinase and deleted in liver cancer 1 (DLC1) to regulate GTPase activity.
Collapse
|
80
|
Furlong H, Smith R, Wang J, Seymour C, Mothersill C, Howe O. Identification of Key Proteins in Human Epithelial Cells Responding to Bystander Signals From Irradiated Trout Skin. Dose Response 2015; 13:1559325815597669. [PMID: 26673684 PMCID: PMC4674182 DOI: 10.1177/1559325815597669] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Radiation-induced bystander signaling has been found to occur in live rainbow trout fish (Oncorhynchus mykiss). This article reports identification of key proteomic changes in a bystander reporter cell line (HaCaT) grown in low-dose irradiated tissue-conditioned media (ITCM) from rainbow trout fish. In vitro explant cultures were generated from the skin of fish previously exposed to low doses (0.1 and 0.5 Gy) of X-ray radiation in vivo. The ITCM was harvested from all donor explant cultures and placed on recipient HaCaT cells to observe any change in protein expression caused by the bystander signals. Proteomic methods using 2-dimensional (2D) gel electrophoresis and mass spectroscopy were employed to screen for novel proteins expressed. The proteomic changes measured in HaCaT cells receiving the ITCM revealed that exposure to 0.5 Gy induced an upregulation of annexin A2 and cingulin and a downregulation of Rho-GDI2, F-actin-capping protein subunit beta, microtubule-associated protein RP/EB family member, and 14-3-3 proteins. The 0.1 Gy dose also induced a downregulation of Rho-GDI2, hMMS19, F-actin-capping protein subunit beta, and microtubule-associated protein RP/EB family member proteins. The proteins reported may influence apoptotic signaling, as the results were suggestive of an induction of cell communication, repair mechanisms, and dysregulation of growth signals.
Collapse
Affiliation(s)
- Hayley Furlong
- DIT Centre for Radiation and Environmental Science, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
- School of Biological Sciences, College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| | - Richard Smith
- Medical Physics and Applied Radiation Sciences, Nuclear Research Building, Hamilton, Canada
| | - Jiaxi Wang
- Queen’s Mass Spectrometry and Proteomics Unit, Department of Chemistry, Queen’s University, Bader Lane, Kingston, Canada
| | - Colin Seymour
- Medical Physics and Applied Radiation Sciences, Nuclear Research Building, Hamilton, Canada
| | - Carmel Mothersill
- Medical Physics and Applied Radiation Sciences, Nuclear Research Building, Hamilton, Canada
| | - Orla Howe
- DIT Centre for Radiation and Environmental Science, Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland
- School of Biological Sciences, College of Sciences and Health, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
81
|
Selamat W, Tay PLF, Baskaran Y, Manser E. The Cdc42 Effector Kinase PAK4 Localizes to Cell-Cell Junctions and Contributes to Establishing Cell Polarity. PLoS One 2015; 10:e0129634. [PMID: 26068882 PMCID: PMC4466050 DOI: 10.1371/journal.pone.0129634] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/11/2015] [Indexed: 01/22/2023] Open
Abstract
The serine/threonine kinase PAK4 is a Cdc42 effector whose role is not well understood; overexpression of PAK4 has been associated with some cancers, and there are reports that correlate kinase level with increased cell migration in vitro. Here we report that PAK4 is primarily associated with cell-cell junctions in all the cell lines we tested, and fails to accumulate at focal adhesions or at the leading edge of migrating cells. In U2OS osteosarcoma and MCF-7 breast cancer cell lines, PAK4 depletion did not affect collective cell migration, but affected cell polarization. By contrast, Cdc42 depletion (as reported by many studies) caused a strong defect in junctional assembly in multiple cells lines. We also report that the depletion of PAK4 protein or treatment of cells with the PAK4 inhibitor PF-3758309 can lead to defects in centrosome reorientation (polarization) after cell monolayer wounding. These experiments are consistent with PAK4 forming part of a conserved cell-cell junctional polarity Cdc42 complex. We also confirm β-catenin as a target for PAK4 in these cells. Treatment of cells with PF-3758309 caused inhibition of β-catenin Ser-675 phosphorylation, which is located predominantly at cell-cell junctions.
