101
|
Wu J, Kent IA, Shekhar N, Chancellor TJ, Mendonca A, Dickinson RB, Lele TP. Actomyosin pulls to advance the nucleus in a migrating tissue cell. Biophys J 2014; 106:7-15. [PMID: 24411232 DOI: 10.1016/j.bpj.2013.11.4489] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 10/29/2013] [Accepted: 11/19/2013] [Indexed: 01/14/2023] Open
Abstract
The cytoskeletal forces involved in translocating the nucleus in a migrating tissue cell remain unresolved. Previous studies have variously implicated actomyosin-generated pushing or pulling forces on the nucleus, as well as pulling by nucleus-bound microtubule motors. We found that the nucleus in an isolated migrating cell can move forward without any trailing-edge detachment. When a new lamellipodium was triggered with photoactivation of Rac1, the nucleus moved toward the new lamellipodium. This forward motion required both nuclear-cytoskeletal linkages and myosin activity. Apical or basal actomyosin bundles were found not to translate with the nucleus. Although microtubules dampen fluctuations in nuclear position, they are not required for forward translocation of the nucleus during cell migration. Trailing-edge detachment and pulling with a microneedle produced motion and deformation of the nucleus suggestive of a mechanical coupling between the nucleus and the trailing edge. Significantly, decoupling the nucleus from the cytoskeleton with KASH overexpression greatly decreased the frequency of trailing-edge detachment. Collectively, these results explain how the nucleus is moved in a crawling fibroblast and raise the possibility that forces could be transmitted from the front to the back of the cell through the nucleus.
Collapse
Affiliation(s)
- Jun Wu
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - Ian A Kent
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - Nandini Shekhar
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - T J Chancellor
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - Agnes Mendonca
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, Florida
| | - Tanmay P Lele
- Department of Chemical Engineering, University of Florida, Gainesville, Florida.
| |
Collapse
|
102
|
Crespo CL, Vernieri C, Keller PJ, Garrè M, Bender JR, Wittbrodt J, Pardi R. The PAR complex controls the spatiotemporal dynamics of F-actin and the MTOC in directionally migrating leukocytes. J Cell Sci 2014; 127:4381-95. [PMID: 25179599 PMCID: PMC4197085 DOI: 10.1242/jcs.146217] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inflammatory cells acquire a polarized phenotype to migrate towards sites of infection or injury. A conserved polarity complex comprising PAR-3, PAR-6 and atypical protein kinase C (aPKC) relays extracellular polarizing cues to control cytoskeletal and signaling networks affecting morphological and functional polarization. However, there is no evidence that myeloid cells use PAR signaling to migrate vectorially in three-dimensional (3D) environments in vivo. Using genetically encoded bioprobes and high-resolution live imaging, we reveal the existence of F-actin oscillations in the trailing edge and constant repositioning of the microtubule organizing center (MTOC) to direct leukocyte migration in wounded medaka fish larvae (Oryzias latipes). Genetic manipulation in live myeloid cells demonstrates that the catalytic activity of aPKC and the regulated interaction with PAR-3 and PAR-6 are required for consistent F-actin oscillations, MTOC perinuclear mobility, aPKC repositioning and wound-directed migration upstream of Rho kinase (also known as ROCK or ROK) activation. We propose that the PAR complex coordinately controls cytoskeletal changes affecting both the generation of traction force and the directionality of leukocyte migration to sites of injury.
Collapse
Affiliation(s)
- Carolina Lage Crespo
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Vernieri
- IFOM Foundation, Institute FIRC of Molecular Oncology, 20139 Milan, Italy
| | - Philipp J Keller
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, 20147 VI, USA
| | - Massimiliano Garrè
- IFOM Foundation, Institute FIRC of Molecular Oncology, 20139 Milan, Italy
| | - Jeffrey R Bender
- Department of Medicine, Raymond and Beverly Sackler Foundation Cardiovascular Laboratory, Yale University, New Haven, 06511 CT, USA
| | - Joachim Wittbrodt
- Center for Organismal Studies Heidelberg, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ruggero Pardi
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy
| |
Collapse
|
103
|
Wang F, Lin SL. Knockdown of kinesin KIF11 abrogates directed migration in response to epidermal growth factor-mediated chemotaxis. Biochem Biophys Res Commun 2014; 452:642-8. [PMID: 25193695 DOI: 10.1016/j.bbrc.2014.08.136] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 02/04/2023]
Abstract
Establishment of microtubule polarity is critical for directional cell migration involved in morphogenesis, differentiation, cell division, and metastasis. Current models, involving iterative microtubule capture and inactivation of microtubule depolymerizing mechanisms at the leading edge, cannot account for the biased migration exhibited by cells in culture in the absence of directional cues, suggesting central mechanisms governing microtubule polarity remain unknown. We engineered two human MDA-MB-231/IMP1 breast carcinoma cell lines, denoted kdKIF11-1 and kdKIF11-2, in which the kinesin KIF11 (also known as Eg5) was stably knocked down by two different shRNAs. Western blot analysis showed knockdown by each shRNA decreased KIF11 expression by 58% and 79% for kdKIF11-1 and kdKIF11-2, respectively, whereas Rac1 expression was unaffected. All cell lines retained a well-defined microtubule structure. Compared to cells infected with the control viral vector, both KIF11 knockdown cell lines displayed a 14-45% increase in cell motility in a scratch wound healing assay. In contrast, KIF11 knockdown decreased invasion by 70%, compared to the control, as measured by invasion through Matrigel-coated transwells. To determine whether the reduction in invasion was due to reduced chemotaxis, we substituted collagen for Matrigel in the transwell assay and similarly observed a 44-54% reduction in migration, using EGF as the chemoattractant. However, when including EGF in both the upper and lower chambers of the transwell to stimulate migration but eliminate chemotaxis, transwell migration decreased for the control cell line only, indicating that KIF11 knockdown did not impair migration, but severely impaired chemotaxis. We conclude KIF11 is a key downstream molecule that responds to directional cues in chemotaxis to govern the direction of migration.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pathophysiology, Key Immunology Laboratory of Guangdong Province, Shantou University College of Medicine, 22 Xinling Road, Shantou, Guangdong 515041, China; Center for Neuroscience, Shantou University College of Medicine, 22 Xinling Road, Shantou, Guangdong 515041, China
| | - Stanley Li Lin
- Department of Pathophysiology, Key Immunology Laboratory of Guangdong Province, Shantou University College of Medicine, 22 Xinling Road, Shantou, Guangdong 515041, China; Center for Neuroscience, Shantou University College of Medicine, 22 Xinling Road, Shantou, Guangdong 515041, China.
