1
|
Purkerson MM, Amend SR, Pienta KJ. Bystanders or active players: the role of extra centrosomes as signaling hubs. Cancer Metastasis Rev 2024; 44:1. [PMID: 39570514 PMCID: PMC11582193 DOI: 10.1007/s10555-024-10224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Centrosomes serve as microtubule-organizing organelles that function in spindle pole organization, cell cycle progression, and cilia formation. A non-canonical role of centrosomes that has gained traction in recent years is the ability to act as signal transduction centers. Centrosome amplification, which includes numerical and structural aberrations of centrosomes, is a candidate hallmark of cancer. The function of centrosomes as signaling centers in cancer cells with centrosome amplification is poorly understood. Establishing a model of how cancer cells utilize centrosomes as signaling platforms will help elucidate the role of extra centrosomes in cancer cell survival and tumorigenesis. Centrosomes act in a diverse array of cellular processes, including cell migration, cell cycle progression, and proteasomal degradation. Given that cancer cells with amplified centrosomes exhibit an increased number and larger area of these signaling platforms, extra centrosomes may be acting to promote tumor development by enhancing signaling kinetics in pathways that are essential for the formation and growth of cancer. In this review, we identify the processes centrosomes are involved in as signal transduction platforms and highlight ways in which cancer cells with centrosome amplification may be taking advantage of these mechanisms.
Collapse
Affiliation(s)
- Madison M Purkerson
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Sarah R Amend
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Tam R, Harris TJ. Centrosome-organized plasma membrane infoldings linked to growth of a cortical actin domain. J Cell Biol 2024; 223:e202403115. [PMID: 38935075 PMCID: PMC11215285 DOI: 10.1083/jcb.202403115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/27/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Regulated cell shape change requires the induction of cortical cytoskeletal domains. Often, local changes to plasma membrane (PM) topography are involved. Centrosomes organize cortical domains and can affect PM topography by locally pulling the PM inward. Are these centrosome effects coupled? At the syncytial Drosophila embryo cortex, centrosome-induced actin caps grow into dome-like compartments for mitoses. We found the nascent cap to be a collection of PM folds and tubules formed over the astral centrosomal MT array. The localized infoldings require centrosome and dynein activities, and myosin-based surface tension prevents them elsewhere. Centrosome-engaged PM infoldings become specifically enriched with an Arp2/3 induction pathway. Arp2/3 actin network growth between the infoldings counterbalances centrosomal pulling forces and disperses the folds for actin cap expansion. Abnormal domain topography with either centrosome or Arp2/3 disruption correlates with decreased exocytic vesicle association. Together, our data implicate centrosome-organized PM infoldings in coordinating Arp2/3 network growth and exocytosis for cortical domain assembly.
Collapse
Affiliation(s)
- Rebecca Tam
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Tony J.C. Harris
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
3
|
Fujii K, Kondo T, Kimura A. Enucleation of the C. elegans embryo revealed dynein-dependent spacing between microtubule asters. Life Sci Alliance 2024; 7:e202302427. [PMID: 37931957 PMCID: PMC10627822 DOI: 10.26508/lsa.202302427] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023] Open
Abstract
The intracellular positioning of the centrosome, a major microtubule-organizing center, is important for cellular functions. One of the features of centrosome positioning is the spacing between centrosomes; however, the underlying mechanisms are not fully understood. To characterize the spacing activity in Caenorhabditis elegans embryos, a genetic setup was developed to produce enucleated embryos. The centrosome was duplicated multiple times in the enucleated embryo, which enabled us to characterize the chromosome-independent spacing activity between sister and non-sister centrosome pairs. We found that the timely spacing depended on cytoplasmic dynein, and we propose a stoichiometric model of cortical and cytoplasmic pulling forces for the spacing between centrosomes. We also observed dynein-independent but non-muscle myosin II-dependent movement of centrosomes in the later cell cycle phase. The spacing mechanisms revealed in this study are expected to function between centrosomes in general, regardless of the presence of a chromosome/nucleus between them, including centrosome separation and spindle elongation.
Collapse
Affiliation(s)
- Ken Fujii
- Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies) Mishima, Japan
- Cell Architecture Laboratory, National Institute of Genetics, Mishima, Japan
| | - Tomo Kondo
- Cell Architecture Laboratory, National Institute of Genetics, Mishima, Japan
| | - Akatsuki Kimura
- Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies) Mishima, Japan
- Cell Architecture Laboratory, National Institute of Genetics, Mishima, Japan
| |
Collapse
|
4
|
Obino D, Maurin M, Dingli F, Loew D, Lescure A, Terriac E, Goudot C, Malbec O, Lankar D, Yuseff MI, Lennon-Duménil AM, Moreau HD. Medium-throughput image-based phenotypic siRNA screen to unveil the molecular basis of B cell polarization. Sci Data 2023; 10:401. [PMID: 37353541 PMCID: PMC10290135 DOI: 10.1038/s41597-023-02301-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/12/2023] [Indexed: 06/25/2023] Open
Abstract
Cell polarity is an essential and highly conserved process governing cell function. Cell polarization is generally triggered by an external signal that induces the relocation of the centrosome, thus defining the polarity axis of the cell. Here, we took advantage of B cells as a model to study cell polarity and perform a medium-throughput siRNA-based imaging screen to identify new molecular regulators of polarization. We first identified candidates based on a quantitative proteomic analysis of proteins differentially associated with the centrosome of resting non-polarized and stimulated polarized B cells. We then targeted 233 candidates in a siRNA screen and identified hits regulating the polarization of the centrosome and/or lysosomes in B cells upon stimulation. Our dataset of proteomics, images, and polarity indexes provides a valuable source of information for a broad community of scientists interested in the molecular mechanisms regulating cell polarity.
Collapse
Affiliation(s)
- Dorian Obino
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France.
| | - Mathieu Maurin
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, 75005, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, 75005, Paris, France
| | - Aurianne Lescure
- Institut Curie, PSL Research University, Translational Research Department, BioPhenics Platform, PICT-IBISA, Paris, France
| | - Emmanuel Terriac
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Christel Goudot
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France
| | - Odile Malbec
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France
| | - Danielle Lankar
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France
| | - Maria-Isabel Yuseff
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France
- Department of Cellular and Molecular Biology, Faculty of Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Hélène D Moreau
- Institut Curie, PSL Research University, Inserm U932, Immunity and Cancer, 75005, Paris, France.
| |
Collapse
|
5
|
Goo BS, Mun DJ, Kim S, Nhung TTM, Lee SB, Woo Y, Kim SJ, Suh BK, Park SJ, Lee HE, Park K, Jang H, Rah JC, Yoon KJ, Baek ST, Park SY, Park SK. Schizophrenia-associated Mitotic Arrest Deficient-1 (MAD1) regulates the polarity of migrating neurons in the developing neocortex. Mol Psychiatry 2023; 28:856-870. [PMID: 36357673 PMCID: PMC9908555 DOI: 10.1038/s41380-022-01856-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Although large-scale genome-wide association studies (GWAS) have identified an association between MAD1L1 (Mitotic Arrest Deficient-1 Like 1) and the pathology of schizophrenia, the molecular mechanisms underlying this association remain unclear. In the present study, we aimed to address these mechanisms by examining the role of MAD1 (the gene product of MAD1L1) in key neurodevelopmental processes in mice and human organoids. Our findings indicated that MAD1 is highly expressed during active cortical development and that MAD1 deficiency leads to impairments in neuronal migration and neurite outgrowth. We also observed that MAD1 is localized to the Golgi apparatus and regulates vesicular trafficking from the Golgi apparatus to the plasma membrane, which is required for the growth and polarity of migrating neurons. In this process, MAD1 physically interacts and collaborates with the kinesin-like protein KIFC3 (kinesin family member C3) to regulate the morphology of the Golgi apparatus and neuronal polarity, thereby ensuring proper neuronal migration and differentiation. Consequently, our findings indicate that MAD1 is an essential regulator of neuronal development and that alterations in MAD1 may underlie schizophrenia pathobiology.
Collapse
Affiliation(s)
- Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seunghyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Sung Jin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Hee-Eun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Kunyou Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
6
|
Shaping subcellular tubes through vesicle trafficking: Common and distinct pathways. Semin Cell Dev Biol 2023; 133:74-82. [PMID: 35365398 DOI: 10.1016/j.semcdb.2022.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 11/23/2022]
Abstract
Cells with subcellular lumens form some of the most miniature tubes in the tubular organs of animals. These are often crucial components of the system, executing functions at remote body locations. Unlike tubes formed by intercellular or autocellular junctions, the cells with junctionless subcellular lumens face unique challenges in modifying the cell shape and plasma membrane organization to incorporate a membrane-bound tube within, often associated with dramatic cellular growth and extensions. Results in the recent years have shown that membrane dynamics, including both the primary delivery and recycling, is crucial in providing the cell with the flexibility to face these challenges. A significant portion of this information has come from two in vivo invertebrate models; the Drosophila tracheal terminal cells and the C. elegans excretory cell. This review focuses on the data obtained from these systems in the recent past about how trafficking pathways influence subcellular tube and branching morphogenesis. Given that such tubes occur in vertebrate vasculature, these insights are relevant to human health, and we contrast our conclusions with the less understood subcellular tubes of angiogenesis.