Collapse
Affiliation(s)
- Widyawilis Selamat
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei-Ling Felicia Tay
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yohendran Baskaran
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ed Manser
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
82
|
Lim SY, Mah W. Abnormal Astrocytosis in the Basal Ganglia Pathway of Git1(-/-) Mice. Mol Cells 2015; 38:540-7. [PMID: 25997734 PMCID: PMC4469912 DOI: 10.14348/molcells.2015.0041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 11/27/2022] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, affecting approximately 5% of children. However, the neural mechanisms underlying its development and treatment are yet to be elucidated. In this study, we report that an ADHD mouse model, which harbors a deletion in the Git1 locus, exhibits severe astrocytosis in the globus pallidus (GP) and thalamic reticular nucleus (TRN), which send modulatory GABAergic inputs to the thalamus. A moderate level of astrocytosis was displayed in other regions of the basal ganglia pathway, including the ventrobasal thalamus and cortex, but not in other brain regions, such as the caudate putamen, basolateral amygdala, and hippocampal CA1. This basal ganglia circuit-selective astrocytosis was detected in both in adult (2-3 months old) and juvenile (4 weeks old) Git1(-/-) mice, suggesting a developmental origin. Astrocytes play an active role in the developing synaptic circuit; therefore, we performed an immunohistochemical analysis of synaptic markers. We detected increased and decreased levels of GABA and parvalbumin (PV), respectively, in the GP. This suggests that astrocytosis may alter synaptic transmission in the basal ganglia. Intriguingly, increased GABA expression colocalized with the astrocyte marker, GFAP, indicative of an astrocytic origin. Collectively, these results suggest that defects in basal ganglia circuitry, leading to impaired inhibitory modulation of the thalamus, are neural correlates for the ADHD-associated behavioral manifestations in Git1(-/-) mice.
Collapse
Affiliation(s)
- Soo-Yeon Lim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412,
Korea
| | - Won Mah
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412,
Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| |
Collapse
|
83
|
Chiu CT, Liao CK, Shen CC, Tang TK, Jow GM, Wang HS, Wu JC. HYS-32-Induced Microtubule Catastrophes in Rat Astrocytes Involves the PI3K-GSK3beta Signaling Pathway. PLoS One 2015; 10:e0126217. [PMID: 25938237 PMCID: PMC4418738 DOI: 10.1371/journal.pone.0126217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/31/2015] [Indexed: 02/06/2023] Open
Abstract
HYS-32 is a novel derivative of combretastatin-A4 (CA-4) previously shown to induce microtubule coiling in rat primary astrocytes. In this study, we further investigated the signaling mechanism and EB1, a microtubule-associated end binding protein, involved in HYS-32-induced microtubule catastrophes. Confocal microscopy with double immunofluorescence staining revealed that EB1 accumulates at the growing microtubule plus ends, where they exhibit a bright comet-like staining pattern in control astrocytes. HYS-32 induced microtubule catastrophes in both a dose- and time-dependent manner and dramatically increased the distances between microtubule tips and the cell border. Treatment of HYS-32 (5 μM) eliminated EB1 localization at the microtubule plus ends and resulted in an extensive redistribution of EB1 to the microtubule lattice without affecting the β-tubulin or EB1 protein expression. Time-lapse experiments with immunoprecipitation further displayed that the association between EB-1 and β-tubulin was significantly decreased following a short-term treatment (2 h), but gradually increased in a prolonged treatment (6-24 h) with HYS-32. Further, HYS-32 treatment induced GSK3β phosphorylation at Y216 and S9, where the ratio of GSK3β-pY216 to GSK3β-pS9 was first elevated followed by a decrease over time. Co-treatment of astrocytes with HYS-32 and GSK3β inhibitor SB415286 attenuated the HYS-32-induced microtubule catastrophes and partially prevented EB1 dissociation from the plus end of microtubules. Furthermore, co-treatment with PI3K inhibitor LY294002 inhibited HYS-32-induced GSK3β-pS9 and partially restored EB1 distribution from the microtubule lattice to plus ends. Together these findings suggest that HYS-32 induces microtubule catastrophes by preventing EB1 from targeting to microtubule plus ends through the GSK3β signaling pathway.
Collapse
Affiliation(s)
- Chi-Ting Chiu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chih-Kai Liao
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chien-Chang Shen
- Division of Medicinal Chemistry, National Research Institute of Chinese Medicine, Taipei 11221, Taiwan
| | - Tswen-Kei Tang
- Department of Nursing, College of Health and Nursing, National Quemoy University, Kinmen 89250, Taiwan
| | - Guey-Mei Jow
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan
| | - Hwai-Shi Wang
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Jiahn-Chun Wu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
84
|
Yamaoka M, Ishizaki T, Kimura T. Interplay between Rab27a effectors in pancreatic β-cells. World J Diabetes 2015; 6:508-516. [PMID: 25897360 PMCID: PMC4398906 DOI: 10.4239/wjd.v6.i3.508] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/24/2014] [Accepted: 02/09/2015] [Indexed: 02/05/2023] Open
Abstract
The small GTPase Rab27a is a member of the Rab family that is involved in membrane trafficking in various kinds of cells. Rab27a has GTP- and GDP-bound forms, and their interconversion regulates intracellular signaling pathways. Typically, only a GTP-bound GTPase binds its specific effectors with the resulting downstream signals controlling specific cellular functions. We previously identified novel Rab27a-interacting proteins. Surprisingly, some of these proteins interacted with GDP-bound Rab27a. The present study reviews recent progress in our understanding of the roles of Rab27a and its effectors in the secretory process. In pancreatic β-cells, GTP-bound Rab27a regulates insulin secretion at the pre-exocytotic stages via its GTP-specific effectors such as Exophilin8/Slac2-c/MyRIP and Slp4/Granuphilin. Glucose stimulation causes insulin exocytosis. Glucose stimulation also converts Rab27a from its GTP- to its GDP-bound form. GDP-bound Rab27a interacts with GDP-specific effectors and controls endocytosis of the secretory membrane. Thus, Rab27a cycling between GTP- and GDP-bound forms synchronizes with the recycling of secretory membrane to re-use the membrane and keep the β-cell volume constant.
