51
|
Planar Cell Polarity Signaling in Mammalian Cardiac Morphogenesis. Pediatr Cardiol 2018; 39:1052-1062. [PMID: 29564519 PMCID: PMC5959767 DOI: 10.1007/s00246-018-1860-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 01/16/2023]
Abstract
The mammalian heart is the first organ to form and is critical for embryonic survival and development. With an occurrence of 1%, congenital heart defects (CHDs) are also the most common birth defects in humans, and major cause of childhood morbidity and mortality (Hoffman and Kaplan in J Am Coll Cardiol 39(12):1890-1900, 2002; Samanek in Cardiol Young 10(3):179-185, 2000). Understanding how the heart forms will not only help to determine the etiology and to design diagnostic and therapeutic approaches for CHDs, but may also provide insight into regenerative medicine to repair injured adult hearts. Mammalian heart development requires precise orchestration of growth, differentiation, and morphogenesis to remodel a simple linear heart tube into an intricate, four-chambered heart with properly connected pulmonary artery and aorta, a structural basis for establishing the pulmonary and systemic circulation. Here we will review the recent advance in our understanding of how the planar cell polarity pathway, a highly conserved morphogenetic engine in vertebrates, regulates polarized morphogenetic processes to contribute to both the arterial and venous poles development of the heart.
Collapse
|
52
|
Harris AR, Jreij P, Fletcher DA. Mechanotransduction by the Actin Cytoskeleton: Converting Mechanical Stimuli into Biochemical Signals. Annu Rev Biophys 2018. [DOI: 10.1146/annurev-biophys-070816-033547] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Force transmission through the actin cytoskeleton plays a central role in cell movements, shape change, and internal organization. Dynamic reorganization of actin filaments by an array of specialized binding proteins creates biochemically and architecturally distinct structures, many of which are finely tuned to exert or resist mechanical loads. The molecular complexity of the actin cytoskeleton continues to be revealed by detailed biochemical assays, and the architectural diversity and dynamics of actin structures are being uncovered by advances in super-resolution fluorescence microscopy and electron microscopy. However, our understanding of how mechanical forces feed back on cytoskeletal architecture and actin-binding protein organization is comparatively limited. In this review, we discuss recent work investigating how mechanical forces applied to cytoskeletal proteins are transduced into biochemical signals. We explore multiple mechanisms for mechanical signal transduction, including the mechanosensitive behavior of actin-binding proteins, the effect of mechanical force on actin filament dynamics, and the influence of mechanical forces on the structure of single actin filaments. The emerging picture is one in which the actin cytoskeleton is defined not only by the set of proteins that constitute a network but also by the constant interplay of mechanical forces and biochemistry.
Collapse
Affiliation(s)
- Andrew R. Harris
- Department of Bioengineering, and Biophysics Program, University of California, Berkeley, California 94720, USA
| | - Pamela Jreij
- Department of Bioengineering, and Biophysics Program, University of California, Berkeley, California 94720, USA
| | - Daniel A. Fletcher
- Department of Bioengineering, and Biophysics Program, University of California, Berkeley, California 94720, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| |
Collapse
|
53
|
Ossipova O, Kerney R, Saint-Jeannet JP, Sokol SY. Regulation of neural crest development by the formin family protein Daam1. Genesis 2018; 56:e23108. [PMID: 29673042 DOI: 10.1002/dvg.23108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/16/2018] [Accepted: 03/26/2018] [Indexed: 01/12/2023]
Abstract
The neural crest (NC) multipotent progenitor cells form at the neural plate border and migrate to diverse locations in the embryo to differentiate into many cell types. NC is specified by several embryonic pathways, however the role of noncanonical Wnt signaling in this process remains poorly defined. Daam1 is a formin family protein that is present in embryonic ectoderm at the time of NC formation and can mediate noncanonical Wnt signaling. Our interference experiments indicated that Daam1 is required for NC gene activation. To further study the function of Daam1 in NC development we used a transgenic reporter Xenopus line, in which GFP transcription is driven by sox10 upstream regulatory sequences. The activation of the sox10:GFP reporter in a subset of NC cells was suppressed after Daam1 depletion and in embryos expressing N-Daam1, a dominant interfering construct. Moreover, N-Daam1 blocked reporter activation in neuralized ectodermal explants in response to Wnt11, but not Wnt8 or Wnt3a, confirming that the downstream pathways are different. In complementary experiments, a constitutively active Daam1 fragment expanded the NC territory, but this gain-of-function activity was eliminated in a construct with a point mutation in the FH2 domain that is critical for actin polymerization. These observations suggest a new role of Daam1 and actin remodeling in NC specification.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ryan Kerney