Collapse
|
7
|
Ignacio DP, Kravtsova N, Henry J, Palomares RH, Dawes AT. Dynein localization and pronuclear movement in the C. elegans zygote. Cytoskeleton (Hoboken) 2022; 79:133-143. [PMID: 36214774 PMCID: PMC10092226 DOI: 10.1002/cm.21733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 01/30/2023]
Abstract
Centrosomes serve as a site for microtubule nucleation and these microtubules will grow and interact with the motor protein dynein at the cortex. The position of the centrosomes determines where the mitotic spindle will develop across all cell types. Centrosome positioning is achieved through dynein and microtubule-mediated force generation. The mechanism and regulation of force generation during centrosome positioning are not fully understood. Centrosome and pronuclear movement in the first cell cycle of the Caenorhabditis elegans early embryo undergoes both centration and rotation prior to cell division. The proteins LET-99 and GPB-1 have been postulated to have a role in force generation associated with pronuclear centration and rotation dynamics. When the expression of these proteins is perturbed, pronuclear positioning exhibits a movement defect characterized by oscillatory ("wobble") behavior of the pronuclear complex (PNC). To determine if this movement defect is due to an effect on cortical dynein distribution, we utilize RNAi-mediated knockdown of LET-99 and GPB-1 to induce wobble and assay for any effects on GFP-tagged dynein localization in the early C. elegans embryo. To compare and quantify the movement defect produced by the knockdown of LET-99 and GPB-1, we devised a quantification method that measures the strength of wobble ("wobble metric") observed under these experimental conditions. Our quantification of pronuclear complex dynamics and dynein localization shows that loss of LET-99 and GPB-1 induces a similar movement defect which is independent of cortical dynein localization in the early C. elegans embryo.
Collapse
Affiliation(s)
- David P Ignacio
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio, USA
| | - Natalia Kravtsova
- Department of Mathematics, Ohio State University, Columbus, Ohio, USA
| | - John Henry
- Department of Mathematics, Ohio State University, Columbus, Ohio, USA
| | | | - Adriana T Dawes
- Department of Mathematics, Department of Molecular Genetics, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
8
|
Infante E, Etienne-Manneville S. Intermediate filaments: Integration of cell mechanical properties during migration. Front Cell Dev Biol 2022; 10:951816. [PMID: 35990612 PMCID: PMC9389290 DOI: 10.3389/fcell.2022.951816] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022] Open
Abstract
Cell migration is a vital and dynamic process required for the development of multicellular organisms and for immune system responses, tissue renewal and wound healing in adults. It also contributes to a variety of human diseases such as cancers, autoimmune diseases, chronic inflammation and fibrosis. The cytoskeleton, which includes actin microfilaments, microtubules, and intermediate filaments (IFs), is responsible for the maintenance of animal cell shape and structural integrity. Each cytoskeletal network contributes its unique properties to dynamic cell behaviour, such as cell polarization, membrane protrusion, cell adhesion and contraction. Hence, cell migration requires the dynamic orchestration of all cytoskeleton components. Among these, IFs have emerged as a molecular scaffold with unique mechanical features and a key player in the cell resilience to mechanical stresses during migration through complex 3D environment. Moreover, accumulating evidence illustrates the participation of IFs in signalling cascades and cytoskeletal crosstalk. Teaming up with actin and microtubules, IFs contribute to the active generation of forces required for cell adhesion and mesenchymal migration and invasion. Here we summarize and discuss how IFs integrate mechanical properties and signalling functions to control cell migration in a wide spectrum of physiological and pathological situations.
Collapse
Affiliation(s)
- Elvira Infante
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Université Paris-Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Université Paris-Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| |
Collapse
|
9
|
Yamamoto S, Gaillard J, Vianay B, Guerin C, Orhant-Prioux M, Blanchoin L, Théry M. Actin network architecture can ensure robust centering or sensitive decentering of the centrosome. EMBO J 2022; 41:e111631. [PMID: 35916262 PMCID: PMC9574749 DOI: 10.15252/embj.2022111631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 01/17/2023] Open
Abstract
The orientation of cell polarity depends on the position of the centrosome, the main microtubule-organizing center (MTOC). Microtubules (MTs) transmit pushing forces to the MTOC as they grow against the cell periphery. How the actin network regulates these forces remains unclear. Here, in a cell-free assay, we used purified proteins to reconstitute the interaction of a microtubule aster with actin networks of various architectures in cell-sized microwells. In the absence of actin filaments, MTOC positioning was highly sensitive to variations in microtubule length. The presence of a bulk actin network limited microtubule displacement, and MTOCs were held in place. In contrast, the assembly of a branched actin network along the well edges centered the MTOCs by maintaining an isotropic balance of pushing forces. An anisotropic peripheral actin network caused the MTOC to decenter by focusing the pushing forces. Overall, our results show that actin networks can limit the sensitivity of MTOC positioning to microtubule length and enforce robust MTOC centering or decentering depending on the isotropy of its architecture.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Jérémie Gaillard
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Benoit Vianay
- Institut de Recherche Saint Louis, UMRS1160-HIPI, CytoMorpho Lab, University of Paris, CEA, INSERM, Paris, France
| | - Christophe Guerin
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Magali Orhant-Prioux
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France
| | - Laurent Blanchoin
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France.,Institut de Recherche Saint Louis, UMRS1160-HIPI, CytoMorpho Lab, University of Paris, CEA, INSERM, Paris, France
| | - Manuel Théry
- Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, France.,Institut de Recherche Saint Louis, UMRS1160-HIPI, CytoMorpho Lab, University of Paris, CEA, INSERM, Paris, France
| |
Collapse
|
10
|
Ordóñez AJL, Fasiczka R, Fernández B, Naaldijk Y, Fdez E, Ramírez MB, Phan S, Boassa D, Hilfiker S. The LRRK2 signaling network converges on a centriolar phospho-Rab10/RILPL1 complex to cause deficits in centrosome cohesion and cell polarization. Biol Open 2022; 11:275880. [PMID: 35776681 PMCID: PMC9346292 DOI: 10.1242/bio.059468] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
The Parkinson's-disease-associated LRRK2 kinase phosphorylates multiple Rab GTPases including Rab8 and Rab10, which enhances their binding to RILPL1 and RILPL2. The nascent interaction between phospho-Rab10 and RILPL1 blocks ciliogenesis in vitro and in the intact brain, and interferes with the cohesion of duplicated centrosomes in dividing cells. We show here that regulators of the LRRK2 signaling pathway including vps35 and PPM1H converge upon causing centrosomal deficits. The cohesion alterations do not require the presence of other LRRK2 kinase substrates including Rab12, Rab35 and Rab43 or the presence of RILPL2. Rather, they depend on the RILPL1-mediated centrosomal accumulation of phosphorylated Rab10. RILPL1 localizes to the subdistal appendage of the mother centriole, followed by recruitment of the LRRK2-phosphorylated Rab proteins to cause the centrosomal defects. The centrosomal alterations impair cell polarization as monitored by scratch wound assays which is reverted by LRRK2 kinase inhibition. These data reveal a common molecular pathway by which enhanced LRRK2 kinase activity impacts upon centrosome-related events to alter the normal biology of a cell. Summary: The Parkinson's disease LRRK2 signaling pathway converges upon the formation of a complex at the subdistal appendage of the mother centriole which causes centrosomal deficits and impairs appropriate cell polarization.
Collapse
Affiliation(s)
- Antonio Jesús Lara Ordóñez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Rachel Fasiczka
- Department of Anesthesiology and Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Belén Fernández
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Yahaira Naaldijk
- Department of Anesthesiology and Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Elena Fdez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Marian Blanca Ramírez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Sébastien Phan
- Department of Neurosciences and National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, USA
| | - Daniela Boassa
- Department of Neurosciences and National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, USA
| | - Sabine Hilfiker
- Department of Anesthesiology and Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
11
|
Vaidžiulytė K, Macé AS, Battistella A, Beng W, Schauer K, Coppey M. Persistent cell migration emerges from a coupling between protrusion dynamics and polarized trafficking. eLife 2022; 11:69229. [PMID: 35302488 PMCID: PMC8963884 DOI: 10.7554/elife.69229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 02/26/2022] [Indexed: 11/13/2022] Open
Abstract
Migrating cells present a variety of paths, from random to highly directional ones. While random movement can be explained by basal intrinsic activity, persistent movement requires stable polarization. Here, we quantitatively address emergence of persistent migration in (hTERT)–immortalizedRPE1 (retinal pigment epithelial) cells over long timescales. By live cell imaging and dynamic micropatterning, we demonstrate that the Nucleus-Golgi axis aligns with direction of migration leading to efficient cell movement. We show that polarized trafficking is directed toward protrusions with a 20-min delay, and that migration becomes random after disrupting internal cell organization. Eventually, we prove that localized optogenetic Cdc42 activation orients the Nucleus-Golgi axis. Our work suggests that polarized trafficking stabilizes the protrusive activity of the cell, while protrusive activity orients this polarity axis, leading to persistent cell migration. Using a minimal physical model, we show that this feedback is sufficient to recapitulate the quantitative properties of cell migration in the timescale of hours.
Collapse
Affiliation(s)
| | | | | | | | - Kristine Schauer
- Tumor Cell Dynamics Unit, Institut Gustave Roussy, Villejuif, France
| | | |
Collapse
|
12
|
Hornak I, Rieger H. Stochastic model of T Cell repolarization during target elimination (II). Biophys J 2022; 121:1246-1265. [PMID: 35196513 PMCID: PMC9034251 DOI: 10.1016/j.bpj.2022.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/08/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Cytotoxic T lymphocytes (T cells) and natural killer cells form a tight contact, the immunological synapse (IS), with target cells, where they release their lytic granules containing perforin/granzyme and cytokine-containing vesicles. During this process the cell repolarizes and moves the microtubule organizing center (MTOC) toward the IS. In the first part of our work we developed a computational model for the molecular-motor-driven motion of the microtubule cytoskeleton during T cell polarization and analyzed the effects of cortical-sliding and capture-shrinkage mechanisms. Here we use this model to analyze the dynamics of the MTOC repositioning in situations in which 1) the IS is in an arbitrary position with respect to the initial position of the MTOC and 2) the T cell has two IS at two arbitrary positions. In the case of one IS, we found that the initial position determines which mechanism is dominant and that the time of repositioning does not rise monotonously with the MTOC-IS distance. In the case of two IS, we observe several scenarios that have also been reported experimentally: the MTOC alternates stochastically (but with a well-defined average transition time) between the two IS; it wiggles in between the two IS without transiting to one of the two; or it is at some point pulled to one of the two IS and stays there. Our model allows one to predict which scenario emerges in dependency of the mechanisms in action and the number of dyneins present. We report that the presence of capture-shrinkage mechanism in at least one IS is necessary to assure the transitions in every cell configuration. Moreover, the frequency of transitions does not decrease with the distance between the two IS and is the highest when both mechanisms are present in both IS.