| |
Collapse
|
104
|
Prager-Khoutorsky M, Khoutorsky A, Bourque CW. Unique interweaved microtubule scaffold mediates osmosensory transduction via physical interaction with TRPV1. Neuron 2014; 83:866-78. [PMID: 25123313 DOI: 10.1016/j.neuron.2014.07.023] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2014] [Indexed: 01/19/2023]
Abstract
The electrical activity of mammalian osmosensory neurons (ONs) is increased by plasma hypertonicity to command thirst, antidiuretic hormone release, and increased sympathetic tone during dehydration. Osmosensory transduction is a mechanical process whereby decreases in cell volume cause the activation of transient receptor potential vanilloid type-1 (TRPV1) channels to induce depolarization and increase spiking activity in ONs. However, it is not known how cell shrinking is mechanically coupled to channel activation. Using superresolution imaging, we found that ONs are endowed with a uniquely interweaved scaffold of microtubules throughout their somata. Microtubules physically interact with the C terminus of TRPV1 at the cell surface and provide a pushing force that drives channels activation during shrinking. Moreover, we found that changes in the density of these interactions can bidirectionally modulate osmosensory gain. Microtubules are thus an essential component of the vital neuronal mechanotransduction apparatus that allows the brain to monitor and correct body hydration.
Collapse
Affiliation(s)
- Masha Prager-Khoutorsky
- Center for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC H3G 1A4, Canada
| | - Arkady Khoutorsky
- Department of Biochemistry and Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Charles W Bourque
- Center for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC H3G 1A4, Canada.
| |
Collapse
|
105
|
Werner ME, Mitchell JW, Putzbach W, Bacon E, Kim SK, Mitchell BJ. Radial intercalation is regulated by the Par complex and the microtubule-stabilizing protein CLAMP/Spef1. J Cell Biol 2014; 206:367-76. [PMID: 25070955 PMCID: PMC4121976 DOI: 10.1083/jcb.201312045] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/25/2014] [Indexed: 01/28/2023] Open
Abstract
The directed movement of cells is critical for numerous developmental and disease processes. A developmentally reiterated form of migration is radial intercalation; the process by which cells move in a direction orthogonal to the plane of the tissue from an inner layer to an outer layer. We use the radial intercalation of cells into the skin of Xenopus laevis embryos as a model to study directed cell migration within an epithelial tissue. We identify a novel function for both the microtubule-binding protein CLAMP and members of the microtubule-regulating Par complex during intercalation. Specifically, we show that Par3 and aPKC promote the apical positioning of centrioles, whereas CLAMP stabilizes microtubules along the axis of migration. We propose a model in which the Par complex defines the orientation of apical migration during intercalation and in which subcellular localization of CLAMP promotes the establishment of an axis of microtubule stability required for the active migration of cells into the outer epithelium.
Collapse
Affiliation(s)
- Michael E Werner
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Jennifer W Mitchell
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - William Putzbach
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Elizabeth Bacon
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Sun K Kim
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Brian J Mitchell
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
106
|
Besnoitia besnoiti and Toxoplasma gondii: two apicomplexan strategies to manipulate the host cell centrosome and Golgi apparatus. Parasitology 2014; 141:1436-54. [DOI: 10.1017/s0031182014000493] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SUMMARYBesnoitia besnoiti and Toxoplasma gondii are two closely related parasites that interact with the host cell microtubule cytoskeleton during host cell invasion. Here we studied the relationship between the ability of these parasites to invade and to recruit the host cell centrosome and the Golgi apparatus. We observed that T. gondii recruits the host cell centrosome towards the parasitophorous vacuole (PV), whereas B. besnoiti does not. Notably, both parasites recruit the host Golgi apparatus to the PV but its organization is affected in different ways. We also investigated the impact of depleting and over-expressing the host centrosomal protein TBCCD1, involved in centrosome positioning and Golgi apparatus integrity, on the ability of these parasites to invade and replicate. Toxoplasma gondii replication rate decreases in cells over-expressing TBCCD1 but not in TBCCD1-depleted cells; while for B. besnoiti no differences were found. However, B. besnoiti promotes a reorganization of the Golgi ribbon previously fragmented by TBCCD1 depletion. These results suggest that successful establishment of PVs in the host cell requires modulation of the Golgi apparatus which probably involves modifications in microtubule cytoskeleton organization and dynamics. These differences in how T. gondii and B. besnoiti interact with their host cells may indicate different evolutionary paths.
Collapse
|
107
|
Luxton GWG, Starr DA. KASHing up with the nucleus: novel functional roles of KASH proteins at the cytoplasmic surface of the nucleus. Curr Opin Cell Biol 2014; 28:69-75. [PMID: 24704701 DOI: 10.1016/j.ceb.2014.03.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/05/2014] [Accepted: 03/08/2014] [Indexed: 10/25/2022]
Abstract
Nuclear-cytoskeletal connections are central to fundamental cellular processes, including nuclear positioning and chromosome movements in meiosis. The cytoskeleton is coupled to the nucleoskeleton through conserved KASH-SUN bridges, or LINC complexes, that span the nuclear envelope. KASH proteins localize to the outer nuclear membrane where they connect the nucleus to the cytoskeleton. New findings have expanded the functional diversity of KASH proteins, showing that they interact with microtubule motors, actin, intermediate filaments, a nonconventional myosin, RanGAP, and each other. The role of KASH proteins in cellular mechanics is discussed. Genetic mutations in KASH proteins are associated with autism, hearing loss, cancer, muscular dystrophy and other diseases.
Collapse
Affiliation(s)
- G W Gant Luxton
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, United States.
| | - Daniel A Starr
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, United States.
| |
Collapse
|
108
|
Kiss A, Horvath P, Rothballer A, Kutay U, Csucs G. Nuclear motility in glioma cells reveals a cell-line dependent role of various cytoskeletal components. PLoS One 2014; 9:e93431. [PMID: 24691067 PMCID: PMC3972233 DOI: 10.1371/journal.pone.0093431] [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] [Received: 10/15/2013] [Accepted: 03/05/2014] [Indexed: 12/31/2022] Open
Abstract
Nuclear migration is a general term for the movement of the nucleus towards a specific site in the cell. These movements are involved in a number of fundamental biological processes, such as fertilization, cell division, and embryonic development. Despite of its importance, the mechanism of nuclear migration is still poorly understood in mammalian cells. In order to shed light on the mechanical processes underlying nuclear movements, we adapted a micro-patterning based assay. C6 rat and U87 human glioma cells seeded on fibronectin patterns - thereby forced into a bipolar morphology - displayed oscillatory movements of the nucleus or the whole cell, respectively. We found that both the actomyosin system and microtubules are involved in the nuclear/cellular movements of both cell lines, but their contributions are cell-/migration-type specific. Dynein activity was necessary for nuclear migration of C6 cells but active myosin-II was dispensable. On the other hand, coupled nuclear and cellular movements of U87 cells were driven by actomyosin contraction. We explain these cell-line dependent effects by the intrinsic differences in the overall mechanical tension due to the various cytoskeletal elements inside the cell. Our observations showed that the movements of the nucleus and the centrosome are strongly correlated and display large variation, indicating a tight but flexible coupling between them. The data also indicate that the forces responsible for nuclear movements are not acting directly via the centrosome. Based on our observations, we propose a new model for nuclear oscillations in C6 cells in which dynein and microtubule dynamics are the main drivers of nuclear movements. This mechanism is similar to the meiotic nuclear oscillations of Schizosaccharomyces pombe and may be evolutionary conserved.