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania
| | - Jean-Pierre Saint-Jeannet
- Department of Basic Science and Craniofacial Biology,College of Dentistry, New York University, New York, New York
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
54
|
El-Hayek S, Yang Q, Abbassi L, FitzHarris G, Clarke HJ. Mammalian Oocytes Locally Remodel Follicular Architecture to Provide the Foundation for Germline-Soma Communication. Curr Biol 2018; 28:1124-1131.e3. [PMID: 29576478 DOI: 10.1016/j.cub.2018.02.039] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/02/2018] [Accepted: 02/15/2018] [Indexed: 01/11/2023]
Abstract
Germ cells develop in a microenvironment created by the somatic cells of the gonad [1-3]. Although in males, the germ and somatic support cells lie in direct contact, in females, a thick extracellular coat surrounds the oocyte, physically separating it from the somatic follicle cells [4]. To bypass this barrier to communication, narrow cytoplasmic extensions of the follicle cells traverse the extracellular coat to reach the oocyte plasma membrane [5-9]. These delicate structures provide the sole platform for the contact-mediated communication between the oocyte and its follicular environment that is indispensable for production of a fertilizable egg [8, 10-15]. Identifying the mechanisms underlying their formation should uncover conserved regulators of fertility. We show here in mice that these structures, termed transzonal projections (TZPs), are specialized filopodia whose number amplifies enormously as oocytes grow, enabling increased germ-soma communication. By creating chimeric complexes of genetically tagged oocytes and follicle cells, we demonstrate that follicle cells elaborate new TZPs that push through the extracellular coat to reach the oocyte surface. We further show that growth-differentiation factor 9, produced by the oocyte, drives the formation of new TZPs, uncovering a key yet unanticipated role for the germ cell in building these essential bridges of communication. Moreover, TZP number and germline-soma communication are strikingly reduced in reproductively aged females. Thus, the growing oocyte locally remodels follicular architecture to ensure that its developmental needs are met, and an inability of somatic follicle cells to respond appropriately to oocyte-derived cues may contribute to human infertility.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Department of Biology, McGill University, Montreal, QC, Canada; Research Institute, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC H4A 3J1, Canada
| | - Qin Yang
- Research Institute, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC H4A 3J1, Canada
| | - Laleh Abbassi
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Research Institute, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC H4A 3J1, Canada
| | - Greg FitzHarris
- Centre Recherche CHUM and Département d'Obstétrique et de Gynécologie, Université de Montréal, 900 rue St-Denis, Montreal, QC H2X 0A9, Canada
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Department of Biology, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Research Institute, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
55
|
Xiong H, Yan T, Zhang W, Shi F, Jiang X, Wang X, Li S, Chen Y, Chen C, Zhu Y. miR-613 inhibits cell migration and invasion by downregulating Daam1 in triple-negative breast cancer. Cell Signal 2018; 44:33-42. [PMID: 29339084 DOI: 10.1016/j.cellsig.2018.01.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 11/25/2022]
Abstract
Dishevelled-associated activator of morphogenesis 1 (Daam1) is a formin protein and participates in regulating cell migration of triple-negative breast cancer (TNBC) cells. The specific miRNA targeting Daam1 and mediating cell migration and invasion remains obscure. This experiment investigated the suppressive role of miR-613 in TNBC cells. The luciferase activity of Daam1 3'-untranslated region (3'-UTR) based reporters constructed in HEK-293T and MCF-7 cells suggested that Daam1 was the target gene of miR-613. Overexpressed miR-613 reduced the protein level of Daam1, weakened RhoA activity, and retarded the cell migration, cell invasion and colony formation of TNBC cells. Overexpression of Daam1 or RhoA rescued cell migration and invasion in miR-613-overexpressed TNBC cells, but failed to reverse colony formation. MiR-613 was significantly downregulated in breast cancer tissues compared with that in adjacent normal tissues. This downregulation in TNBC tissues and lymphnode metastatic breast cancer tissues was more obvious than that in non-TNBC tissues and non-metastatic cancer tissues, respectively. MiR-613 weakens the resistance of TNBC cells against paclitaxel rather than adriamycin, cyclophosphamide, docetaxel, and kaempferol. Taken together, miR-613 is involved in cell migration and invasion of TNBC cells via targeting Daam1/RhoA signaling pathway.