Collapse
Affiliation(s)
- Ivan Hornak
- Department of Theoretical Physics, Center for Biophysics, Saarland University, Saarbrücken, Germany.
| | - Heiko Rieger
- Department of Theoretical Physics, Center for Biophysics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
13
|
Mastrogiovanni M, Di Bartolo V, Alcover A. Cell Polarity Regulators, Multifunctional Organizers of Lymphocyte Activation and Function. Biomed J 2021; 45:299-309. [PMID: 34626864 PMCID: PMC9250085 DOI: 10.1016/j.bj.2021.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/01/2021] [Accepted: 10/01/2021] [Indexed: 11/27/2022] Open
Abstract
Cell polarity regulators are ubiquitous, evolutionary conserved multifunctional proteins. They contain a variety of protein–protein interaction domains endowing them the capacity to interact with cytoskeleton structures, membrane components and multiple regulatory proteins. In this way, they act in complexes and are pivotal for cell growth and differentiation, tissue formation, stability and turnover, cell migration, wound healing, and others. Hence some of these proteins are tumor suppressors. These cellular processes rely on the establishment of cell polarity characterized by the asymmetric localization of proteins, RNAs, membrane domains, or organelles that together condition cell shape and function. Whether apparently stable, as in epithelia or neurons, or very dynamic, as in immune cells, cell polarity is an active process. It involves cytoskeleton reorganization and targeted intracellular traffic, and results in cellular events such as protein synthesis, secretion and assembly taking place at defined cell poles. Multiple polarity regulators orchestrate these processes. Immune cells are particularly versatile in rapidly polarizing and assuming different shapes, so to swiftly adopt specialized behaviors and functions. Polarity regulators act in various ways in different immune cell types and at their distinct differentiation states. Here we review how cell polarity regulators control different processes and functions along T lymphocyte physiology, including cell migration through different tissues, immunological synapse formation and effector functions.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris. France
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France
| | - Andrés Alcover
- Lymphocyte Cell Biology Unit, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, Department of Immunology, Institut Pasteur, INSERM-U1224. F-75015 Paris, France.
| |
Collapse
|
14
|
Nechipurenko I, Lavrentyeva S, Sengupta P. GRDN-1/Girdin regulates dendrite morphogenesis and cilium position in two specialized sensory neuron types in C. elegans. Dev Biol 2021; 472:38-51. [PMID: 33460640 DOI: 10.1016/j.ydbio.2020.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
Primary cilia are located at the dendritic tips of sensory neurons and house the molecular machinery necessary for detection and transduction of sensory stimuli. The mechanisms that coordinate dendrite extension with cilium position during sensory neuron development are not well understood. Here, we show that GRDN-1, the Caenorhabditis elegans ortholog of the highly conserved scaffold and signaling protein Girdin/GIV, regulates both cilium position and dendrite extension in the postembryonic AQR and PQR gas-sensing neurons. Mutations in grdn-1 disrupt dendrite outgrowth and mislocalize cilia to the soma or proximal axonal segments in AQR, and to a lesser extent, in PQR. GRDN-1 is localized to the basal body and regulates localization of HMR-1/Cadherin to the distal AQR dendrite. However, knockdown of HMR-1 and/or loss of SAX-7/LICAM, molecules previously implicated in sensory dendrite development in C. elegans, do not alter AQR dendrite morphology or cilium position. We find that GRDN-1 localization in AQR is regulated by UNC-116/Kinesin-1, and that correspondingly, unc-116 mutants exhibit severe AQR dendrite outgrowth and cilium positioning defects. In contrast, GRDN-1 and cilium localization in PQR is modulated by LIN-44/Wnt signaling. Together, these findings identify upstream regulators of GRDN-1, and describe new cell-specific roles for this multifunctional protein in sensory neuron development.
Collapse
Affiliation(s)
- Inna Nechipurenko
- Department of Biology, Brandeis University, Waltham, MA, USA; Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA.
| | | | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
15
|
Becsky D, Szabo K, Gyulai-Nagy S, Gajdos T, Bartos Z, Balind A, Dux L, Horvath P, Erdelyi M, Homolya L, Keller-Pinter A. Syndecan-4 Modulates Cell Polarity and Migration by Influencing Centrosome Positioning and Intracellular Calcium Distribution. Front Cell Dev Biol 2020; 8:575227. [PMID: 33178691 PMCID: PMC7593626 DOI: 10.3389/fcell.2020.575227] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Efficient cell migration requires cellular polarization, which is characterized by the formation of leading and trailing edges, appropriate positioning of the nucleus and reorientation of the Golgi apparatus and centrosomes toward the leading edge. Migration also requires the development of an asymmetrical front-to-rear calcium (Ca2+) gradient to regulate focal adhesion assembly and actomyosin contractility. Here we demonstrate that silencing of syndecan-4, a transmembrane heparan sulfate proteoglycan, interferes with the correct polarization of migrating mammalian myoblasts (i.e., activated satellite stem cells). In particular, syndecan-4 knockdown completely abolished the intracellular Ca2+ gradient, abrogated centrosome reorientation and thus decreased cell motility, demonstrating the role of syndecan-4 in cell polarity. Additionally, syndecan-4 exhibited a polarized distribution during migration. Syndecan-4 knockdown cells exhibited decreases in the total movement distance during directional migration, maximum and vectorial distances from the starting point, as well as average and maximum cell speeds. Super-resolution direct stochastic optical reconstruction microscopy images of syndecan-4 knockdown cells revealed nanoscale changes in the actin cytoskeletal architecture, such as decreases in the numbers of branches and individual branch lengths in the lamellipodia of the migrating cells. Given the crucial importance of myoblast migration during embryonic development and postnatal muscle regeneration, we conclude that our results could facilitate an understanding of these processes and the general role of syndecan-4 during cell migration.
Collapse
Affiliation(s)
- Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Kitti Szabo
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamas Gajdos
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsuzsa Bartos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Arpad Balind
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Peter Horvath
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklos Erdelyi
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Laszlo Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
16
|
Fructuoso M, Legrand M, Mousson A, Steffan T, Vauchelles R, De Mey J, Sick E, Rondé P, Dujardin D. FAK regulates dynein localisation and cell polarity in migrating mouse fibroblasts. Biol Cell 2020; 112:53-72. [PMID: 31859373 DOI: 10.1111/boc.201900041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 01/26/2023]
Abstract
BACKGROUND Fibroblasts executing directional migration position their centrosome, and their Golgi apparatus, in front of the nucleus towards the cell leading edge. Centrosome positioning relative to the nucleus has been associated to mechanical forces exerted on the centrosome by the microtubule-dependent molecular motor cytoplasmic dynein 1, and to nuclear movements such as rearward displacement and rotation events. Dynein has been proposed to regulate the position of the centrosome by exerting pulling forces on microtubules from the cell leading edge, where the motor is enriched during migration. However, the mechanism explaining how dynein acts at the front of the cells has not been elucidated. RESULTS We present here results showing that the protein Focal Adhesion Kinase (FAK) interacts with dynein and regulates the enrichment of the dynein/dynactin complex at focal adhesions at the cell the leading edge of migrating fibroblasts. This suggests that focal adhesions provide anchoring sites for dynein during the polarisation process. In support of this, we present evidence indicating that the interaction between FAK and dynein, which is regulated by the phosphorylation of FAK on its Ser732 residue, is required for proper centrosome positioning. Our results further show that the polarisation of the centrosome can occur independently of nuclear movements. Although FAK regulates both nuclear and centrosome motilities, downregulating the interaction between FAK and dynein affects only the nuclear independent polarisation of the centrosome. CONCLUSIONS Our work highlights the role of FAK as a key player in the regulation of several aspects of cell polarity. We thus propose a model in which the transient localisation of dynein with focal adhesions provides a tuneable mechanism to bias dynein traction forces on microtubules allowing proper centrosome positioning in front of the nucleus. SIGNIFICANCE We unravel here a new role for the cancer therapeutic target FAK in the regulation of cell morphogenesis.