Collapse
Affiliation(s)
- Alexa Kiss
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| | - Peter Horvath
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| | | | - Ulrike Kutay
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| | - Gabor Csucs
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; Light Microscopy and Screening Centre, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
109
|
Singh AV, Mehta KK, Worley K, Dordick JS, Kane RS, Wan LQ. Carbon nanotube-induced loss of multicellular chirality on micropatterned substrate is mediated by oxidative stress. ACS NANO 2014; 8:2196-2205. [PMID: 24559311 DOI: 10.1021/nn405253d] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Carbon nanotubes (CNTs) are receiving much attention in medicine, electronics, consumer products, and next-generation nanocomposites because of their unique nanoscale properties. However, little is known about the toxicity and oxidative stress related anomalies of CNTs on complex multicellular behavior. This includes cell chirality, a newly discovered cellular property important for embryonic morphogenesis and demonstrated by directional migration and biased alignment on micropatterned surfaces. In this study, we report the influence of single-walled carbon nanotubes (SWCNTs) on multicellular chirality. The incubation of human umbilical vein endothelial cells (hUVECs) and mouse myoblasts (C2C12) with CNTs at different doses and time points stimulates reactive oxygen species (ROS) production and intra- and extracellular oxidative stress (OS). The OS-mediated noxious microenvironment influences vital subcellular organelles (e.g., mitochondria and centrosomes), cytoskeletal elements (microtubules), and vinculin rich focal adhesions. The disorientated nuclear-centrosome (NC) axis and centriole disintegration lead to a decreased migration rate and loss of directional alignment on micropatterned surfaces. These findings suggest that CNT-mediated OS leads to loss of multicellular chirality. Furthermore, the in vitro microscale system presented here to measure cell chirality can be extended as a prototype for testing toxicity of other nanomaterials.
Collapse
Affiliation(s)
- Ajay V Singh
- Department of Biomedical Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | | | | | | | | | | |
Collapse
|
110
|
Morris EJ, Nader GPF, Ramalingam N, Bartolini F, Gundersen GG. Kif4 interacts with EB1 and stabilizes microtubules downstream of Rho-mDia in migrating fibroblasts. PLoS One 2014; 9:e91568. [PMID: 24658398 PMCID: PMC3962350 DOI: 10.1371/journal.pone.0091568] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 02/12/2014] [Indexed: 01/03/2023] Open
Abstract
Selectively stabilized microtubules (MTs) form in the lamella of fibroblasts and contribute to cell migration. A Rho-mDia-EB1 pathway regulates the formation of stable MTs, yet how selective stabilization of MTs is achieved is unknown. Kinesin activity has been implicated in selective MT stabilization and a number of kinesins regulate MT dynamics both in vitro and in cells. Here, we show that the mammalian homolog of Xenopus XKLP1, Kif4, is both necessary and sufficient for the induction of selective MT stabilization in fibroblasts. Kif4 localized to the ends of stable MTs and participated in the Rho-mDia-EB1 MT stabilization pathway since Kif4 depletion blocked mDia- and EB1-induced selective MT stabilization and EB1 was necessary for Kif4 induction of stable MTs. Kif4 and EB1 interacted in cell extracts, and binding studies revealed that the tail domain of Kif4 interacted directly with the N-terminal domain of EB1. Consistent with its role in regulating formation of stable MTs in interphase cells, Kif4 knockdown inhibited migration of cells into wounded monolayers. These data identify Kif4 as a novel factor in the Rho-mDia-EB1 MT stabilization pathway and cell migration.
Collapse
Affiliation(s)
- Edward J. Morris
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| | - Guilherme P. F. Nader
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| | - Nagendran Ramalingam
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| | - Gregg G. Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
111
|
Sharma M, Johnson M, Brocardo M, Jamieson C, Henderson BR. Wnt signaling proteins associate with the nuclear pore complex: implications for cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 773:353-72. [PMID: 24563356 DOI: 10.1007/978-1-4899-8032-8_16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Several components of the Wnt signaling pathway have in recent years been linked to the nuclear pore complex. β-catenin, the primary transducer of Wnt signals from the plasma membrane to the nucleus, has been shown to transiently associate with different FG-repeat containing nucleoporins (Nups) and to translocate bidirectionally through pores of the nuclear envelope in a manner independent of classical transport receptors and the Ran GTPase. Two key regulators of β-catenin, IQGAP1 and APC, have also been reported to bind specific Nups or to locate at the nuclear pore complex. The interaction between these Wnt signaling proteins and different Nups may have functional implications beyond nuclear transport in cellular processes that include mitotic regulation, centrosome positioning and cell migration, nuclear envelope assembly/disassembly, and the DNA replication checkpoint. The broad implications of interactions between Wnt signaling proteins and Nups will be discussed in the context of cancer.
Collapse
Affiliation(s)
- Manisha Sharma
- Westmead Institute for Cancer Research, Westmead Millennium Institute at Westmead Hospital, The University of Sydney, Darcy Road, 412, Westmead, NSW, 2145, Australia,
| | | | | | | | | |
Collapse
|
112
|
Ogden A, Rida PCG, Aneja R. Heading off with the herd: how cancer cells might maneuver supernumerary centrosomes for directional migration. Cancer Metastasis Rev 2013; 32:269-87. [PMID: 23114845 DOI: 10.1007/s10555-012-9413-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The complicity of centrosomes in carcinogenesis is unmistakable. Mounting evidence clearly implicates a robust correlation between centrosome amplification (CA) and malignant transformation in diverse tissue types. Furthermore, CA has been suggested as a marker of cancer aggressiveness, in particular the invasive phenotype, in breast and prostate cancers. One means by which CA promotes malignancy is through induction of transient spindle multipolarity during mitosis, which predisposes the cell to karyotypic changes arising from low-grade chromosome mis-segregation. It is well recognized that during cell migration in interphase, centrosome-mediated nucleation of a radial microtubule array is crucial for establishing a polarized Golgi apparatus, without which directionality is precluded. The question of how cancer cells maneuver their supernumerary centrosomes to achieve directionality during cell migration is virtually uncharted territory. Given that CA is a hallmark of cancers and has been correlated with cancer aggressiveness, malignant cells are presumably competent in managing their centrosome surfeit during directional migration, although the cellular logistics of this process remain unexplored. Another key angle worth pondering is whether an overabundance of centrosomes confers some advantage on cancer cells in terms of their migratory and invasive capabilities. Recent studies have uncovered a remarkable strategy that cancer cells employ to deal with the problem of excess centrosomes and ensure bipolar mitoses, viz., centrosome clustering. This review aims to change the narrative by exploring how an increased centrosome complement may, via aneuploidy-independent modulation of the microtubule cytoskeleton, enhance directional migration and invasion of malignant cells. We postulate that CA imbues cancer cells with cytoskeletal advantages that enhance cell polarization, Golgi-dependent vesicular trafficking, stromal invasion, and other aspects of metastatic progression. We also propose that centrosome declustering may represent a novel, cancer cell-specific antimetastatic strategy, as cancer cells may rely on centrosome clustering during migration as they do in mitosis. Elucidation of these details offers an exciting avenue for future research, as does investigating how CA may promote metastasis through enhanced directional migration.