Collapse
Affiliation(s)
- Huaping Xiong
- Department of Oncology, Traditional Chinese Medical Hospital of Siyang County, Siyang 223700, Jiangsu, China
| | - Ting Yan
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Weijie Zhang
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210009, Jiangsu, China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu, China
| | - Xuesong Jiang
- Department of Radiotherapy, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, Jiangsu, China
| | - Xiaohua Wang
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, Jiangsu, China
| | - Shoushan Li
- Department of Oncology, Traditional Chinese Medical Hospital of Siyang County, Siyang 223700, Jiangsu, China
| | - Ying Chen
- Department of Oncology, Traditional Chinese Medical Hospital of Siyang County, Siyang 223700, Jiangsu, China
| | - Cheng Chen
- Department of Radiotherapy, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, Jiangsu, China.
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
56
|
Abstract
The planar cell polarity (PCP) pathway is best known for its role in polarizing epithelial cells within the plane of a tissue but it also plays a role in a range of cell migration events during development. The mechanism by which the PCP pathway polarizes stationary epithelial cells is well characterized, but how PCP signaling functions to regulate more dynamic cell behaviors during directed cell migration is much less understood. Here, we review recent discoveries regarding the localization of PCP proteins in migrating cells and their impact on the cell biology of collective and individual cell migratory behaviors.
Collapse
Affiliation(s)
- Crystal F Davey
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, B2-159, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | - Cecilia B Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, B2-159, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| |
Collapse
|
57
|
Wilking-Busch MJ, Ndiaye MA, Liu X, Ahmad N. RNA interference-mediated knockdown of SIRT1 and/or SIRT2 in melanoma: Identification of downstream targets by large-scale proteomics analysis. J Proteomics 2017; 170:99-109. [PMID: 28882678 DOI: 10.1016/j.jprot.2017.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/14/2017] [Accepted: 09/03/2017] [Indexed: 02/07/2023]
Abstract
Melanoma is the most notorious and fatal of all skin cancers and the existing treatment options have not been proven to effectively manage this neoplasm, especially the metastatic disease. Sirtuin (SIRT) proteins have been shown to be differentially expressed in melanoma. We have shown that SIRTs 1 and 2 were overexpressed in melanoma and inhibition of SIRT1 imparts anti-proliferative responses in human melanoma cells. To elucidate the impact of SIRT 1 and/or 2 in melanoma, we created stable knockdowns of SIRTs 1, 2, and their combination using shRNA mediated RNA interference in A375 human melanoma cells. We found that SIRT1 and SIRT1&2 combination knockdown caused a decreased cellular proliferation in melanoma cells. Further, the knockdown of SIRT 1 and/or 2 resulted in a decreased colony formation in melanoma cells. To explore the downstream targets of SIRTs 1 and/or 2, we employed a label-free quantitative nano-LC-MS/MS proteomics analysis using the stable lines. We found aberrant levels of proteins involved in many vital cellular processes, including cytoskeletal organization, ribosomal activity, oxidative stress response, and angiogenesis. These findings provide clear evidence of cellular systems undergoing alterations in response to sirtuin inhibition, and have unveiled several excellent candidates for future study. SIGNIFICANCE Melanoma is the deadliest form of skin cancer, due to its aggressive nature, metastatic potential, and a lack of sufficient treatment options for advanced disease. Therefore, detailed investigations into the molecular mechanisms of melanoma growth and progression are needed. In the search for candidate genes to serve as therapeutic targets, the sirtuins show promise as they have been found to be upregulated in melanoma and they regulate a large number of proteins involved in cellular processes known to affect tumor growth, such as DNA damage repair, cell cycle arrest, and apoptosis. In this study, we used a large-scale label-free comparative proteomics system to identify novel protein targets that are affected following knockdown of SIRT1 and/or 2 in A375 metastatic melanoma cell line. Our study offers important insight into the potential downstream targets of SIRTs 1 and/or 2. This may unravel new potential areas of exploration in melanoma research.