Collapse
Affiliation(s)
- Marta Fructuoso
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France.,ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marlène Legrand
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Antoine Mousson
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Tania Steffan
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Romain Vauchelles
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Jan De Mey
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Emilie Sick
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Philippe Rondé
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| | - Denis Dujardin
- Migration, invasion and microenvironnement, Faculté de Pharmacie, UMR7021 CNRS, LBP, Université de Strasbourg, Illkirch, France
| |
Collapse
|
17
|
Bance B, Seetharaman S, Leduc C, Boëda B, Etienne-Manneville S. Microtubule acetylation but not detyrosination promotes focal adhesion dynamics and astrocyte migration. J Cell Sci 2019; 132:jcs.225805. [PMID: 30858195 DOI: 10.1242/jcs.225805] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/25/2019] [Indexed: 01/12/2023] Open
Abstract
Microtubules play a crucial role in mesenchymal migration by controlling cell polarity and the turnover of cell adhesive structures on the extracellular matrix. The polarized functions of microtubules imply that microtubules are locally regulated. Here, we investigated the regulation and role of two major tubulin post-translational modifications, acetylation and detyrosination, which have been associated with stable microtubules. Using primary astrocytes in a wound healing assay, we show that these tubulin modifications are independently regulated during cell polarization and differently affect cell migration. In contrast to microtubule detyrosination, αTAT1 (ATAT1)-mediated microtubule acetylation increases in the vicinity of focal adhesions and promotes cell migration. We further demonstrate that αTAT1 increases focal adhesion turnover by promoting Rab6-positive vesicle fusion at focal adhesions. Our results highlight the specificity of microtubule post-translational modifications and bring new insight into the regulatory functions of tubulin acetylation.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Bertille Bance
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France.,Sorbonne Université, Collège doctoral, F-75005 Paris, France
| | - Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, F-75006 Paris, France
| | - Cécile Leduc
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France
| | - Batiste Boëda
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France
| |
Collapse
|
18
|
Som S, Chatterjee S, Paul R. Mechanistic three-dimensional model to study centrosome positioning in the interphase cell. Phys Rev E 2019; 99:012409. [PMID: 30780383 DOI: 10.1103/physreve.99.012409] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Indexed: 01/28/2023]
Abstract
During the interphase in mammalian cells, the position of the centrosome is actively maintained at a small but finite distance away from the nucleus. The perinuclear positioning of the centrosome is crucial for cellular trafficking and progression into mitosis. Although the literature suggests that the contributions of the microtubule-associated forces bring the centrosome to the center of the cell, the position of the centrosome was merely investigated in the absence of the nucleus. Upon performing a coarse-grained simulation study with mathematical analysis, we show that the combined effect of the forces due to the cell cortex and the nucleus facilitate the centrosome positioning. Our study also demonstrates that in the absence of nucleus-based forces, the centrosome collapses on the nucleus due to cortical forces. Depending upon the magnitudes of the cortical forces and the nucleus-based forces, the centrosome appears to stay at various distances away from the nucleus. Such null force regions are found to be stable as well as unstable fixed points. This study uncovers a set of redundant schemes that the cell may adopt to produce the required cortical and nucleus-based forces stabilizing the centrosome at a finite distance away from the nucleus.
Collapse
Affiliation(s)
- Subhendu Som
- Indian Association for the Cultivation of Science, Kolkata - 700032, India
| | | | - Raja Paul
- Indian Association for the Cultivation of Science, Kolkata - 700032, India
| |
Collapse
|
19
|
Roman AC, Garrido-Jimenez S, Diaz-Chamorro S, Centeno F, Carvajal-Gonzalez JM. Centriole Positioning: Not Just a Little Dot in the Cell. Results Probl Cell Differ 2019; 67:201-221. [PMID: 31435796 DOI: 10.1007/978-3-030-23173-6_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Organelle positioning as many other morphological parameters in a cell is not random. Centriole positioning as centrosomes or ciliary basal bodies is not an exception to this rule in cell biology. Indeed, centriole positioning is a tightly regulated process that occurs during development, and it is critical for many organs to function properly, not just during development but also in the adulthood. In this book chapter, we overview our knowledge on centriole positioning in different and highly specialized animal cells like photoreceptor or ependymal cells. We will also discuss recent advances in the discovery of molecular pathways involved in this process, mostly related to the cytoskeleton and the cell polarity pathways. And finally, we present quantitative methods that have been used to assess centriole positioning in different cell types although mostly in epithelial cells.
Collapse
Affiliation(s)
- Angel-Carlos Roman
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Sergio Garrido-Jimenez
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Selene Diaz-Chamorro
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Francisco Centeno
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain
| | - Jose Maria Carvajal-Gonzalez
- Facultad de Ciencias, Departamento de Bioquímica, Biología Molecular y Genética, Universidad de Extremadura, Badajoz, Spain.
| |
Collapse
|
20
|
Cao Y, Lei Y, Luo Y, Tan T, Du B, Zheng Y, Sun L, Liang Q. The actomyosin network is influenced by NMHC IIA and regulated by Crp F46, which is involved in controlling cell migration. Exp Cell Res 2018; 373:119-131. [PMID: 30336116 DOI: 10.1016/j.yexcr.2018.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Abstract
When a cell migrates, the centrosome positions between the nucleus and the leading edge of migration via the microtubule system. The protein CrpF46 (centrosome-related protein F46) has a known role during mitosis and centrosome duplication. However, how CrpF46 efficiently regulates centrosome-related cell migration is unclear. Here, we report that knockdown of CrpF46 resulted in the disruption of microtubule arrangement, with impaired centrosomal reorientation, and slowed down cell migration. In cells that express low levels of CrpF46, stress fibers were weakened, which could be rescued by recovering Flag-CrpF46. We also found that CrpF46 interacted with non-muscle myosin high chain IIA (NMHC IIA) and that its three coiled-coil domains are pivotal for its binding to NMHC IIA. Additionally, analyses of phosphorylation of NMHC IIA and RLC (regulatory light chain) demonstrated that CrpF46 was associated with myosin IIA during filament formation. Indirect immunofluorescence images indicated that NM IIA filaments were inhibited when CrpF46 was under-expressed. Thus, CrpF46 regulates cell migration by centrosomal reorientation and altering the function of the actomyosin network by controlling specific phosphorylation of myosin.
Collapse
Affiliation(s)
- Yang Cao
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yan Lei
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yang Luo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Tan Tan
- School of Pharmacology and Biology, University of South China, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, Hengyang 421001, PR China
| | - Baochen Du
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yanbo Zheng
- The Institute of Medical Biotechnology (IMB) of the Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Le Sun
- AbMax Biotechnology Co., Beijing 101111, PR China
| | - Qianjin Liang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China.
| |
Collapse
|
21
|
Chang B, Svoboda KKH, Liu X. Cell polarization: From epithelial cells to odontoblasts. Eur J Cell Biol 2018; 98:1-11. [PMID: 30473389 DOI: 10.1016/j.ejcb.2018.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
Cell polarity identifies the asymmetry of a cell. Various types of cells, including odontoblasts and epithelial cells, polarize to fulfil their destined functions. Odontoblast polarization is a prerequisite and fundamental step for tooth development and tubular dentin formation. Current knowledge of odontoblast polarization, however, is very limited, which greatly impedes the development of novel approaches for regenerative endodontics. Compared to odontoblasts, epithelial cell polarization has been extensively studied over the last several decades. The knowledge obtained from epithelia polarization has been found applicable to other cell types, which is particularly useful considering the remarkable similarities of the morphological and compositional features between polarized odontoblasts and epithelia. In this review, we first discuss the characteristics, the key regulatory factors, and the process of epithelial polarity. Next, we compare the known facts of odontoblast polarization with epithelial cells. Lastly, we clarify knowledge gaps in odontoblast polarization and propose the directions for future research to fill the gaps, leading to the advancement of regenerative endodontics.
Collapse
Affiliation(s)
- Bei Chang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Kathy K H Svoboda
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA.
| |
Collapse
|
22
|
Madero-Pérez J, Fernández B, Lara Ordóñez AJ, Fdez E, Lobbestael E, Baekelandt V, Hilfiker S. RAB7L1-Mediated Relocalization of LRRK2 to the Golgi Complex Causes Centrosomal Deficits via RAB8A. Front Mol Neurosci 2018; 11:417. [PMID: 30483055 PMCID: PMC6243087 DOI: 10.3389/fnmol.2018.00417] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 10/25/2018] [Indexed: 11/30/2022] Open
Abstract
Mutations in the LRRK2 gene cause autosomal-dominant Parkinson’s disease (PD), and both LRRK2 as well as RAB7L1 have been implicated in increased susceptibility to idiopathic PD. RAB7L1 has been shown to increase membrane-association and kinase activity of LRRK2, and both seem to be mechanistically implicated in the same pathway. Another RAB protein, RAB8A, has been identified as a prominent LRRK2 kinase substrate, and our recent work demonstrates that aberrant LRRK2-mediated phosphorylation of RAB8A leads to centrosomal alterations. Here, we show that RAB7L1 recruits LRRK2 to the Golgi complex, which causes accumulation of phosphorylated RAB8A in a pericentrosomal/centrosomal location as well as centrosomal deficits identical to those observed with pathogenic LRRK2. The centrosomal alterations induced by wildtype LRRK2 in the presence of RAB7L1 depend on Golgi integrity. This is in contrast to pathogenic LRRK2 mutants, which cause centrosomal deficits independent of Golgi integrity or largely independent on RAB7L1 expression. Furthermore, centrosomal alterations in the presence of wildtype LRRK2 and RAB7L1 are at least in part mediated by aberrant LRRK2-mediated RAB8A phosphorylation, as abolished by kinase inhibitors and reduced upon knockdown of RAB8A. These results indicate that pathogenic LRRK2, as well as increased levels of RAB7L1, cause centrosomal deficits in a manner dependent on aberrant RAB8A phosphorylation and centrosomal/pericentrosomal accumulation, suggesting that centrosomal cohesion deficits may comprise a useful cellular readout for a broader spectrum of the disease.
Collapse
Affiliation(s)
- Jesús Madero-Pérez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Belén Fernández
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Antonio Jesús Lara Ordóñez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena Fdez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Sabine Hilfiker
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
23
|
Hua K, Ferland RJ. Primary Cilia Reconsidered in the Context of Ciliopathies: Extraciliary and Ciliary Functions of Cilia Proteins Converge on a Polarity theme? Bioessays 2018; 40:e1700132. [PMID: 29882973 PMCID: PMC6239423 DOI: 10.1002/bies.201700132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/09/2018] [Indexed: 12/13/2022]
Abstract
Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.