Collapse
Affiliation(s)
- Angela Ogden
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | | |
Collapse
|
113
|
Soares H, Marinho HS, Real C, Antunes F. Cellular polarity in aging: role of redox regulation and nutrition. GENES AND NUTRITION 2013; 9:371. [PMID: 24306961 DOI: 10.1007/s12263-013-0371-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 11/19/2013] [Indexed: 02/06/2023]
Abstract
Cellular polarity concerns the spatial asymmetric organization of cellular components and structures. Such organization is important not only for biological behavior at the individual cell level, but also for the 3D organization of tissues and organs in living organisms. Processes like cell migration and motility, asymmetric inheritance, and spatial organization of daughter cells in tissues are all dependent of cell polarity. Many of these processes are compromised during aging and cellular senescence. For example, permeability epithelium barriers are leakier during aging; elderly people have impaired vascular function and increased frequency of cancer, and asymmetrical inheritance is compromised in senescent cells, including stem cells. Here, we review the cellular regulation of polarity, as well as the signaling mechanisms and respective redox regulation of the pathways involved in defining cellular polarity. Emphasis will be put on the role of cytoskeleton and the AMP-activated protein kinase pathway. We also discuss how nutrients can affect polarity-dependent processes, both by direct exposure of the gastrointestinal epithelium to nutrients and by indirect effects elicited by the metabolism of nutrients, such as activation of antioxidant response and phase-II detoxification enzymes through the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2). In summary, cellular polarity emerges as a key process whose redox deregulation is hypothesized to have a central role in aging and cellular senescence.
Collapse
Affiliation(s)
- Helena Soares
- Departamento de Química e Bioquímica, Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | | | | | | |
Collapse
|
114
|
Desai SP, Bhatia SN, Toner M, Irimia D. Mitochondrial localization and the persistent migration of epithelial cancer cells. Biophys J 2013; 104:2077-88. [PMID: 23663851 DOI: 10.1016/j.bpj.2013.03.025] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 02/18/2013] [Accepted: 03/07/2013] [Indexed: 01/02/2023] Open
Abstract
During cancer cell invasion, faster moving cancer cells play a dominant role by invading further and metastasizing earlier. Despite the importance of these outlier cells, the source of heterogeneity in their migratory behavior remains poorly understood. Here, we show that anterior localization of mitochondria, in between the nucleus and the leading edge of migrating epithelial cancer cells, correlates with faster migration velocities and increased directional persistence. The asymmetry of mitochondrial localization along the axis of migration is absent during spontaneous cell migration on two-dimensional surfaces and only occurs in the presence of chemical attractant cues or in conditions of mechanical confinement. Moreover, perturbing the asymmetric distribution of mitochondria within migrating cells by interfering with mitochondrial fusion (opa-1) or fission (drp-1) proteins, significantly reduces the number of cells with anterior localization of mitochondria and significantly decreases the velocity and directional persistence of the fastest moving cells. We also observed similar changes after perturbing the linkage between mitochondria and microtubules by the knockdown of mitochondrial rhoGTPase-1 (miro-1). Taken together, the changes in migration velocity and directional persistence in cells with anterior-localized mitochondria could account for an order of magnitude differences in invasive abilities between cells from otherwise homogenous cell populations.
Collapse
Affiliation(s)
- Salil P Desai
- Harvard-MIT Division of Health-Science Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
115
|
Vandenberg LN, Lemire JM, Levin M. It's never too early to get it Right: A conserved role for the cytoskeleton in left-right asymmetry. Commun Integr Biol 2013; 6:e27155. [PMID: 24505508 PMCID: PMC3912007 DOI: 10.4161/cib.27155] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 01/08/2023] Open
Abstract
For centuries, scientists and physicians have been captivated by the consistent left-right (LR) asymmetry of the heart, viscera, and brain. A recent study implicated tubulin proteins in establishing laterality in several experimental models, including asymmetric chemosensory receptor expression in C. elegans neurons, polarization of HL-60 human neutrophil-like cells in culture, and asymmetric organ placement in Xenopus. The same mutations that randomized asymmetry in these diverse systems also affect chirality in Arabidopsis, revealing a remarkable conservation of symmetry-breaking mechanisms among kingdoms. In Xenopus, tubulin mutants only affected LR patterning very early, suggesting that this axis is established shortly after fertilization. This addendum summarizes and extends the knowledge of the cytoskeleton's role in the patterning of the LR axis. Results from many species suggest a conserved role for the cytoskeleton as the initiator of asymmetry, and indicate that symmetry is first broken during early embryogenesis by an intracellular process.