Collapse
Affiliation(s)
- Melissa J Wilking-Busch
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Mary A Ndiaye
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, 175 S. University Street, West Lafayette, IN, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA; William S. Middleton VA Medical Center, 2500 Overlook Terrace, Madison, WI 53705, USA.
| |
Collapse
|
58
|
Lu Y, Zhang Y, Pan MH, Kim NH, Sun SC, Cui XS. Daam1 regulates fascin for actin assembly in mouse oocyte meiosis. Cell Cycle 2017; 16:1350-1356. [PMID: 28682694 DOI: 10.1080/15384101.2017.1325045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
As a formin protein, Daam1 (Dishevelled-associated activator of morphogenesis 1) is reported to regulate series of cell processes like endocytosis, cell morphology and migration via its effects on actin assembly in mitosis. However, whether Daam1 plays roles in female meiosis remains uncertain. In this study, we investigated the expression and functions of Daam1 during mouse oocyte meiosis. Our results indicated that Daam1 localized at the cortex of oocytes, which was similar with actin filaments. After Daam1 morpholino (MO) microinjection, the expression of Daam1 significantly decreased, which resulted in the failure of oocyte polar body extrusion. These results might be due to the defects of actin assembly, since the decreased fluorescence intensity of actin filaments in oocyte cortex and cytoplasm were observed. However, Daam1 knockdown seemed not to affect the meiotic spindle movement. In addition, we found that fascin might be the down effector of Daam1, since the protein expression of fascin decreased after Daam1 knockdown. Thus, our data suggested that Daam1 affected actin assembly during oocyte meiotic division via the regulation of fascin expression.
Collapse
Affiliation(s)
- Yujie Lu
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yu Zhang
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Meng-Hao Pan
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Nam-Hyung Kim
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| | - Shao-Chen Sun
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Xiang-Shun Cui
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| |
Collapse
|
59
|
Juanes MA, Bouguenina H, Eskin JA, Jaiswal R, Badache A, Goode BL. Adenomatous polyposis coli nucleates actin assembly to drive cell migration and microtubule-induced focal adhesion turnover. J Cell Biol 2017; 216:2859-2875. [PMID: 28663347 PMCID: PMC5584174 DOI: 10.1083/jcb.201702007] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/14/2017] [Accepted: 05/25/2017] [Indexed: 02/07/2023] Open
Abstract
Cell motility depends on tight coordination between the microtubule (MT) and actin cytoskeletons, but the mechanisms underlying this MT-actin cross talk have remained poorly understood. Here, we show that the tumor suppressor protein adenomatous polyposis coli (APC), which is a known MT-associated protein, directly nucleates actin assembly to promote directed cell migration. By changing only two residues in APC, we generated a separation-of-function mutant, APC (m4), that abolishes actin nucleation activity without affecting MT interactions. Expression of full-length APC carrying the m4 mutation (APC (m4)) rescued cellular defects in MT organization, MT dynamics, and mitochondrial distribution caused by depletion of endogenous APC but failed to restore cell migration. Wild-type APC and APC (m4) localized to focal adhesions (FAs), and APC (m4) was defective in promoting actin assembly at FAs to facilitate MT-induced FA turnover. These results provide the first direct evidence for APC-mediated actin assembly in vivo and establish a role for APC in coordinating MTs and actin at FAs to direct cell migration.