Collapse
Affiliation(s)
- Kiet Hua
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
| | - Russell J Ferland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA, 12208
- Department of Neurology, Albany Medical College, Albany, New York, USA, 12208
| |
Collapse
|
24
|
Agüera-González S, Burton OT, Vázquez-Chávez E, Cuche C, Herit F, Bouchet J, Lasserre R, Del Río-Iñiguez I, Di Bartolo V, Alcover A. Adenomatous Polyposis Coli Defines Treg Differentiation and Anti-inflammatory Function through Microtubule-Mediated NFAT Localization. Cell Rep 2018; 21:181-194. [PMID: 28978472 DOI: 10.1016/j.celrep.2017.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/04/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022] Open
Abstract
Adenomatous polyposis coli (APC) is a polarity regulator and tumor suppressor associated with familial adenomatous polyposis and colorectal cancer development. Although extensively studied in epithelial transformation, the effect of APC on T lymphocyte activation remains poorly defined. We found that APC ensures T cell receptor-triggered activation through Nuclear Factor of Activated T cells (NFAT), since APC is necessary for NFAT's nuclear localization in a microtubule-dependent fashion and for NFAT-driven transcription leading to cytokine gene expression. Interestingly, NFAT forms clusters juxtaposed with microtubules. Ultimately, mouse Apc deficiency reduces the presence of NFAT in the nucleus of intestinal regulatory T cells (Tregs) and impairs Treg differentiation and the acquisition of a suppressive phenotype, which is characterized by the production of the anti-inflammatory cytokine IL-10. These findings suggest a dual role for APC mutations in colorectal cancer development, where mutations drive the initiation of epithelial neoplasms and also reduce Treg-mediated suppression of the detrimental inflammation that enhances cancer growth.
Collapse
Affiliation(s)
- Sonia Agüera-González
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France.
| | - Oliver T Burton
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Vázquez-Chávez
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Céline Cuche
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Floriane Herit
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Jérôme Bouchet
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Rémi Lasserre
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Iratxe Del Río-Iñiguez
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Vincenzo Di Bartolo
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France
| | - Andrés Alcover
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, 75015 Paris, France; CNRS URA1961, 75015 Paris, France; INSERM U1221, 75015 Paris, France.
| |
Collapse
|
25
|
Martin M, Veloso A, Wu J, Katrukha EA, Akhmanova A. Control of endothelial cell polarity and sprouting angiogenesis by non-centrosomal microtubules. eLife 2018; 7:33864. [PMID: 29547120 PMCID: PMC5898915 DOI: 10.7554/elife.33864] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/13/2018] [Indexed: 12/11/2022] Open
Abstract
Microtubules control different aspects of cell polarization. In cells with a radial microtubule system, a pivotal role in setting up asymmetry is attributed to the relative positioning of the centrosome and the nucleus. Here, we show that centrosome loss had no effect on the ability of endothelial cells to polarize and move in 2D and 3D environments. In contrast, non-centrosomal microtubules stabilized by the microtubule minus-end-binding protein CAMSAP2 were required for directional migration on 2D substrates and for the establishment of polarized cell morphology in soft 3D matrices. CAMSAP2 was also important for persistent endothelial cell sprouting during in vivo zebrafish vessel development. In the absence of CAMSAP2, cell polarization in 3D could be partly rescued by centrosome depletion, indicating that in these conditions the centrosome inhibited cell polarity. We propose that CAMSAP2-protected non-centrosomal microtubules are needed for establishing cell asymmetry by enabling microtubule enrichment in a single-cell protrusion. Networks of blood vessels grow like trees. Sprouts appear on existing vessels, stretching out to form new branches in a process called angiogenesis. The cells responsible are the same cells that line the finished vessels. These “endothelial cells” start the process by reorganizing themselves to face the direction of the new sprout, changing shape to become asymmetrical, and then they begin to migrate. Beneath the surface, a network of protein scaffolding supports each migrating cell. The scaffolding includes tube-like fibers called microtubules that extend towards the cell membrane and organize the inside of the cell. Destroying microtubules damages blood vessel formation, but their exact role remains unclear. A structure called the centrosome can organize microtubules within cells. The centrosome was generally believed to act like a compass, pointing in the direction that the cell will move. Microtubules can anchor to the centrosome, and this structure is thought to play an important role in cell migration. Yet, many microtubules organize without it; these microtubules instead are organized by a compartment of the cell called the Golgi apparatus and are stabilized by a protein named CAMSAP2. Martin et al. now report that removing the cells’ centrosomes did not affect cell migration, but getting rid of CAMSAP2 did. Analysis of cell shape and movement in cells grown in the laboratory and in living animals revealed that cells cannot become asymmetrical, or “polarize”, and migrate without CAMSAP2. In a two-dimensional wound-healing assay, a sheet of cells originally grown from the vessels of a human umbilical cord was scratched, and a microscope was then used to record the cell’s movement as they repaired the injury. Normally, the cells on either side move in a straight line using their microtubules, and though the process was not affected in cells without centrosomes, it was in those without CAMSAP2. Even more striking results were seen in three-dimensional assays. When the same blood vessel cells from human umbilical cords are grown as spheres inside collagen gels, they form sprouts as they would in the body. Without CAMSAP2, the cells could not organize their microtubules and they were unable to elongate in one direction and form stable sprouts. Lastly, depleting CAMSAP2 also prevented the normal formation of blood vessels in zebrafish embryos. Taken together, these findings change our understanding of how microtubules affect cell movement and how important the centrosome is for this process. Further work could have an impact on human health, not least in cancer research. Tumors need a good blood supply to grow, so understanding how to block blood vessel formation could lead to new treatments. Microtubules are already a target for cancer therapy, so future work could help to optimize the use of existing drugs.
Collapse
Affiliation(s)
- Maud Martin
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alexandra Veloso
- Interdisciplinary Cluster for Applied Genoproteomics, University of Liège, Liège, Belgium.,GIGA-Molecular Biology in Diseases, University of Liège, Liège, Belgium
| | - Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
26
|
Madero-Pérez J, Fdez E, Fernández B, Lara Ordóñez AJ, Blanca Ramírez M, Gómez-Suaga P, Waschbüsch D, Lobbestael E, Baekelandt V, Nairn AC, Ruiz-Martínez J, Aiastui A, López de Munain A, Lis P, Comptdaer T, Taymans JM, Chartier-Harlin MC, Beilina A, Gonnelli A, Cookson MR, Greggio E, Hilfiker S. Parkinson disease-associated mutations in LRRK2 cause centrosomal defects via Rab8a phosphorylation. Mol Neurodegener 2018; 13:3. [PMID: 29357897 PMCID: PMC5778812 DOI: 10.1186/s13024-018-0235-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 01/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Mutations in LRRK2 are a common genetic cause of Parkinson's disease (PD). LRRK2 interacts with and phosphorylates a subset of Rab proteins including Rab8a, a protein which has been implicated in various centrosome-related events. However, the cellular consequences of such phosphorylation remain elusive. METHODS Human neuroblastoma SH-SY5Y cells stably expressing wildtype or pathogenic LRRK2 were used to test for polarity defects in the context of centrosomal positioning. Centrosomal cohesion deficits were analyzed from transiently transfected HEK293T cells, as well as from two distinct peripheral cell types derived from LRRK2-PD patients. Kinase assays, coimmunoprecipitation and GTP binding/retention assays were used to address Rab8a phosphorylation by LRRK2 and its effects in vitro. Transient transfections and siRNA experiments were performed to probe for the implication of Rab8a and its phosphorylated form in the centrosomal deficits caused by pathogenic LRRK2. RESULTS Here, we show that pathogenic LRRK2 causes deficits in centrosomal positioning with effects on neurite outgrowth, cell polarization and directed migration. Pathogenic LRRK2 also causes deficits in centrosome cohesion which can be detected in peripheral cells derived from LRRK2-PD patients as compared to healthy controls, and which are reversed upon LRRK2 kinase inhibition. The centrosomal cohesion and polarity deficits can be mimicked when co-expressing wildtype LRRK2 with wildtype but not phospho-deficient Rab8a. The centrosomal defects induced by pathogenic LRRK2 are associated with a kinase activity-dependent increase in the centrosomal localization of phosphorylated Rab8a, and are prominently reduced upon RNAi of Rab8a. CONCLUSIONS Our findings reveal a new function of LRRK2 mediated by Rab8a phosphorylation and related to various centrosomal defects.