Collapse
Affiliation(s)
- Laura N Vandenberg
- Biology Department; Center for Regenerative and Developmental Biology; Tufts University; Medford, MA USA ; Current affiliation: Department of Public Health; Division of Environmental Health Sciences; University of Massachusetts, Amherst; Amherst, MA USA
| | - Joan M Lemire
- Biology Department; Center for Regenerative and Developmental Biology; Tufts University; Medford, MA USA
| | - Michael Levin
- Biology Department; Center for Regenerative and Developmental Biology; Tufts University; Medford, MA USA
| |
Collapse
|
116
|
Chang W, Folker ES, Worman HJ, Gundersen GG. Emerin organizes actin flow for nuclear movement and centrosome orientation in migrating fibroblasts. Mol Biol Cell 2013; 24:3869-80. [PMID: 24152738 PMCID: PMC3861083 DOI: 10.1091/mbc.e13-06-0307] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Emerin, a nuclear membrane protein, and myosin IIB contribute to nuclear movement by regulating the directionality of nuclear movement and dorsal actin cable flow. Emerin interacts with myosin IIB and is required for its perinuclear localization. The results show that the nuclear envelope actively organizes cytoplasmic polarity. In migrating fibroblasts, rearward movement of the nucleus orients the centrosome toward the leading edge. Nuclear movement results from coupling rearward-moving, dorsal actin cables to the nucleus by linear arrays of nesprin-2G and SUN2, termed transmembrane actin-associated nuclear (TAN) lines. A-type lamins anchor TAN lines, prompting us to test whether emerin, a nuclear membrane protein that interacts with lamins and TAN line proteins, contributes to nuclear movement. In fibroblasts depleted of emerin, nuclei moved nondirectionally or completely failed to move. Consistent with these nuclear movement defects, dorsal actin cable flow was nondirectional in cells lacking emerin. TAN lines formed normally in cells lacking emerin and were coordinated with the erratic nuclear movements, although in 20% of the cases, TAN lines slipped over immobile nuclei. Myosin II drives actin flow, and depletion of myosin IIB, but not myosin IIA, showed similar nondirectional nuclear movement and actin flow as in emerin-depleted cells. Myosin IIB specifically coimmunoprecipitated with emerin, and emerin depletion prevented myosin IIB localization near nuclei. These results show that emerin functions with myosin IIB to polarize actin flow and nuclear movement in fibroblasts, suggesting a novel function for the nuclear envelope in organizing directional actin flow and cytoplasmic polarity.
Collapse
Affiliation(s)
- Wakam Chang
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032 Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | | | | | | |
Collapse
|
117
|
Peng H, Ong YM, Shah WA, Holland PC, Carbonetto S. Integrins regulate centrosome integrity and astrocyte polarization following a wound. Dev Neurobiol 2013; 73:333-53. [PMID: 22949126 DOI: 10.1002/dneu.22055] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/14/2012] [Accepted: 08/27/2012] [Indexed: 12/31/2022]
Abstract
In response to a wound, astrocytes in culture extend microtubule-rich processes and polarize, orienting their centrosomes and Golgi apparatus woundside. β1 Integrin null astrocytes fail to extend processes toward the wound, and are disoriented, and often migrate away orthogonal, to the wound. The centrosome is unusually fragmented in β1 integrin null astrocytes. Expression of a β1 integrin cDNA in the null background yields cells with intact centrosomes that polarize and extend processes normally. Fragmented centrosomes rapidly assemble following integrin ligation and cell attachment. However, several experiments indicated that cell adhesion is not necessary. For example, astrocytes in suspension expressing a chimeric β1 subunit that can be activated by an antibody assemble centrosomes suggesting that β1 activation is sufficient to cause centrosome assembly in the absence of cell adhesion. siRNA knockdown of PCM1, a major centrosomal protein, inhibits cell polarization, consistent with the notion that centrosomes are necessary for polarity and that integrins regulate polarity via centrosome integrity. Screening inhibitors of molecules downstream of integrins indicate that neither FAK nor ILK is involved in regulation of centrosome integrity. In contrast, blebbistatin, a specific inhibitor of non-muscle myosin II (NMII), mimics the response of β1 integrin null astrocytes by disrupting centrosome integrity and cell polarization. Blebbistatin also inhibits integrin-mediated centrosome assembly in astrocytes attaching to fibronectin, consistent with the hypothesis that NMII functions downstream of integrins in regulating centrosome integrity.
Collapse
Affiliation(s)
- Huashan Peng
- Centre for Research in Neuroscience, McGill University Health Centre, Montreal, Quebec, H3G 1A4, Canada
| | | | | | | | | |
Collapse
|
118
|
Integrating mitochondrial organization and dynamics with cellular architecture. Curr Opin Cell Biol 2013; 26:34-40. [PMID: 24529244 DOI: 10.1016/j.ceb.2013.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/08/2013] [Accepted: 09/16/2013] [Indexed: 12/22/2022]
Abstract
Mitochondrial organization, dynamics, and interactions with other intracellular structures and organelles are crucial for proper cell physiology. In this review we will discuss recent work on the significance of mitochondrial organization in regulating the size and distribution of mitochondrial DNA nucleoids and emphasize the importance of a new role for actin in regulating mitochondrial dynamics. We will also highlight new and unexpected examples of how mitochondria are integrated with many aspects of cell behavior, including cell migration, cell division, and the proper functioning of specialized cells such as neurons and immune cells. Together, these recent studies demonstrate the importance of mitochondrial organization in generating cellular architecture and vice versa.
Collapse
|
119
|
|
120
|
Chang SS, Guo WH, Kim Y, Wang YL. Guidance of cell migration by substrate dimension. Biophys J 2013; 104:313-21. [PMID: 23442853 DOI: 10.1016/j.bpj.2012.12.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 11/25/2012] [Accepted: 12/03/2012] [Indexed: 11/29/2022] Open
Abstract
There is increasing evidence to suggest that physical parameters, including substrate rigidity, topography, and cell geometry, play an important role in cell migration. As there are significant differences in cell behavior when cultured in 1D, 2D, or 3D environments, we hypothesize that migrating cells are also able to sense the dimension of the environment as a guidance cue. NIH 3T3 fibroblasts were cultured on micropatterned substrates where the path of migration alternates between 1D lines and 2D rectangles. We found that 3T3 cells had a clear preference to stay on 2D rather than 1D substrates. Cells on 2D surfaces generated stronger traction stress than did those on 1D surfaces, but inhibition of myosin II caused cells to lose their sensitivity to substrate dimension, suggesting that myosin-II-dependent traction forces are the determining factor for dimension sensing. Furthermore, oncogene-transformed fibroblasts are defective in mechanosensing while generating similar traction forces on 1D and 2D surfaces. Dimension sensing may be involved in guiding cell migration for both physiological functions and tissue engineering, and for maintaining normal cells in their home tissue.