Collapse
Affiliation(s)
| | - Habib Bouguenina
- Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Institut Paoli-Calmettes, Aix-Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | | | - Richa Jaiswal
- Department of Biology, Brandeis University, Waltham, MA
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille, Institut National de la Santé et de la Recherche Médicale, Institut Paoli-Calmettes, Aix-Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - Bruce L Goode
- Department of Biology, Brandeis University, Waltham, MA
| |
Collapse
|
60
|
Vig AT, Földi I, Szikora S, Migh E, Gombos R, Tóth MÁ, Huber T, Pintér R, Talián GC, Mihály J, Bugyi B. The activities of the C-terminal regions of the formin protein disheveled-associated activator of morphogenesis (DAAM) in actin dynamics. J Biol Chem 2017. [PMID: 28642367 DOI: 10.1074/jbc.m117.799247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Disheveled-associated activator of morphogenesis (DAAM) is a diaphanous-related formin protein essential for the regulation of actin cytoskeleton dynamics in diverse biological processes. The conserved formin homology 1 and 2 (FH1-FH2) domains of DAAM catalyze actin nucleation and processively mediate filament elongation. These activities are indirectly regulated by the N- and C-terminal regions flanking the FH1-FH2 domains. Recently, the C-terminal diaphanous-autoregulatory domain (DAD) and the C terminus (CT) of formins have also been shown to regulate actin assembly by directly interacting with actin. Here, to better understand the biological activities of DAAM, we studied the role of DAD-CT regions of Drosophila DAAM in its interaction with actin with in vitro biochemical and in vivo genetic approaches. We found that the DAD-CT region binds actin in vitro and that its main actin-binding element is the CT region, which does not influence actin dynamics on its own. However, we also found that it can tune the nucleating activity and the filament end-interaction properties of DAAM in an FH2 domain-dependent manner. We also demonstrate that DAD-CT makes the FH2 domain more efficient in antagonizing with capping protein. Consistently, in vivo data suggested that the CT region contributes to DAAM-mediated filopodia formation and dynamics in primary neurons. In conclusion, our results demonstrate that the CT region of DAAM plays an important role in actin assembly regulation in a biological context.
Collapse
Affiliation(s)
- Andrea Teréz Vig
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - István Földi
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Szilárd Szikora
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Ede Migh
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Rita Gombos
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Mónika Ágnes Tóth
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Tamás Huber
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Réka Pintér
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Gábor Csaba Talián
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - József Mihály
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Beáta Bugyi
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624, .,the Szentágothai Research Center, Ifjúság Str. 34, Pécs H-7624, Hungary
| |
Collapse
|
61
|
Szikora S, Földi I, Tóth K, Migh E, Vig A, Bugyi B, Maléth J, Hegyi P, Kaltenecker P, Sanchez-Soriano N, Mihály J. The formin DAAM is required for coordination of the actin and microtubule cytoskeleton in axonal growth cones. J Cell Sci 2017; 130:2506-2519. [PMID: 28606990 DOI: 10.1242/jcs.203455] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/05/2017] [Indexed: 01/10/2023] Open
Abstract
Directed axonal growth depends on correct coordination of the actin and microtubule cytoskeleton in the growth cone. However, despite the relatively large number of proteins implicated in actin-microtubule crosstalk, the mechanisms whereby actin polymerization is coupled to microtubule stabilization and advancement in the peripheral growth cone remained largely unclear. Here, we identified the formin Dishevelled-associated activator of morphogenesis (DAAM) as a novel factor playing a role in concerted regulation of actin and microtubule remodeling in Drosophilamelanogaster primary neurons. In vitro, DAAM binds to F-actin as well as to microtubules and has the ability to crosslink the two filament systems. Accordingly, DAAM associates with the neuronal cytoskeleton, and a significant fraction of DAAM accumulates at places where the actin filaments overlap with that of microtubules. Loss of DAAM affects growth cone and microtubule morphology, and several aspects of microtubule dynamics; and biochemical and cellular assays revealed a microtubule stabilization activity and binding to the microtubule tip protein EB1. Together, these data suggest that, besides operating as an actin assembly factor, DAAM is involved in linking actin remodeling in filopodia to microtubule stabilization during axonal growth.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Krisztina Tóth
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Andrea Vig
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary
| | - Beáta Bugyi
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary.,Szentágothai Research Center, Ifjúság str. 34, Pécs H-7624, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary.,Institute for Translational Medicine, Department of Pathophysiology, University of Pécs, Pécs H-7624, Hungary