Collapse
|
85
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
86
|
Wang Y, Rouabhia M, Lavertu D, Zhang Z. Pulsed electrical stimulation modulates fibroblasts' behaviour through the Smad signalling pathway. J Tissue Eng Regen Med 2015; 11:1110-1121. [PMID: 25712595 DOI: 10.1002/term.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/11/2014] [Accepted: 01/15/2015] [Indexed: 01/14/2023]
Abstract
The aim of this study was to investigate the healing characteristics and the underlying signalling pathway of human dermal fibroblasts under the influence of pulsed electrical stimulation (PES). Primary human dermal fibroblasts were seeded on polypyrrole-coated polyester fabrics and subjected to four different PES protocols. The parameters of the rectangular pulse included potential intensity (50 and 100 mV/mm) and stimulation time (pulse width 300 s within a period of 600 s, and pulse width 10 s within a period of 1200 s). Our study revealed that PES moderately improved the ability of the cells to migrate in association with a statistically significant (p < 0.05) increase of FGF2 secretion by the PES-exposed fibroblasts. These exposed fibroblasts were able to contract collagen gel matrix up to 48 h and this collagen gel contraction paralleled an increase in α-SMA mRNA expression and protein production from the PES-exposed fibroblasts. Interestingly, the effect of PES on the human fibroblasts involved the Smad signalling pathway, as we observed higher levels of phosphorylated Smad2 and Smad3 in the stimulated groups compared to the control groups. Overall, this study demonstrated that PES modulates fibroblast activities through the Smad signalling pathway, thus providing new mechanistic insights related to the use of PES to promote wound healing in humans. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yongliang Wang
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada.,Axe Médecine régénératrice, Centre de Recherche du CHU de Québec, Département de Chirurgie, Faculté de Médecine, Université Laval, QC, Canada
| | - Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada
| | - Denis Lavertu
- Département de Chirurgie Plastique, Hôpital Saint-François d'Assise, Québec, Canada
| | - Ze Zhang
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec, Département de Chirurgie, Faculté de Médecine, Université Laval, QC, Canada
| |
Collapse
|
87
|
RhoGTPases as key players in mammalian cell adaptation to microgravity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:747693. [PMID: 25649831 PMCID: PMC4310447 DOI: 10.1155/2015/747693] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/14/2014] [Accepted: 09/09/2014] [Indexed: 01/03/2023]
Abstract
A growing number of studies are revealing that cells reorganize their cytoskeleton when exposed to conditions of microgravity. Most, if not all, of the structural changes observed on flown cells can be explained by modulation of RhoGTPases, which are mechanosensitive switches responsible for cytoskeletal dynamics control. This review identifies general principles defining cell sensitivity to gravitational stresses. We discuss what is known about changes in cell shape, nucleus, and focal adhesions and try to establish the relationship with specific RhoGTPase activities. We conclude by considering the potential relevance of live imaging of RhoGTPase activity or cytoskeletal structures in order to enhance our understanding of cell adaptation to microgravity-related conditions.
Collapse
|
88
|
Zhao ZH, Tian Y, Yang JP, Zhou J, Chen KS. RhoC, vascular endothelial growth factor and microvascular density in esophageal squamous cell carcinoma. World J Gastroenterol 2015; 21:905-912. [PMID: 25624724 PMCID: PMC4299343 DOI: 10.3748/wjg.v21.i3.905] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/23/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the expression of Ras homolog (Rho)C, vascular endothelial growth factor (VEGF) and CD105 in esophageal squamous cell carcinoma.