Collapse
Affiliation(s)
- Stephanie S Chang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
121
|
Davis FM, Parsonage MT, Cabot PJ, Parat MO, Thompson EW, Roberts-Thomson SJ, Monteith GR. Assessment of gene expression of intracellular calcium channels, pumps and exchangers with epidermal growth factor-induced epithelial-mesenchymal transition in a breast cancer cell line. Cancer Cell Int 2013; 13:76. [PMID: 23890218 PMCID: PMC3733826 DOI: 10.1186/1475-2867-13-76] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 07/24/2013] [Indexed: 01/05/2023] Open
Abstract
Background Epithelial-mesenchymal transition (EMT) is a process implicated in cancer metastasis that involves the conversion of epithelial cells to a more mesenchymal and invasive cell phenotype. In breast cancer cells EMT is associated with altered store-operated calcium influx and changes in calcium signalling mediated by activation of cell surface purinergic receptors. In this study, we investigated whether MDA-MB-468 breast cancer cells induced to undergo EMT exhibit changes in mRNA levels of calcium channels, pumps and exchangers located on intracellular calcium storing organelles, including the Golgi, mitochondria and endoplasmic reticulum (ER). Methods Epidermal growth factor (EGF) was used to induce EMT in MDA-MB-468 breast cancer cells. Serum-deprived cells were treated with EGF (50 ng/mL) for 12 h and gene expression was assessed using quantitative RT-PCR. Results and conclusions These data reveal no significant alterations in mRNA levels of the Golgi calcium pump secretory pathway calcium ATPases (SPCA1 and SPCA2), or the mitochondrial calcium uniporter (MCU) or Na+/Ca2+ exchanger (NCLX). However, EGF-induced EMT was associated with significant alterations in mRNA levels of specific ER calcium channels and pumps, including (sarco)-endoplasmic reticulum calcium ATPases (SERCAs), and inositol 1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RYR) calcium channel isoforms. The most prominent change in gene expression between the epithelial and mesenchymal-like states was RYR2, which was enriched 45-fold in EGF-treated MDA-MB-468 cells. These findings indicate that EGF-induced EMT in breast cancer cells may be associated with major alterations in ER calcium homeostasis.
Collapse
Affiliation(s)
- Felicity M Davis
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | | | | | | | |
Collapse
|
122
|
Lively S, Schlichter LC. The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion. J Neuroinflammation 2013; 10:75. [PMID: 23786632 PMCID: PMC3693964 DOI: 10.1186/1742-2094-10-75] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/06/2013] [Indexed: 12/30/2022] Open
Abstract
Background Microglial cells are highly mobile under many circumstances and, after central nervous system (CNS) damage, they must contend with the dense extracellular matrix (ECM) in order to reach their target sites. In response to damage or disease, microglia undergo complex activation processes that can be modulated by environmental cues and culminate in either detrimental or beneficial outcomes. Thus, there is considerable interest in comparing their pro-inflammatory (‘classical’ activation) and resolving ‘alternative’ activation states. Almost nothing is known about how these activation states affect the ability of microglia to migrate and degrade ECM, or the enzymes used for substrate degradation. This is the subject of the present study. Methods Primary cultured rat microglial cells were exposed to lipopolysaccharide (LPS) to evoke classical activation or IL4 to evoke alternative activation. High-resolution microscopy was used to monitor changes in cell morphology and aspects of the cytoskeleton. We quantified migration in a scratch-wound assay and through open filter holes, and invasion through Matrigel™. A panel of inhibitors was used to analyze contributions of different matrix-degrading enzymes to migration and invasion, and quantitative real-time reverse transcriptase PCR (qRT-PCR) was used to assess changes in their expression. Results Vinculin- and F-actin-rich lamellae were prominent in untreated and IL4-treated microglia (but not after LPS). IL4 increased the migratory capacity of microglia but eliminated the preferential anterior nuclear-centrosomal axis polarity and location of the microtubule organizing center (MTOC). Microglia degraded fibronectin, regardless of treatment, but LPS-treated cells were relatively immobile and IL4-treated cells invaded much more effectively through Matrigel™. For invasion, untreated microglia primarily used cysteine proteases, but IL4-treated cells used a wider range of enzymes (cysteine proteases, cathepsin S and K, heparanase, and matrix metalloproteases). Untreated microglia expressed MMP2, MMP12, heparanase, and four cathepsins (B, K, L1, and S). Each activation stimulus upregulated a different subset of enzymes. IL4 increased MMP2 and cathepsins S and K; whereas LPS increased MMP9, MMP12, MMP14 (MT1-MMP), heparanase, and cathepsin L1. Conclusions Microglial cells migrate during CNS development and after CNS damage or disease. Thus, there are broad implications of the finding that classically and alternatively activated microglia differ in morphology, cytoskeleton, migratory and invasive capacity, and in the usage of ECM-degrading enzymes.
Collapse
Affiliation(s)
- Starlee Lively
- Toronto Western Research Institute, Room MC9-417, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
| | | |
Collapse
|
123
|
Rothballer A, Kutay U. The diverse functional LINCs of the nuclear envelope to the cytoskeleton and chromatin. Chromosoma 2013; 122:415-29. [PMID: 23736899 PMCID: PMC3777164 DOI: 10.1007/s00412-013-0417-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/30/2022]
Abstract
The nuclear envelope (NE) is connected to the different types of cytoskeletal elements by linker of nucleoskeleton and cytoskeleton (LINC) complexes. LINC complexes exist from yeast to humans, and have preserved their general architecture throughout evolution. They are composed of SUN and KASH domain proteins of the inner and the outer nuclear membrane, respectively. These SUN–KASH bridges are used for the transmission of forces across the NE and support diverse biological processes. Here, we review the function of SUN and KASH domain proteins in various unicellular and multicellular species. Specifically, we discuss their influence on nuclear morphology and cytoskeletal organization. Further, emphasis is given on the role of LINC complexes in nuclear anchorage and migration as well as in genome organization.
Collapse
Affiliation(s)
- Andrea Rothballer
- Department of Biology, Institute of Biochemistry, ETH Zurich, Schafmattstrasse 18, 8093 Zurich, Switzerland
| | - Ulrike Kutay
- Department of Biology, Institute of Biochemistry, ETH Zurich, Schafmattstrasse 18, 8093 Zurich, Switzerland
| |
Collapse
|
124
|
Abstract
The nucleus is the largest organelle and is commonly depicted in the center of the cell. Yet during cell division, migration, and differentiation, it frequently moves to an asymmetric position aligned with cell function. We consider the toolbox of proteins that move and anchor the nucleus within the cell and how forces generated by the cytoskeleton are coupled to the nucleus to move it. The significance of proper nuclear positioning is underscored by numerous diseases resulting from genetic alterations in the toolbox proteins. Finally, we discuss how nuclear position may influence cellular organization and signaling pathways.
Collapse
Affiliation(s)
- Gregg G Gundersen
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | |
Collapse
|
125
|
Structural insights into LINC complexes. Curr Opin Struct Biol 2013; 23:285-91. [PMID: 23597672 DOI: 10.1016/j.sbi.2013.03.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 02/21/2013] [Accepted: 03/23/2013] [Indexed: 11/21/2022]
Abstract
Communication between nucleus and cytoplasm extends past molecular exchange and critically includes mechanical wiring. Cytoskeleton and nucleoskeleton are connected via molecular tethers that span the nuclear envelope. Sad1, UNC84 (SUN)-domain proteins spanning the inner nuclear membrane and Klarsicht, ANC-1 and SYNE/Nesprin-1 and -2 Homology (KASH)-peptide bearing proteins residing in the outer nuclear membrane directly bind and constitute the core of the LInkers of Nucleoskeleton and Cytoskeleton (LINC) complex. These connections appear critical for a growing number of biological processes and aberrations are implicated in a host of diverse diseases, including muscular dystrophies, cardiomyopathies, and premature aging. We discuss recent developments in this vibrant research area, particularly in context of first structural insights into LINC complexes reported in the past year.