Collapse
Affiliation(s)
- Jesús Madero-Pérez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Elena Fdez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Belén Fernández
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Antonio J Lara Ordóñez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Marian Blanca Ramírez
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Patricia Gómez-Suaga
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain
| | - Dieter Waschbüsch
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Münster, Münster, Germany
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, KU Leuven, 3000, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, KU Leuven, 3000, Leuven, Belgium
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | | | - Ana Aiastui
- Cell Culture Platform and Division of Neurosciences, Instituto Biodonostia, San Sebastián, Spain
| | - Adolfo López de Munain
- Division of Neurosciences, Instituto Biodonostia-CIBERNED, San Sebastián, Spain.,Division of Neurosciences, Instituto Biodonostia-CIBERNED, University of the Basque Country UPV-EHU, San Sebastián, Spain
| | - Pawel Lis
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Thomas Comptdaer
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Jean-Marc Taymans
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Marie-Christine Chartier-Harlin
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Alexandria Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Adriano Gonnelli
- Department of Biology, University of Padova, 35131, Padova, Italy
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Elisa Greggio
- Department of Biology, University of Padova, 35131, Padova, Italy
| | - Sabine Hilfiker
- Institute of Parasitology and Biomedicine "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Avda del Conocimiento s/n, 18016, Granada, Spain.
| |
Collapse
|
27
|
Mazel T. Crosstalk of cell polarity signaling pathways. PROTOPLASMA 2017; 254:1241-1258. [PMID: 28293820 DOI: 10.1007/s00709-017-1075-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/02/2017] [Indexed: 06/06/2023]
Abstract
Cell polarity, the asymmetric organization of cellular components along one or multiple axes, is present in most cells. From budding yeast cell polarization induced by pheromone signaling, oocyte polarization at fertilization to polarized epithelia and neuronal cells in multicellular organisms, similar mechanisms are used to determine cell polarity. Crucial role in this process is played by signaling lipid molecules, small Rho family GTPases and Par proteins. All these signaling circuits finally govern the cytoskeleton, which is responsible for oriented cell migration, cell shape changes, and polarized membrane and organelle trafficking. Thus, typically in the process of cell polarization, most cellular constituents become polarized, including plasma membrane lipid composition, ion concentrations, membrane receptors, and proteins in general, mRNA, vesicle trafficking, or intracellular organelles. This review gives a brief overview how these systems talk to each other both during initial symmetry breaking and within the signaling feedback loop mechanisms used to preserve the polarized state.
Collapse
Affiliation(s)
- Tomáš Mazel
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague 2, Czech Republic.
- State Institute for Drug Control, Šrobárova 48, 100 41, Prague 10, Czech Republic.
| |
Collapse
|
28
|
Fruleux A, Hawkins RJ. Physical role for the nucleus in cell migration. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2016; 28:363002. [PMID: 27406341 DOI: 10.1088/0953-8984/28/36/363002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cell migration is important for the function of many eukaryotic cells. Recently the nucleus has been shown to play an important role in cell motility. After giving an overview of cell motility mechanisms we review what is currently known about the mechanical properties of the nucleus and the connections between it and the cytoskeleton. We also discuss connections to the extracellular matrix and mechanotransduction. We identify key physical roles of the nucleus in cell migration.
Collapse
Affiliation(s)
- Antoine Fruleux
- Department of Physics and Astronomy, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield S3 7RH, UK
| | | |
Collapse
|
29
|
Pallesi-Pocachard E, Bazellieres E, Viallat-Lieutaud A, Delgrossi MH, Barthelemy-Requin M, Le Bivic A, Massey-Harroche D. Hook2, a microtubule-binding protein, interacts with Par6α and controls centrosome orientation during polarized cell migration. Sci Rep 2016; 6:33259. [PMID: 27624926 PMCID: PMC5021942 DOI: 10.1038/srep33259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Polarity protein complexes function during polarized cell migration and a subset of these proteins localizes to the reoriented centrosome during this process. Despite these observations, the mechanisms behind the recruitment of these polarity complexes such as the aPKC/PAR6α complex to the centrosome are not well understood. Here we identify Hook2 as an interactor for the aPKC/PAR6α complex that functions to localize this complex at the centrosome. We first demonstrate that Hook2 is essential for the polarized Golgi re-orientation towards the migration front. Depletion of Hook2 results in a decrease of PAR6α at the centrosome during cell migration, while overexpression of Hook2 in cells induced the formation of aggresomes with the recruitment of PAR6α, aPKC and PAR3. In addition, we demonstrate that the interaction between the C-terminal domain of Hook2 and the aPKC-binding domain of PAR6α localizes PAR6α to the centrosome during cell migration. Our data suggests that Hook2, a microtubule binding protein, plays an important role in the regulation of PAR6α recruitment to the centrosome to bridge microtubules and the aPKC/PAR complex. This data reveals how some of the polarity protein complexes are recruited to the centrosome and might regulate pericentriolar and microtubule organization and potentially impact on polarized migration.
Collapse
Affiliation(s)
- Emilie Pallesi-Pocachard
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Elsa Bazellieres
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Annelise Viallat-Lieutaud
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Marie-Hélène Delgrossi
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Magali Barthelemy-Requin
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - André Le Bivic
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Dominique Massey-Harroche
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| |
Collapse
|
30
|
Nechipurenko IV, Olivier-Mason A, Kazatskaya A, Kennedy J, McLachlan IG, Heiman MG, Blacque OE, Sengupta P. A Conserved Role for Girdin in Basal Body Positioning and Ciliogenesis. Dev Cell 2016; 38:493-506. [PMID: 27623382 PMCID: PMC5023068 DOI: 10.1016/j.devcel.2016.07.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 03/09/2016] [Accepted: 07/17/2016] [Indexed: 01/16/2023]
Abstract
Primary cilia are ubiquitous sensory organelles that mediate diverse signaling pathways. Cilia position on the cell surface is determined by the location of the basal body (BB) that templates the cilium. The mechanisms that regulate BB positioning in the context of ciliogenesis are largely unknown. Here we show that the conserved signaling and scaffolding protein Girdin localizes to the proximal regions of centrioles and regulates BB positioning and ciliogenesis in Caenorhabditis elegans sensory neurons and human RPE-1 cells. Girdin depletion alters localization of the intercentriolar linker and ciliary rootlet component rootletin, and rootletin knockdown in RPE-1 cells mimics Girdin-dependent phenotypes. C. elegans Girdin also regulates localization of the apical junction component AJM-1, suggesting that in nematodes Girdin may position BBs via rootletin- and AJM-1-dependent anchoring to the cytoskeleton and plasma membrane, respectively. Together, our results describe a conserved role for Girdin in BB positioning and ciliogenesis.
Collapse
Affiliation(s)
- Inna V Nechipurenko
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA.
| | - Anique Olivier-Mason
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Anna Kazatskaya
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA
| | - Julie Kennedy
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ian G McLachlan
- Department of Genetics, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Maxwell G Heiman
- Department of Genetics, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Piali Sengupta
- Department of Biology and National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
31
|
Sharma G, Tsutsumi K, Saito T, Asada A, Ando K, Tomomura M, Hisanaga SI. Kinase activity of endosomal kinase LMTK1A regulates its cellular localization and interactions with cytoskeletons. Genes Cells 2016; 21:1080-1094. [PMID: 27600567 DOI: 10.1111/gtc.12404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/31/2016] [Indexed: 01/07/2023]
Abstract
Neurite formation, a fundamental process in neuronal maturation, requires the coordinated regulation of cytoskeletal reorganization and membrane transport. Compared to the understanding of cytoskeletal functions, less is known about the supply of membranes to growing neurites. Lemur kinase 1A (LMTK1A) is an endosomal protein kinase that is highly expressed in neurons. We recently reported that LMTK1A regulates the trafficking of Rab11-positive recycling endosomes in growing axons and dendrites. Here, we used the kinase-negative (kn) mutant to investigate the role of the kinase activity of LMTK1A in its cellular localization and interactions with the cytoskeleton in Neuro2A and PC-12 cells. Kinase activity was required for the localization of LMTK1A in the perinuclear endocytic recycling compartment. Perinuclear accumulation was microtubule dependent, and LMTK1A wild type (wt) localized mainly on microtubules, whereas kn LMTK1A was found in the actin-rich cell periphery. In the neurites of PC-12 cells, LMTK1A showed contrasting distributions depending on the kinase activity, with wt being located in the microtubule-rich shaft and the kn form in the actin-rich tip. Taken together, these results suggest that the kinase activity of LMTK1A regulates the pathway for endosomal vesicles to transfer from microtubules to actin filaments at the tip of growing neurites.
Collapse
Affiliation(s)
- Govinda Sharma
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Koji Tsutsumi
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Taro Saito
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Akiko Asada
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Kanae Ando
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Mineko Tomomura
- Integral Education Center, Meikai University, Urayasu, Chiba, 279-9950, Japan
| | - Shin-Ichi Hisanaga
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan.
| |
Collapse
|
32
|
Klímová Z, Bráborec V, Maninová M, Čáslavský J, Weber MJ, Vomastek T. Symmetry breaking in spreading RAT2 fibroblasts requires the MAPK/ERK pathway scaffold RACK1 that integrates FAK, p190A-RhoGAP and ERK2 signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2189-200. [PMID: 27212270 DOI: 10.1016/j.bbamcr.2016.05.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/22/2016] [Accepted: 05/17/2016] [Indexed: 11/28/2022]
Abstract
The spreading of adhering cells is a morphogenetic process during which cells break spherical or radial symmetry and adopt migratory polarity with spatially segregated protruding cell front and non-protruding cell rear. The organization and regulation of these symmetry-breaking events, which are both complex and stochastic, are not fully understood. Here we show that in radially spreading cells, symmetry breaking commences with the development of discrete non-protruding regions characterized by large but sparse focal adhesions and long peripheral actin bundles. Establishment of this non-protruding static region specifies the distally oriented protruding cell front and thus determines the polarity axis and the direction of cell migration. The development of non-protruding regions requires ERK2 and the ERK pathway scaffold protein RACK1. RACK1 promotes adhesion-mediated activation of ERK2 that in turn inhibits p190A-RhoGAP signaling by reducing the peripheral localization of p190A-RhoGAP. We propose that sustained ERK signaling at the prospective cell rear induces p190A-RhoGAP depletion from the cell periphery resulting in peripheral actin bundles and cell rear formation. Since cell adhesion activates both ERK and p190A-RhoGAP signaling this constitutes a spatially confined incoherent feed-forward signaling circuit.