| | - Péter Kaltenecker
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - Natalia Sanchez-Soriano
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
62
|
Functional Actin Networks under Construction: The Cooperative Action of Actin Nucleation and Elongation Factors. Trends Biochem Sci 2017; 42:414-430. [DOI: 10.1016/j.tibs.2017.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 03/04/2017] [Accepted: 03/07/2017] [Indexed: 12/31/2022]
|
63
|
Chemek M, Venditti M, Boughamoura S, Mimouna SB, Messaoudi I, Minucci S. Involvement of testicular DAAM1 expression in zinc protection against cadmium‐induced male rat reproductive toxicity. J Cell Physiol 2017; 233:630-640. [DOI: 10.1002/jcp.25923] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/20/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Marouane Chemek
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut de BiotechnologieUniversité de MonastirMonastirTunisia
| | - Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate “F. Bottazzi”Università della Campania Luigi VanvitelliNapoliItaly
| | - Sana Boughamoura
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut de BiotechnologieUniversité de MonastirMonastirTunisia
| | - Safa B. Mimouna
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut de BiotechnologieUniversité de MonastirMonastirTunisia
| | - Imed Messaoudi
- LR11ES41: Génétique, Biodiversité et Valorisation des Bioressources, Institut de BiotechnologieUniversité de MonastirMonastirTunisia
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate “F. Bottazzi”Università della Campania Luigi VanvitelliNapoliItaly
| |
Collapse
|
64
|
Nishimura T, Ito S, Saito H, Hiver S, Shigetomi K, Ikenouchi J, Takeichi M. DAAM1 stabilizes epithelial junctions by restraining WAVE complex-dependent lateral membrane motility. J Cell Biol 2016; 215:559-573. [PMID: 27807130 PMCID: PMC5119936 DOI: 10.1083/jcb.201603107] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/13/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Nishimura et al. show that DAAM1, a formin family actin polymerization regulator, stabilizes epithelial cell junctions by counteracting the WAVE complex, another actin regulator. Loss of DAAM1 promotes the motility of junctional membranes and thereby enhances their invasion of neighboring environments. Epithelial junctions comprise two subdomains, the apical junctional complex (AJC) and the adjacent lateral membrane contacts (LCs), that span the majority of the junction. The AJC is lined with circumferential actin cables, whereas the LCs are associated with less-organized actin filaments whose roles are elusive. We found that DAAM1, a formin family actin regulator, accumulated at the LCs, and its depletion caused dispersion of actin filaments at these sites while hardly affecting circumferential actin cables. DAAM1 loss enhanced the motility of LC-forming membranes, leading to their invasion of neighboring cell layers, as well as disruption of polarized epithelial layers. We found that components of the WAVE complex and its downstream targets were required for the elevation of LC motility caused by DAAM1 loss. These findings suggest that the LC membranes are motile by nature because of the WAVE complex, but DAAM1-mediated actin regulation normally restrains this motility, thereby stabilizing epithelial architecture, and that DAAM1 loss evokes invasive abilities of epithelial cells.
Collapse
Affiliation(s)
- Tamako Nishimura
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Shoko Ito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroko Saito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Sylvain Hiver
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Kenta Shigetomi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | | |
Collapse
|
65
|
Luo W, Lieu ZZ, Manser E, Bershadsky AD, Sheetz MP. Formin DAAM1 Organizes Actin Filaments in the Cytoplasmic Nodal Actin Network. PLoS One 2016; 11:e0163915. [PMID: 27760153 PMCID: PMC5070803 DOI: 10.1371/journal.pone.0163915] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/17/2016] [Indexed: 01/13/2023] Open
Abstract
A nodal cytoplasmic actin network underlies actin cytoplasm cohesion in the absence of stress fibers. We previously described such a network that forms upon Latrunculin A (LatA) treatment, in which formin DAAM1 was localized at these nodes. Knock down of DAAM1 reduced the mobility of actin nodes but the nodes remained. Here we have investigated DAAM1 containing nodes after LatA washout. DAAM1 was found to be distributed between the cytoplasm and the plasma membrane. The membrane binding likely occurs through an interaction with lipid rafts, but is not required for F-actin assembly. Interesting the forced interaction of DAAM1 with plasma membrane through a rapamycin-dependent linkage, enhanced F-actin assembly at the cell membrane (compared to the cytoplasm) after the LatA washout. However, immediately after addition of both rapamycin and LatA, the cytoplasmic actin nodes formed transiently, before DAAM1 moved to the membrane. This was consistent with the idea that DAAM1 was initially anchored to cytoplasmic actin nodes. Further, photoactivatable tracking of DAAM1 showed DAAM1 was immobilized at these actin nodes. Thus, we suggest that DAAM1 organizes actin filaments into a nodal complex, and such nodal complexes seed actin network recovery after actin depolymerization.