METHODS: Semi-quantitative reverse transcriptase polymerase chain reaction, in situ hybridization and immunohistochemical streptavidin-biotin- peroxidase methods were used to detect expression of RhoC mRNA and protein, and VEGF protein in 62 cases with esophageal squamous cell carcinoma, 31 cases with adjacent atypical hyperplastic tissues, and 62 cases with normal esophageal mucosa. CD105 antibody labeling was used to measure microvascular density. Expression levels were compared according to clinicopathologic and patient parameters.
RESULTS: Expression of RhoC mRNA showed a positive correlation with the protein level in esophageal squamous cell carcinoma, as well as with VEGF protein levels. RhoC mRNA expression was mainly located within the cytoplasm of the tumor cells, appearing as blue to purple particles by in situ hybridization. The differences in RhoC mRNA expression in esophageal squamous cell carcinoma, adjacent atypical hyperplasia and normal esophageal mucosa were significant (P < 0.05). The relative expression of RhoC mRNA in cancer tissues with lymph node metastasis was significantly higher than in the tissues without lymph node metastasis (P < 0.05). VEGF protein expression was consistent with microvascular density (t = 25.52, P < 0.05). Positive expression of VEGF protein in esophageal squamous cell carcinoma of different histologic gradings did not differ significantly. Positive expression of VEGF protein in carcinoma tissues with deep infiltration was significantly higher than in tissues with only superficial infiltration (P < 0.05). The positive expression of VEGF protein in cancer tissues with lymph node metastasis was significantly higher than in the tissues without lymph node metastasis (P < 0.05).
CONCLUSION: RhoC protein may upregulate VEGF expression, thereby promoting tumor angiogenesis. RhoC mRNA and protein expression was correlated with metastasis.
Collapse
|
89
|
Imaoka H, Toiyama Y, Saigusa S, Kawamura M, Kawamoto A, Okugawa Y, Hiro J, Tanaka K, Inoue Y, Mohri Y, Kusunoki M. RacGAP1 expression, increasing tumor malignant potential, as a predictive biomarker for lymph node metastasis and poor prognosis in colorectal cancer. Carcinogenesis 2015; 36:346-54. [PMID: 25568185 DOI: 10.1093/carcin/bgu327] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Rac GTPase-activating protein (RacGAP) 1 plays a key role in controlling various cellular phenomena including cytokinesis, transformation, invasive migration and metastasis. This study investigated the function and clinical significance of RacGAP1 expression in colorectal cancer (CRC). The intrinsic functions of RacGAP1 in CRC cells were analyzed using small interfering RNA (siRNA). We analyzed RacGAP1 mRNA expression in surgical specimens from 193 CRC patients (Cohort 1) by real-time PCR. Finally, we validated RacGAP1 protein expression using formalin-fixed paraffin-embedded samples from 298 CRC patients (Cohort 2) by immunohistochemistry. Reduced RacGAP1 expression by siRNA in CRC cell lines showed significantly decreased cellular proliferation, migration and invasion. In Cohort 1, RacGAP1 expression in CRC was significantly higher than in adjacent normal mucosa and increased according to tumor node metastasis stage progression. High RacGAP1 expression in tumors was significantly associated with progression and prognosis. In Cohort 2, RacGAP1 protein was overexpressed mainly in the nuclei of CRC cells; however, its expression was scarcely observed in normal colorectal mucosa. RacGAP1 protein expression was significantly higher in CRC patients with higher T stage, vessel invasion and lymph node and distant metastasis. Increased expression of RacGAP1 protein was significantly associated with poor disease-free and overall survival. Multivariate analyses revealed that high RacGAP1 expression was an independent predictive marker for lymph node metastasis, recurrence and poor prognosis in CRC. Our data provide novel evidence for the biological and clinical significance of RacGAP1 as a potential biomarker for identifying patients with lymph node metastasis and poor prognosis in CRC.