Collapse
Affiliation(s)
| | | | | | | | - Michael J Weber
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Tomáš Vomastek
- Institute of Microbiology AS CR, Prague, Czech Republic.
| |
Collapse
|
33
|
Chiapparo G, Lin X, Lescroart F, Chabab S, Paulissen C, Pitisci L, Bondue A, Blanpain C. Mesp1 controls the speed, polarity, and directionality of cardiovascular progenitor migration. J Cell Biol 2016; 213:463-77. [PMID: 27185833 PMCID: PMC4878090 DOI: 10.1083/jcb.201505082] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 04/18/2016] [Indexed: 01/09/2023] Open
Abstract
During embryonic development, Mesp1 marks the earliest cardiovascular progenitors (CPs) and promotes their specification, epithelial-mesenchymal transition (EMT), and cardiovascular differentiation. However, Mesp1 deletion in mice does not impair initial CP specification and early cardiac differentiation but induces cardiac malformations thought to arise from a defect of CP migration. Using inducible gain-of-function experiments during embryonic stem cell differentiation, we found that Mesp2, its closest homolog, was as efficient as Mesp1 at promoting CP specification, EMT, and cardiovascular differentiation. However, only Mesp1 stimulated polarity and directional cell migration through a cell-autonomous mechanism. Transcriptional analysis and chromatin immunoprecipitation experiments revealed that Mesp1 and Mesp2 activate common target genes that promote CP specification and differentiation. We identified two direct Mesp1 target genes, Prickle1 and RasGRP3, that are strongly induced by Mesp1 and not by Mesp2 and that control the polarity and the speed of cell migration. Altogether, our results identify the molecular interface controlled by Mesp1 that links CP specification and cell migration.
Collapse
Affiliation(s)
- Giuseppe Chiapparo
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Xionghui Lin
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Fabienne Lescroart
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Samira Chabab
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Catherine Paulissen
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Lorenzo Pitisci
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Antoine Bondue
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium Department of Cardiology, Hopital Erasme, Brussels B-1070, Belgium
| | - Cédric Blanpain
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium WELBIO, Université Libre de Bruxelles, Brussels B-1070, Belgium
| |
Collapse
|
34
|
Galletta BJ, Jacobs KC, Fagerstrom CJ, Rusan NM. Asterless is required for centriole length control and sperm development. J Cell Biol 2016; 213:435-50. [PMID: 27185836 PMCID: PMC4878089 DOI: 10.1083/jcb.201501120] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/19/2016] [Indexed: 12/18/2022] Open
Abstract
Loss of the centriole protein Asterless (Asl) prevents centriole duplication, which has limited the study of its function at centrioles. Here, Galletta et al. show that Asl controls centriole length and ensures proper basal body functions during spermatogenesis. Centrioles are the foundation of two organelles, centrosomes and cilia. Centriole numbers and functions are tightly controlled, and mutations in centriole proteins are linked to a variety of diseases, including microcephaly. Loss of the centriole protein Asterless (Asl), the Drosophila melanogaster orthologue of Cep152, prevents centriole duplication, which has limited the study of its nonduplication functions. Here, we identify populations of cells with Asl-free centrioles in developing Drosophila tissues, allowing us to assess its duplication-independent function. We show a role for Asl in controlling centriole length in germline and somatic tissue, functioning via the centriole protein Cep97. We also find that Asl is not essential for pericentriolar material recruitment or centrosome function in organizing mitotic spindles. Lastly, we show that Asl is required for proper basal body function and spermatid axoneme formation. Insights into the role of Asl/Cep152 beyond centriole duplication could help shed light on how Cep152 mutations lead to the development of microcephaly.
Collapse
Affiliation(s)
- Brian J Galletta
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Katherine C Jacobs
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Carey J Fagerstrom
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Nasser M Rusan
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
Impaired immunological synapse in sperm associated antigen 6 (SPAG6) deficient mice. Sci Rep 2016; 6:25840. [PMID: 27169488 PMCID: PMC4864328 DOI: 10.1038/srep25840] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 04/22/2016] [Indexed: 11/18/2022] Open
Abstract
Sperm associated antigen 6 (SPAG6), a component of the central apparatus of the “9 + 2” axoneme, plays a central role in ciliary and flagellar motility; but, its contribution to adaptive immunity and immune system development is completely unknown. While immune cells lack a cilium, the immunological synapse is a surrogate cilium as it utilizes the same machinery as ciliogenesis including the nucleation of microtubules at the centrosome. This prompted our hypothesis that SPAG6 critically regulates the formation and function of immunological synapses. Using bone marrow reconstitution studies of adult WT mice, we demonstrate that SPAG6 is expressed in primary and secondary lymphoid tissues, is associated with the centrosome in lymphocytes, and its deficiency results in synapse disruption due to loss of centrosome polarization and actin clearance at the synaptic cleft. Improper synapse formation in Spag6KO mice was associated with defective CTL functions and impaired humoral immunity as indicated by reduced germinal centers reactions, follicular CD4 T cells, and production of class-switched antibody, together with expansion of B1 B cells. This novel report demonstrates the requirement of SPAG6 for optimal synapse formation and function, its direct role in immune cell function, and provides a novel mechanism for infertility disorders related to SPAG6.
Collapse
|
36
|
Sepich DS, Solnica-Krezel L. Intracellular Golgi Complex organization reveals tissue specific polarity during zebrafish embryogenesis. Dev Dyn 2016; 245:678-91. [PMID: 27043944 DOI: 10.1002/dvdy.24409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 03/15/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cell polarity is essential for directed migration of mesenchymal cells and morphogenesis of epithelial tissues. Studies in cultured cells indicate that a condensed Golgi Complex (GC) is essential for directed protein trafficking to establish cell polarity underlying directed cell migration. Dynamic changes of the GC intracellular organization during early vertebrate development remain to be investigated. RESULTS We used antibody labeling and fusion proteins in vivo to study the organization and intracellular placement of the GC during early zebrafish embryogenesis. We found that the GC was dispersed into several puncta containing cis- and trans-Golgi Complex proteins, presumably ministacks, until the end of the gastrula period. By early segmentation stages, the GC condensed in cells of the notochord, adaxial mesoderm, and neural plate, and its intracellular position became markedly polarized away from borders between these tissues. CONCLUSIONS We find that GC is dispersed in early zebrafish cells, even when cells are engaged in massive gastrulation movements. The GC accumulates into patches in a stage and cell-type specific manner, and becomes polarized away from borders between the embryonic tissues. With respect to tissue borders, intracellular GC polarity in notochord is independent of mature apical/basal polarity, Wnt/PCP, or signals from adaxial mesoderm. Developmental Dynamics 245:678-691, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Diane S Sepich
- Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
37
|
Obino D, Farina F, Malbec O, Sáez PJ, Maurin M, Gaillard J, Dingli F, Loew D, Gautreau A, Yuseff MI, Blanchoin L, Théry M, Lennon-Duménil AM. Actin nucleation at the centrosome controls lymphocyte polarity. Nat Commun 2016; 7:10969. [PMID: 26987298 PMCID: PMC4802043 DOI: 10.1038/ncomms10969] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023] Open
Abstract
Cell polarity is required for the functional specialization of many cell types including lymphocytes. A hallmark of cell polarity is the reorientation of the centrosome that allows repositioning of organelles and vesicles in an asymmetric fashion. The mechanisms underlying centrosome polarization are not fully understood. Here we found that in resting lymphocytes, centrosome-associated Arp2/3 locally nucleates F-actin, which is needed for centrosome tethering to the nucleus via the LINC complex. Upon lymphocyte activation, Arp2/3 is partially depleted from the centrosome as a result of its recruitment to the immune synapse. This leads to a reduction in F-actin nucleation at the centrosome and thereby allows its detachment from the nucleus and polarization to the synapse. Therefore, F-actin nucleation at the centrosome—regulated by the availability of the Arp2/3 complex—determines its capacity to polarize in response to external stimuli. Cell polarity is marked by re-orientation of the centrosome, but the mechanisms governing centrosome polarization are poorly understood. Here Obino et al. show that in lymphocytes centrosome-associated Arp2/3 nucleates actin that tethers the centrosome to the nucleus; activation depletes Arp2/3 from the centrosome and frees it from the nucleus.
Collapse
Affiliation(s)
- Dorian Obino
- INSERM-U932 Immunité et Cancer, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | - Francesca Farina
- CytoMorpho Lab, Biosciences &Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, Grenoble 38054, France
| | - Odile Malbec
- INSERM-U932 Immunité et Cancer, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | - Pablo J Sáez
- INSERM-U932 Immunité et Cancer, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | - Mathieu Maurin
- INSERM-U932 Immunité et Cancer, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | - Jérémie Gaillard
- CytoMorpho Lab, Biosciences &Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, Grenoble 38054, France
| | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| | | | - Maria-Isabel Yuseff
- Departamento de Biologia Celular y Molecular, Pontificia Universidad Catolica de Chile, Santiago 6513677, Chile
| | - Laurent Blanchoin
- CytoMorpho Lab, Biosciences &Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, Grenoble 38054, France
| | - Manuel Théry
- CytoMorpho Lab, Biosciences &Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, Grenoble 38054, France.,CytoMorpho Lab, Hopital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CEA/INSERM/Université Paris Diderot, Paris 75010, France
| | - Ana-Maria Lennon-Duménil
- INSERM-U932 Immunité et Cancer, Institut Curie, PSL Research University, 75248 Paris Cedex 05, France
| |
Collapse
|
38
|
Hung HF, Hehnly H, Doxsey S. The Mother Centriole Appendage Protein Cenexin Modulates Lumen Formation through Spindle Orientation. Curr Biol 2016; 26:793-801. [PMID: 26948879 DOI: 10.1016/j.cub.2016.01.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/04/2015] [Accepted: 01/12/2016] [Indexed: 01/16/2023]
Abstract
Establishing apical-basal polarity is instrumental in the functional shaping of a solitary lumen within an acinus. By exploiting micropatterned slides, wound healing assays, and three-dimensional culture systems, we identified a mother centriole subdistal appendage protein, cenexin, as a critical player in symmetric lumen expansion through the control of microtubule organization. In this regard, cenexin was required for both centrosome positioning in interphase cells and proper spindle orientation during mitosis. In contrast, the essential mother centriole distal appendage protein CEP164 did not play a role in either process, demonstrating the specificity of subdistal appendages for these events. Importantly, upon closer examination we found that cenexin depletion decreased astral microtubule length, disrupted astral microtubule minus-end organization, and increased levels of the polarity protein NuMA at the cell cortex. Interestingly, spindle misorientation and NuMA mislocalization were reversed by treatment with a low dose of the microtubule-stabilizing agent paclitaxel. Taken together, these results suggest that cenexin modulates microtubule organization and stability to mediate spindle orientation.