Collapse
|
126
|
Maier B, Kirsch M, Anderhub S, Zentgraf H, Krämer A. The novel actin/focal adhesion-associated protein MISP is involved in mitotic spindle positioning in human cells. Cell Cycle 2013; 12:1457-71. [PMID: 23574715 PMCID: PMC3674073 DOI: 10.4161/cc.24602] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accurate mitotic spindle positioning is essential for the regulation of cell fate choices, cell size and cell position within tissues. The most prominent model of spindle positioning involves a cortical pulling mechanism, where the minus end-directed microtubule motor protein dynein is attached to the cell cortex and exerts pulling forces on the plus ends of astral microtubules that reach the cortex. In nonpolarized cultured cells integrin-dependent, retraction fiber-mediated cell adhesion is involved in spindle orientation. Proteins serving as intermediaries between cortical actin or retraction fibers and astral microtubules remain largely unknown. In a recent genome-wide RNAi screen we identified a previously uncharacterized protein, MISP (C19ORF21) as being involved in centrosome clustering, a process leading to the clustering of supernumerary centrosomes in cancer cells into a bipolar mitotic spindle array by microtubule tension. Here, we show that MISP is associated with the actin cytoskeleton and focal adhesions and is expressed only in adherent cell types. During mitosis MISP is phosphorylated by Cdk1 and localizes to retraction fibers. MISP interacts with the +TIP EB1 and p150glued, a subunit of the dynein/dynactin complex. Depletion of MISP causes mitotic arrest with reduced tension across sister kinetochores, chromosome misalignment and spindle multipolarity in cancer cells with supernumerary centrosomes. Analysis of spindle orientation revealed that MISP depletion causes randomization of mitotic spindle positioning relative to cell axes and cell center. Together, we propose that MISP links microtubules to the actin cytoskeleton and focal adhesions in order to properly position the mitotic spindle.
Collapse
Affiliation(s)
- Bettina Maier
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
127
|
Chan KT, Jones SW, Brighton HE, Bo T, Cochran SD, Sharpless NE, Bear JE. Intravital imaging of a spheroid-based orthotopic model of melanoma in the mouse ear skin. INTRAVITAL 2013; 2. [PMID: 28748125 DOI: 10.4161/intv.25805] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Multiphoton microscopy is a powerful tool that enables the visualization of fluorescently tagged tumor cells and their stromal interactions within tissues in vivo. We have developed an orthotopic model of implanting multicellular melanoma tumor spheroids into the dermis of the mouse ear skin without the requirement for invasive surgery. Here, we demonstrate the utility of this approach to observe the primary tumor, single cell actin dynamics, and tumor-associated vasculature. These methods can be broadly applied to investigate an array of biological questions regarding tumor cell behavior in vivo.
Collapse
Affiliation(s)
- Keefe T Chan
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Stephen W Jones
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Hailey E Brighton
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Tao Bo
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - Shelly D Cochran
- Department of Biomedical Engineering; North Carolina State University; Raleigh, NC USA
| | - Norman E Sharpless
- Departments of Genetics and Medicine; Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| | - James E Bear
- Department of Cell Biology and Physiology; Howard Hughes Medical Institute, Lineberger Comprehensive Cancer Center; University of North Carolina-Chapel Hill; Chapel Hill, NC USA
| |
Collapse
|
128
|
Maninová M, Klímová Z, Parsons JT, Weber MJ, Iwanicki MP, Vomastek T. The reorientation of cell nucleus promotes the establishment of front-rear polarity in migrating fibroblasts. J Mol Biol 2013; 425:2039-2055. [PMID: 23524135 DOI: 10.1016/j.jmb.2013.02.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 02/10/2013] [Accepted: 02/19/2013] [Indexed: 11/27/2022]
Abstract
The establishment of cell polarity is an essential step in the process of cell migration. This process requires precise spatiotemporal coordination of signaling pathways that in most cells create the typical asymmetrical profile of a polarized cell with nucleus located at the cell rear and the microtubule organizing center (MTOC) positioned between the nucleus and the leading edge. During cell polarization, nucleus rearward positioning promotes correct microtubule organizing center localization and thus the establishment of front-rear polarity and directional migration. We found that cell polarization and directional migration require also the reorientation of the nucleus. Nuclear reorientation is manifested as temporally restricted nuclear rotation that aligns the nuclear axis with the axis of cell migration. We also found that nuclear reorientation requires physical connection between the nucleus and cytoskeleton mediated by the LINC (linker of nucleoskeleton and cytoskeleton) complex. Nuclear reorientation is controlled by coordinated activity of lysophosphatidic acid (LPA)-mediated activation of GTPase Rho and the activation of integrin, FAK (focal adhesion kinase), Src, and p190RhoGAP signaling pathway. Integrin signaling is spatially induced at the leading edge as FAK and p190RhoGAP are predominantly activated or localized at this location. We suggest that integrin activation within lamellipodia defines cell front, and subsequent FAK, Src, and p190RhoGAP signaling represents the polarity signal that induces reorientation of the nucleus and thus promotes the establishment of front-rear polarity.
Collapse
Affiliation(s)
- Miloslava Maninová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Zuzana Klímová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Michael J Weber
- University of Virginia Health System, Charlottesville, VA, USA
| | | | - Tomáš Vomastek
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
129
|
Burakov AV, Nadezhdina ES. Association of nucleus and centrosome: magnet or velcro? Cell Biol Int 2013; 37:95-104. [DOI: 10.1002/cbin.10016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/12/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Anton V. Burakov
- A.N.Belozersky Institute of Physico-Chemical Biology of Lomonosov Moscow State University; Vorobjevy Gory, 1/40, Moscow 119992 Russia
| | - Elena S. Nadezhdina
- A.N.Belozersky Institute of Physico-Chemical Biology of Lomonosov Moscow State University; Vorobjevy Gory, 1/40, Moscow 119992 Russia
- Institute of Protein Research of Russian Academy of Science; Vavilova ul., 34, Moscow 119333 Russia
| |
Collapse
|
130
|
Maninová M, Iwanicki MP, Vomastek T. Emerging role for nuclear rotation and orientation in cell migration. Cell Adh Migr 2013; 8:42-8. [PMID: 24589621 DOI: 10.4161/cam.27761] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nucleus movement, positioning, and orientation is precisely specified and actively regulated within cells, and it plays a critical role in many cellular and developmental processes. Mutation of proteins that regulate the nucleus anchoring and movement lead to diverse pathologies, laminopathies in particular, suggesting that the nucleus correct positioning and movement is essential for proper cellular function. In motile cells that polarize toward the direction of migration, the nucleus undergoes controlled rotation promoting the alignment of the nucleus with the axis of migration. Such spatial organization of the cell appears to be optimal for the cell migration. Nuclear reorientation requires the cytoskeleton to be anchored to the nuclear envelope, which exerts pulling or pushing torque on the nucleus. Here we discuss the possible molecular mechanisms regulating the nuclear rotation and reorientation and the significance of this type of nuclear movement for cell migration.