Collapse
Affiliation(s)
- Hiroki Imaoka
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Susumu Saigusa
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Mikio Kawamura
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Aya Kawamoto
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Junichiro Hiro
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Koji Tanaka
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Yasuhiro Inoue
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Yasuhiko Mohri
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Masato Kusunoki
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| |
Collapse
|
90
|
|
91
|
Matsui T, Watanabe T, Matsuzawa K, Kakeno M, Okumura N, Sugiyama I, Itoh N, Kaibuchi K. PAR3 and aPKC regulate Golgi organization through CLASP2 phosphorylation to generate cell polarity. Mol Biol Cell 2014; 26:751-61. [PMID: 25518939 PMCID: PMC4325844 DOI: 10.1091/mbc.e14-09-1382] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A PAR complex (PAR3, PAR6, and aPKC) plays a central role in the establishment of cell polarity. Another polarity protein, CLASP2, binds directly with PAR3 and is phosphorylated by aPKC. Through CLASP2 phosphorylation, aPKC and PAR3 regulate the localization of CLASP2 to the trans-Golgi network, thereby controlling the Golgi organization. The organization of the Golgi apparatus is essential for cell polarization and its maintenance. The polarity regulator PAR complex (PAR3, PAR6, and aPKC) plays critical roles in several processes of cell polarization. However, how the PAR complex participates in regulating the organization of the Golgi remains largely unknown. Here we demonstrate the functional cross-talk of the PAR complex with CLASP2, which is a microtubule plus-end–tracking protein and is involved in organizing the Golgi ribbon. CLASP2 directly interacted with PAR3 and was phosphorylated by aPKC. In epithelial cells, knockdown of either PAR3 or aPKC induced the aberrant accumulation of CLASP2 at the trans-Golgi network (TGN) concomitantly with disruption of the Golgi ribbon organization. The expression of a CLASP2 mutant that inhibited the PAR3-CLASP2 interaction disrupted the organization of the Golgi ribbon. CLASP2 is known to localize to the TGN through its interaction with the TGN protein GCC185. This interaction was inhibited by the aPKC-mediated phosphorylation of CLASP2. Furthermore, the nonphosphorylatable mutant enhanced the colocalization of CLASP2 with GCC185, thereby perturbing the Golgi organization. On the basis of these observations, we propose that PAR3 and aPKC control the organization of the Golgi through CLASP2 phosphorylation.
Collapse
Affiliation(s)
- Toshinori Matsui
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kenji Matsuzawa
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mai Kakeno
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nobumasa Okumura
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Ikuko Sugiyama
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Norimichi Itoh
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
92
|
Shah B, Püschel AW. In vivo functions of small GTPases in neocortical development. Biol Chem 2014; 395:465-76. [PMID: 24391191 DOI: 10.1515/hsz-2013-0277] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/24/2013] [Indexed: 11/15/2022]
Abstract
The complex mammalian cortex develops from a simple neuroepithelium through the proliferation of neuronal progenitors, their asymmetric division and cell migration. Newly generated neurons transiently assume a multipolar morphology before they polarize to form a trailing axon and a leading process that is required for their radial migration. The polarization and migration events during cortical development are under the control of multiple signaling cascades that coordinate the different cellular processes involved in neuronal differentiation. GTPases perform essential functions at different stages of neuronal development as central components of these pathways. They have been widely studied using cell lines and primary neuronal cultures but their physiological function in vivo still remains to be explored in many cases. Here we review the function of GTPases that have been studied genetically by the analysis of the embryonic nervous system in knockout mice. The phenotype of these mutants has highlighted the importance of GTPases for different steps of development by orchestrating cytoskeletal rearrangements and neuronal polarization.
Collapse
|
93
|
Darido C, Jane SM. Grhl3 and GEF19 in the front rho. Small GTPases 2014; 1:104-107. [PMID: 21686262 DOI: 10.4161/sgtp.1.2.13620] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 11/19/2022] Open
Abstract
Directional migration is a critical component of cell motility is observed in many diverse processes including embryogenesis, immune surveillance and wound repair. A central aspect of directional migration is cellular polarity, which is established through several signaling pathways that converge on the small GTPases. These factors orchestrate precise spatial and temporal organization of the actin cytoskeleton at the leading edge of the cell, and induce polarized capture and stabilization of microtubules and their associated microtubule organizing center (MTOC). Studies of the regulation of the GTPases have predominantly focused on post-translational mechanisms involving guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and guanine nucleotide dissociation inhibitors (GDIs). In this commentary, we examine the transcriptional regulation of these factors, focusing on the recently described regulation of RhoGEF19, an activator of RhoA, by the epidermal-specific transcription factor GRHL3, and the importance of this regulatory mechanism in wound repair. Our findings establish novel links between epidermal cell migration in wound healing and the planar cell polarity (PCP) signaling pathway, and establish a paradigm for tissue-specific regulation of Rho GTPase activity.