Collapse
Affiliation(s)
- Hui-Fang Hung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heidi Hehnly
- Department of Cell and Developmental Biology, State University of New York Upstate Medical School, Syracuse, NY 13210, USA.
| | - Stephen Doxsey
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
39
|
Chang W, Antoku S, Östlund C, Worman HJ, Gundersen GG. Linker of nucleoskeleton and cytoskeleton (LINC) complex-mediated actin-dependent nuclear positioning orients centrosomes in migrating myoblasts. Nucleus 2015; 6:77-88. [PMID: 25587885 DOI: 10.1080/19491034.2015.1004947] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Myoblast migration is essential for muscle development and repair; however, the factors that contribute to the polarity of migrating myoblasts are relatively unknown. We find that randomly migrating C2C12 myoblasts orient their centrosomes in the direction of migration. Using wounded monolayers, we further show that centrosome orientation is stimulated by the serum factor lysophosphatidic acid (LPA) and involves the rearward movement of the nucleus while the centrosome is maintained at the cell centroid. The rate of nuclear movement correlated with that of actin retrograde flow and both cytochalasin D and blebbistatin prevented nuclear movement and centrosome orientation. Actin-dependent rearward nuclear movement in fibroblasts is mediated by assembly of nuclear membrane nesprin-2G and SUN2 LINC complexes into transmembrane actin-associated nuclear (TAN) lines anchored by A-type lamins and emerin. In C2C12 myoblasts, depletion of nesprin-2G, SUN2 or lamin A/C prevented nuclear movement and endogenous nesprin-2G and a chimeric GFP-mini-nesprin-2G formed TAN lines during nuclear movement. Depleting nesprin-2G strongly interfered with directed cell migration and reduced the efficiency of myoblast fusion into multinucleated myotubes. Our results show that nuclear movement contributes to centrosome orientation and polarity for efficient migration and fusion of myoblasts. Given that mutations in the genes encoding A-type lamins, nesprin-2 and SUN2 cause Emery-Dreifuss muscular dystrophy and related myopathies, our results have implications for understanding the mechanism of disease pathogenesis.
Collapse
Affiliation(s)
- Wakam Chang
- a Department of Pathology and Cell Biology ; College of Physicians and Surgeons; Columbia University ; New York , NY USA
| | | | | | | | | |
Collapse
|
40
|
Maurizio E, Wiśniewski JR, Ciani Y, Amato A, Arnoldo L, Penzo C, Pegoraro S, Giancotti V, Zambelli A, Piazza S, Manfioletti G, Sgarra R. Translating Proteomic Into Functional Data: An High Mobility Group A1 (HMGA1) Proteomic Signature Has Prognostic Value in Breast Cancer. Mol Cell Proteomics 2015; 15:109-23. [PMID: 26527623 PMCID: PMC4762532 DOI: 10.1074/mcp.m115.050401] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Indexed: 12/11/2022] Open
Abstract
Cancer is a very heterogeneous disease, and biological variability adds a further level of complexity, thus limiting the ability to identify new genes involved in cancer development. Oncogenes whose expression levels control cell aggressiveness are very useful for developing cellular models that permit differential expression screenings in isogenic contexts. HMGA1 protein has this unique property because it is a master regulator in breast cancer cells that control the transition from a nontumorigenic epithelial-like phenotype toward a highly aggressive mesenchymal-like one. The proteins extracted from HMGA1-silenced and control MDA-MB-231 cells were analyzed using label-free shotgun mass spectrometry. The differentially expressed proteins were cross-referenced with DNA microarray data obtained using the same cellular model and the overlapping genes were filtered for factors linked to poor prognosis in breast cancer gene expression meta-data sets, resulting in an HMGA1 protein signature composed of 21 members (HRS, HMGA1 reduced signature). This signature had a prognostic value (overall survival, relapse-free survival, and distant metastasis-free survival) in breast cancer. qRT-PCR, Western blot, and immunohistochemistry analyses validated the link of three members of this signature (KIFC1, LRRC59, and TRIP13) with HMGA1 expression levels both in vitro and in vivo and wound healing assays demonstrated that these three proteins are involved in modulating tumor cell motility. Combining proteomic and genomic data with the aid of bioinformatic tools, our results highlight the potential involvement in neoplastic transformation of a restricted list of factors with an as-yet-unexplored role in cancer. These factors are druggable targets that could be exploited for the development of new, targeted therapeutic approaches in triple-negative breast cancer.
Collapse
Affiliation(s)
- Elisa Maurizio
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Jacek R Wiśniewski
- §Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Yari Ciani
- ¶Laboratorio Nazionale CIB, (LNCIB), Area Science Park, 34149 Trieste, Italy
| | - Angela Amato
- ¶¶Laboratory of Experimental Oncology and Pharmacogenomics IRCCS - Salvatore Maugeri Foundation, 27100 Pavia, Italy
| | - Laura Arnoldo
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Carlotta Penzo
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Silvia Pegoraro
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Vincenzo Giancotti
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Alberto Zambelli
- ‖Department of Medical Oncology, Hospital Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Silvano Piazza
- ¶Laboratorio Nazionale CIB, (LNCIB), Area Science Park, 34149 Trieste, Italy
| | | | - Riccardo Sgarra
- From the ‡Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
41
|
Shahbazi MN, Perez-Moreno M. Connections between cadherin-catenin proteins, spindle misorientation, and cancer. Tissue Barriers 2015; 3:e1045684. [PMID: 26451345 DOI: 10.1080/21688370.2015.1045684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/18/2015] [Accepted: 04/21/2015] [Indexed: 10/25/2022] Open
Abstract
Cadherin-catenin mediated adhesion is an important determinant of tissue architecture in multicellular organisms. Cancer progression and maintenance is frequently associated with loss of their expression or functional activity, which not only leads to decreased cell-cell adhesion, but also to enhanced tumor cell proliferation and loss of differentiated characteristics. This review is focused on the emerging implications of cadherin-catenin proteins in the regulation of polarized divisions through their connections with the centrosomes, cytoskeleton, tissue tension and signaling pathways; and illustrates how alterations in cadherin-catenin levels or functional activity may render cells susceptible to transformation through the loss of their proliferation-differentiation balance.
Collapse
Affiliation(s)
- Marta N Shahbazi
- Department of Physiology, Development, and Neuroscience; University of Cambridge ; Cambridge, UK
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group; Cancer Cell Biology Program; Spanish National Cancer Research Centre ; Madrid, Spain
| |
Collapse
|
42
|
Abstract
As a large, nonmembrane bound organelle, the centrosome must rely heavily on protein-protein interactions to assemble itself in the cytoplasm and perform its functions as a microtubule-organizing center. Therefore, to understand how this organelle is built and functions, one must understand the protein-protein interactions made by each centrosome protein. Unfortunately, the highly interconnected nature of the centrosome, combined with its predicted unstructured, coil-rich proteins, has made the use of many standard approaches to studying protein-protein interactions very challenging. The yeast-two hybrid (Y2H) system is well suited for studying the centrosome and is an important complement to other biochemical approaches. In this chapter we describe how to carry out a directed Y2H screen to identify the direct interactions between a given centrosome protein and a library of others. Specifically, we detail using a bioinformatics-based approach (structure prediction programs) to subdivide proteins and screen for interactions using an array-based Y2H approach. We also describe how to use the interaction information garnered from this screen to generate mutations to disrupt specific interactions using mutagenic-PCR and a "reverse" Y2H screen. Finally, we discuss how information from such a screen can be integrated into existing models of centrosome assembly and how it can initiate and guide extensive in vitro and in vivo experimentation to test these models.
Collapse
|
43
|
Ballister ER, Aonbangkhen C, Mayo AM, Lampson MA, Chenoweth DM. Localized light-induced protein dimerization in living cells using a photocaged dimerizer. Nat Commun 2014; 5:5475. [PMID: 25400104 PMCID: PMC4308733 DOI: 10.1038/ncomms6475] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/06/2014] [Indexed: 12/17/2022] Open
Abstract
Regulated protein localization is critical for many cellular processes. Several techniques have been developed for experimental control over protein localization, including chemically induced and light-induced dimerization, which both provide temporal control. Light-induced dimerization offers the distinct advantage of spatial precision within subcellular length scales. A number of elegant systems have been reported that utilize natural light-sensitive proteins to induce dimerization via direct protein-protein binding interactions, but the application of these systems at cellular locations beyond the plasma membrane has been limited. Here we present a new technique to rapidly and reversibly control protein localization in living cells with subcellular spatial resolution using a cell-permeable, photoactivatable chemical inducer of dimerization. We demonstrate light-induced recruitment of a cytosolic protein to individual centromeres, kinetochores, mitochondria and centrosomes in human cells, indicating that our system is widely applicable to many cellular locations.
Collapse
Affiliation(s)
- Edward R Ballister
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Chanat Aonbangkhen
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Alyssa M Mayo
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David M Chenoweth
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|