Collapse
Affiliation(s)
- Miloslava Maninová
- Cell and Molecular Microbiology Division; Institute of Microbiology; Prague, Czech Republic
| | | | - Tomáš Vomastek
- Cell and Molecular Microbiology Division; Institute of Microbiology; Prague, Czech Republic
| |
Collapse
|
131
|
Rodríguez-Fraticelli AE, Auzan M, Alonso MA, Bornens M, Martín-Belmonte F. Cell confinement controls centrosome positioning and lumen initiation during epithelial morphogenesis. ACTA ACUST UNITED AC 2012; 198:1011-23. [PMID: 22965908 PMCID: PMC3444774 DOI: 10.1083/jcb.201203075] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epithelial organ morphogenesis involves sequential acquisition of apicobasal polarity by epithelial cells and development of a functional lumen. In vivo, cells perceive signals from components of the extracellular matrix (ECM), such as laminin and collagens, as well as sense physical conditions, such as matrix stiffness and cell confinement. Alteration of the mechanical properties of the ECM has been shown to promote cell migration and invasion in cancer cells, but the effects on epithelial morphogenesis have not been characterized. We analyzed the effects of cell confinement on lumen morphogenesis using a novel, micropatterned, three-dimensional (3D) Madin-Darby canine kidney cell culture method. We show that cell confinement, by controlling cell spreading, limits peripheral actin contractility and promotes centrosome positioning and lumen initiation after the first cell division. In addition, peripheral actin contractility is mediated by master kinase Par-4/LKB1 via the RhoA-Rho kinase-myosin II pathway, and inhibition of this pathway restores lumen initiation in minimally confined cells. We conclude that cell confinement controls nuclear-centrosomal orientation and lumen initiation during 3D epithelial morphogenesis.
Collapse
Affiliation(s)
- Alejo E Rodríguez-Fraticelli
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | | | | | |
Collapse
|
132
|
Sugioka K, Sawa H. Formation and functions of asymmetric microtubule organization in polarized cells. Curr Opin Cell Biol 2012; 24:517-25. [DOI: 10.1016/j.ceb.2012.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/23/2012] [Indexed: 01/20/2023]
|
133
|
Johnson MA, Henderson BR. The scaffolding protein IQGAP1 co-localizes with actin at the cytoplasmic face of the nuclear envelope: implications for cytoskeletal regulation. BIOARCHITECTURE 2012; 2:138-42. [PMID: 22964981 PMCID: PMC3675075 DOI: 10.4161/bioa.21182] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IQGAP1 is an important cytoskeletal regulator, known to act at the plasma membrane to bundle and cap actin filaments, and to tether the cortical actin meshwork to microtubules via plus-end binding proteins. Here we describe the novel subcellular localization of IQGAP1 at the cytoplasmic face of the nuclear envelope, where it co-located with F-actin. The IQGAP1 and F-actin staining overlapped that of microtubules at the nuclear envelope, revealing a pattern strikingly similar to that observed at the plasma membrane. In detergent-extracted cells IQGAP1 was retained at cytoskeletal structures at the nuclear envelope. This finding has new implications for involvement of IQGAP1 in cell polarization and migration events and potentially in cell cycle-associated nuclear envelope assembly/disassembly.
Collapse
Affiliation(s)
- Michael A Johnson
- Westmead Institute for Cancer Research; University of Sydney; Westmead Millennium Institute at Westmead Hospital; Westmead, Australia
| | - Beric R Henderson
- Westmead Institute for Cancer Research; University of Sydney; Westmead Millennium Institute at Westmead Hospital; Westmead, Australia
| |
Collapse
|
134
|
Neuronal polarity: demarcation, growth and commitment. Curr Opin Cell Biol 2012; 24:547-53. [PMID: 22726583 DOI: 10.1016/j.ceb.2012.05.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/23/2012] [Indexed: 02/03/2023]
Abstract
In a biological sense, polarity refers to the extremity of the main axis of an organelle, cell, or organism. In neurons, morphological polarity begins with the appearance of the first neurite from the cell body. In multipolar neurons, a second phase of polarization occurs when a single neurite initiates a phase of rapid growth to become the neuron's axon, while the others later differentiate as dendrites. Finally, during a third phase, axons and dendrites develop an elaborate architecture, acquiring special morphological and molecular features that commit them to their final identities. Mechanistically, each phase must be preceded by spatial restriction of growth activity. We will review recent work on the mechanisms underlying the polarized growth of neurons.
Collapse
|
135
|
Kawabata W, Yonezawa S, Hayashi K. Rear-side localization of the centrosome in migrating neuroblastoma Neuro-2a cells and its roles in process elongation. Dev Neurosci 2012; 34:20-9. [PMID: 22677609 DOI: 10.1159/000338599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/21/2012] [Indexed: 11/19/2022] Open
Abstract
Axon elongation is usually performed by the migration of growth cones that leave axons. Axon microtubules are generated by enhanced polymerization of tubulin in the growth cones. Some kinds of neurons like cerebellar granule cells, however, generate axons as a result of migration of the cell body leaving axons at the rear. The mechanism to generate microtubules during such growth cone-independent elongation of axons is not well understood. To establish an experimental model to study this mechanism, we cultured neuroblastoma (Neuro-2a) cells on substrates that facilitate cell migration. When cultured on laminin-treated substrate, cells migrated actively and left processes at the rear. We investigated the role of the centrosome in this process formation. The centrosomes were always located at the base of the processes, i.e., at the rear side of the migrating cell body. Close observation of cytoskeletons revealed microtubules limited around the centrosomes, but concentrated at the periphery of the cells or within the processes. Microtubule regrowth experiments showed the ability of the centrosomes to nucleate microtubules. We thus examined the role of microtubule release from the centrosomes, by knocking down the expression of spastin, a microtubule-severing enzyme. Introducing siRNA for spastin into Neuro-2a cells reduced both the migration speed and the length of the processes. Taken together, Neuro-2a cells on laminin proved useful as a model to study the alternative type of axon elongation in which cell migration leaves axons at the rear. This model provided evidence for the involvement of microtubule release from centrosomes in the mechanisms for this type of process elongation.
Collapse
Affiliation(s)
- Wataru Kawabata
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Tokyo, Japan
| | | | | |
Collapse
|