Collapse
Affiliation(s)
- Charbel Darido
- Rotary Bone Marrow Research Laboratories; Parkville, Victoria Australia
| | | |
Collapse
|
94
|
Ko HK, Guo LW, Su B, Gao L, Gelman IH. Suppression of chemotaxis by SSeCKS via scaffolding of phosphoinositol phosphates and the recruitment of the Cdc42 GEF, Frabin, to the leading edge. PLoS One 2014; 9:e111534. [PMID: 25356636 PMCID: PMC4214753 DOI: 10.1371/journal.pone.0111534] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/28/2014] [Indexed: 01/09/2023] Open
Abstract
Chemotaxis is controlled by interactions between receptors, Rho-family GTPases, phosphatidylinositol 3-kinases, and cytoskeleton remodeling proteins. We investigated how the metastasis suppressor, SSeCKS, attenuates chemotaxis. Chemotaxis activity inversely correlated with SSeCKS levels in mouse embryo fibroblasts (MEF), DU145 and MDA-MB-231 cancer cells. SSeCKS loss induced chemotactic velocity and linear directionality, correlating with replacement of leading edge lamellipodia with fascin-enriched filopodia-like extensions, the formation of thickened longitudinal F-actin stress fibers reaching to filopodial tips, relative enrichments at the leading edge of phosphatidylinositol (3,4,5)P3 (PIP3), Akt, PKC-ζ, Cdc42-GTP and active Src (SrcpoY416), and a loss of Rac1. Leading edge lamellipodia and chemotaxis inhibition in SSeCKS-null MEF could be restored by full-length SSeCKS or SSeCKS deleted of its Src-binding domain (ΔSrc), but not by SSeCKS deleted of its three MARCKS (myristylated alanine-rich C kinase substrate) polybasic domains (ΔPBD), which bind PIP2 and PIP3. The enrichment of activated Cdc42 in SSeCKS-null leading edge filopodia correlated with recruitment of the Cdc42-specific guanine nucleotide exchange factor, Frabin, likely recruited via multiple PIP2/3-binding domains. Frabin knockdown in SSeCKS-null MEF restores leading edge lamellipodia and chemotaxis inhibition. However, SSeCKS failed to co-immunoprecipitate with Rac1, Cdc42 or Frabin. Consistent with the notion that chemotaxis is controlled by SSeCKS-PIP (vs. -Src) scaffolding activity, constitutively-active phosphatidylinositol 3-kinase could override the ability of the Src inhibitor, SKI-606, to suppress chemotaxis and filopodial enrichment of Frabin in SSeCKS-null MEF. Our data suggest a role for SSeCKS in controlling Rac1 vs. Cdc42-induced cellular dynamics at the leading chemotactic edge through the scaffolding of phospholipids and signal mediators, and through the reorganization of the actin cytoskeleton controlling directional movement.
Collapse
Affiliation(s)
- Hyun-Kyung Ko
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Li-wu Guo
- Div. of Genetic & Reproductive Toxicology, National Center for Toxicological Research, Jefferson, Arkansas, United States of America
| | - Bing Su
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Lingqiu Gao
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Irwin H. Gelman
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
95
|
Wilmes A, Bielow C, Ranninger C, Bellwon P, Aschauer L, Limonciel A, Chassaigne H, Kristl T, Aiche S, Huber CG, Guillou C, Hewitt P, Leonard MO, Dekant W, Bois F, Jennings P. Mechanism of cisplatin proximal tubule toxicity revealed by integrating transcriptomics, proteomics, metabolomics and biokinetics. Toxicol In Vitro 2014; 30:117-27. [PMID: 25450742 DOI: 10.1016/j.tiv.2014.10.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/18/2014] [Accepted: 10/02/2014] [Indexed: 11/19/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for the treatment of solid tumours. The major dose-limiting factor is nephrotoxicity, in particular in the proximal tubule. Here, we use an integrated omics approach, including transcriptomics, proteomics and metabolomics coupled to biokinetics to identify cell stress response pathways induced by cisplatin. The human renal proximal tubular cell line RPTEC/TERT1 was treated with sub-cytotoxic concentrations of cisplatin (0.5 and 2 μM) in a daily repeat dose treating regime for up to 14 days. Biokinetic analysis showed that cisplatin was taken up from the basolateral compartment, transported to the apical compartment, and accumulated in cells over time. This is in line with basolateral uptake of cisplatin via organic cation transporter 2 and bioactivation via gamma-glutamyl transpeptidase located on the apical side of proximal tubular cells. Cisplatin affected several pathways including, p53 signalling, Nrf2 mediated oxidative stress response, mitochondrial processes, mTOR and AMPK signalling. In addition, we identified novel pathways changed by cisplatin, including eIF2 signalling, actin nucleation via the ARP/WASP complex and regulation of cell polarization. In conclusion, using an integrated omic approach together with biokinetics we have identified both novel and established mechanisms of cisplatin toxicity.
Collapse
Affiliation(s)
- Anja Wilmes
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria.
| | - Chris Bielow
- Institute of Computer Science, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin 14195, Germany
| | - Christina Ranninger
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg 5020, Austria
| | - Patricia Bellwon
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Lydia Aschauer
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Alice Limonciel
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Hubert Chassaigne
- European Commission, Joint Research Centre (JRC), Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Via Enrico Fermi 2749, I-21027 Ispra, Italy
| | - Theresa Kristl
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg 5020, Austria
| | - Stephan Aiche
- Institute of Computer Science, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin 14195, Germany
| | - Christian G Huber
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Claude Guillou
- European Commission, Joint Research Centre (JRC), Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Via Enrico Fermi 2749, I-21027 Ispra, Italy
| | - Philipp Hewitt
- Merck KGaA, Merck Serono, Nonclinical Safety, Darmstadt 64293, Germany
| | - Martin O Leonard
- Centre for Radiation, Chemical and Environmental Hazard, Public Health England, Chilton, Didcot OX11 0RQ, UK
| | - Wolfgang Dekant
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Frederic Bois
- Université de Technologie de Compiègne, Compiègne Cedex 60205, France
| | - Paul Jennings
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
96
|
Liu Q, Song B. Electric field regulated signaling pathways. Int J Biochem Cell Biol 2014; 55:264-8. [DOI: 10.1016/j.biocel.2014.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 02/01/2023]
|
97
|
Huda S, Pilans D, Makurath M, Hermans T, Kandere-Grzybowska K, Grzybowski BA. Microfabricated Systems and Assays for Studying the Cytoskeletal Organization, Micromechanics, and Motility Patterns of Cancerous Cells. ADVANCED MATERIALS INTERFACES 2014; 1:1400158. [PMID: 26900544 PMCID: PMC4757490 DOI: 10.1002/admi.201400158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cell motions are driven by coordinated actions of the intracellular cytoskeleton - actin, microtubules (MTs) and substrate/focal adhesions (FAs). This coordination is altered in metastatic cancer cells resulting in deregulated and increased cellular motility. Microfabrication tools, including photolithography, micromolding, microcontact printing, wet stamping and microfluidic devices have emerged as a powerful set of experimental tools with which to probe and define the differences in cytoskeleton organization/dynamics and cell motility patterns in non-metastatic and metastatic cancer cells. In this review, we discuss four categories of microfabricated systems: (i) micropatterned substrates for studying of cell motility sub-processes (for example, MT targeting of FAs or cell polarization); (ii) systems for studying cell mechanical properties, (iii) systems for probing overall cell motility patterns within challenging geometric confines relevant to metastasis (for example, linear and ratchet geometries), and (iv) microfluidic devices that incorporate co-cultures of multiple cells types and chemical gradients to mimic in vivo intravasation/extravasation steps of metastasis. Together, these systems allow for creating controlled microenvironments that not only mimic complex soft tissues, but are also compatible with live cell high-resolution imaging and quantitative analysis of single cell behavior.
Collapse
Affiliation(s)
- Sabil Huda
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Didzis Pilans
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Monika Makurath
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Thomas Hermans
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Kristiana Kandere-Grzybowska
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Bartosz A Grzybowski
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA; Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| |
Collapse
|
98
|
Poliakova K, Adebola A, Leung CL, Favre B, Liem RKH, Schepens I, Borradori L. BPAG1a and b associate with EB1 and EB3 and modulate vesicular transport, Golgi apparatus structure, and cell migration in C2.7 myoblasts. PLoS One 2014; 9:e107535. [PMID: 25244344 PMCID: PMC4171495 DOI: 10.1371/journal.pone.0107535] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 08/18/2014] [Indexed: 11/19/2022] Open
Abstract
BPAG1a and BPAG1b (BPAG1a/b) constitute two major isoforms encoded by the dystonin (Dst) gene and show homology with MACF1a and MACF1b. These proteins are members of the plakin family, giant multi-modular proteins able to connect the intermediate filament, microtubule and microfilament cytoskeletal networks with each other and to distinct cell membrane sites. They also serve as scaffolds for signaling proteins that modulate cytoskeletal dynamics. To gain better insights into the functions of BPAG1a/b, we further characterized their C-terminal region important for their interaction with microtubules and assessed the role of these isoforms in the cytoskeletal organization of C2.7 myoblast cells. Our results show that alternative splicing does not only occur at the 5′ end of Dst and Macf1 pre-mRNAs, as previously reported, but also at their 3′ end, resulting in expression of additional four mRNA variants of BPAG1 and MACF1. These isoform-specific C-tails were able to bundle microtubules and bound to both EB1 and EB3, two microtubule plus end proteins. In the C2.7 cell line, knockdown of BPAG1a/b had no major effect on the organization of the microtubule and microfilament networks, but negatively affected endocytosis and maintenance of the Golgi apparatus structure, which became dispersed. Finally, knockdown of BPAG1a/b caused a specific decrease in the directness of cell migration, but did not impair initial cell adhesion. These data provide novel insights into the complexity of alternative splicing of Dst pre-mRNAs and into the role of BPAG1a/b in vesicular transport, Golgi apparatus structure as well as in migration in C2.7 myoblasts.
Collapse
Affiliation(s)
- Kseniia Poliakova
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- * E-mail:
| | - Adijat Adebola
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Conrad L. Leung
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Bertrand Favre
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Ronald K. H. Liem
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Isabelle Schepens
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Luca Borradori
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
99
|
Boczonadi V, Gillespie R, Keenan I, Ramsbottom SA, Donald-Wilson C, Al Nazer M, Humbert P, Schwarz RJ, Chaudhry B, Henderson DJ. Scrib:Rac1 interactions are required for the morphogenesis of the ventricular myocardium. Cardiovasc Res 2014; 104:103-15. [PMID: 25139745 PMCID: PMC4174891 DOI: 10.1093/cvr/cvu193] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aims The organization and maturation of ventricular cardiomyocytes from the embryonic to the adult form is crucial for normal cardiac function. We have shown that a polarity protein, Scrib, may be involved in regulating the early stages of this process. Our goal was to establish whether Scrib plays a cell autonomous role in the ventricular myocardium, and whether this involves well-known polarity pathways. Methods and results Deletion of Scrib in cardiac precursors utilizing Scribflox mice together with the Nkx2.5-Cre driver resulted in disruption of the cytoarchitecture of the forming trabeculae and ventricular septal defects. Although the majority of mice lacking Scrib in the myocardium survived to adulthood, they developed marked cardiac fibrosis. Scrib did not physically interact with the planar cell polarity (PCP) protein, Vangl2, in early cardiomyocytes as it does in other tissues, suggesting that the anomalies did not result from disruption of PCP signalling. However, Scrib interacted with Rac1 physically in embryonic cardiomyocytes and genetically to result in ventricular abnormalities, suggesting that this interaction is crucial for the development of the early myocardium. Conclusions The Scrib–Rac1 interaction plays a crucial role in the organization of developing cardiomyocytes and formation of the ventricular myocardium. Thus, we have identified a novel signalling pathway in the early, functioning, heart muscle. These data also show that the foetus can recover from relatively severe abnormalities in prenatal ventricular development, although cardiac fibrosis can be a long-term consequence.
Collapse
Affiliation(s)
- Veronika Boczonadi
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rachel Gillespie
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Iain Keenan
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Simon A Ramsbottom
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | | | - Mariana Al Nazer
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Patrick Humbert
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Australia Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Melbourne, Australia Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Melbourne, Australia Department of Pathology, University of Melbourne, Parkville, Melbourne, Australia
| | | | - Bill Chaudhry
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Deborah J Henderson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
100
|
Yan G, Zou R, Chen Z, Fan B, Wang Z, Wang Y, Yin X, Zhang D, Tong L, Yang F, Jiang W, Fu W, Zheng J, Bergo MO, Dalin M, Zheng J, Chen S, Zhou J. Silencing RhoA inhibits migration and invasion through Wnt/β-catenin pathway and growth through cell cycle regulation in human tongue cancer. Acta Biochim Biophys Sin (Shanghai) 2014; 46:682-90. [PMID: 25001480 DOI: 10.1093/abbs/gmu051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ras homolog gene family member A (RhoA) has been identified as a critical regulator of tumor aggressive behavior. In this study, we assessed the role of RhoA in the mechanisms underlying growth, migration, and invasion of squamous cell carcinoma of tongue (TSCC). Stable RhoA knockdown of TSCC cell lines SCC-4 and CAL27 were achieved using Lentiviral transfection. The effects of RhoA depletion on cell migration, invasion, and cell proliferation were determined. The possible underlying mechanism of RhoA depletion on TSCC cell line was also evaluated by determining the expression of Galectin-3 (Gal-3), β-catenin, and matrix metalloproteinase-9 (MMP-9) in vivo. Meanwhile, the underlying mechanism of TSCC growth was studied by analysis of cyclin D1/2, p21CIP1/WAF1, and p27Kip1 protein levels. Immunohistochemical assessments were performed to further prove the alteration of Gal-3 and β-catenin expression. We found that, in mice injected with human TSCC cells in the tongue, RhoA levels were higher in primary tumors and metastasized lymph nodes compared with those in the normal tissues. Silencing of RhoA significantly reduced the tumor growth, decreased the levels of Gal-3, β-catenin, MMP-9, and cyclin D1/2, and increased the levels of p21CIP1/WAF1 and p27Kip1. In vitro, RhoA knockdown also led to inhibition of cell migration, invasion, and proliferation. Our data suggest that RhoA plays a significant role in TSCC progression by regulating cell migration and invasion through Wnt/β-catenin signaling pathway and cell proliferation through cell cycle regulation, respectively. RhoA might be a novel therapeutic target of TSCC.
Collapse
|