Collapse
Affiliation(s)
- Weiwei Luo
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Zi Zhao Lieu
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Ed Manser
- sGSK Group, Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Proteos Building, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Alexander D. Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Michael P. Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Department of Biological Sciences, Columbia University, New York, New York, 10027, United States of America
- * E-mail:
| |
Collapse
|
66
|
Finkenstaedt-Quinn SA, Qiu TA, Shin K, Haynes CL. Super-resolution imaging for monitoring cytoskeleton dynamics. Analyst 2016; 141:5674-5688. [PMID: 27549146 DOI: 10.1039/c6an00731g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cytoskeleton is a key cellular structure that is important in the control of cellular movement, structure, and sensing. To successfully image the individual cytoskeleton components, high resolution and super-resolution fluorescence imaging methods are needed. This review covers the three basic cytoskeletal elements and the relative benefits and drawbacks of fixed versus live cell imaging before moving on to recent studies using high resolution and super-resolution techniques. The techniques covered include the near-diffraction limited imaging methods of confocal microscopy and TIRF microscopy and the super-resolution fluorescence imaging methods of STORM, PALM, and STED.
Collapse
|
67
|
Hong JH, Kwak Y, Woo Y, Park C, Lee SA, Lee H, Park SJ, Suh Y, Suh BK, Goo BS, Mun DJ, Sanada K, Nguyen MD, Park SK. Regulation of the actin cytoskeleton by the Ndel1-Tara complex is critical for cell migration. Sci Rep 2016; 6:31827. [PMID: 27546710 PMCID: PMC4992831 DOI: 10.1038/srep31827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022] Open
Abstract
Nuclear distribution element-like 1 (Ndel1) plays pivotal roles in diverse biological processes and is implicated in the pathogenesis of multiple neurodevelopmental disorders. Ndel1 function by regulating microtubules and intermediate filaments; however, its functional link with the actin cytoskeleton is largely unknown. Here, we show that Ndel1 interacts with TRIO-associated repeat on actin (Tara), an actin-bundling protein, to regulate cell movement. In vitro wound healing and Boyden chamber assays revealed that Ndel1- or Tara-deficient cells were defective in cell migration. Moreover, Tara overexpression induced the accumulation of Ndel1 at the cell periphery and resulted in prominent co-localization with F-actin. This redistribution of Ndel1 was abolished by deletion of the Ndel1-interacting domain of Tara, suggesting that the altered peripheral localization of Ndel1 requires a physical interaction with Tara. Furthermore, co-expression of Ndel1 and Tara in SH-SY5Y cells caused a synergistic increase in F-actin levels and filopodia formation, suggesting that Tara facilitates cell movement by sequestering Ndel1 at peripheral structures to regulate actin remodeling. Thus, we demonstrated that Ndel1 interacts with Tara to regulate cell movement. These findings reveal a novel role of the Ndel1-Tara complex in actin reorganization during cell movement.
Collapse
Affiliation(s)
- Ji-Ho Hong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Yongdo Kwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Cana Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Seol-Ae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Haeryun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Sung Jin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Kamon Sanada
- Molecular Genetics Research Laboratory, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Departments of Clinical Neurosciences, Cell Biology and Anatomy, and Biochemistry and Molecular Biology, University of Calgary, Calgary T2N 4N1, Canada
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| |
Collapse
|
68
|
Devenport D. Tissue morphodynamics: Translating planar polarity cues into polarized cell behaviors. Semin Cell Dev Biol 2016; 55:99-110. [PMID: 26994528 DOI: 10.1016/j.semcdb.2016.03.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/15/2016] [Indexed: 12/21/2022]
Abstract
The ability of cells to collectively orient and align their behaviors is essential in multicellular organisms for unidirectional cilia beating, collective cell movements, oriented cell divisions, and asymmetric cell fate specification. The planar cell polarity pathway coordinates a vast and diverse array of collective cell behaviors by intersecting with downstream pathways that regulate cytoskeletal dynamics and intercellular signaling. How the planar polarity pathway translates directional cues to produce polarized cell behaviors is the focus of this review.
Collapse
Affiliation(s)
- Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
69
|
Chin SM, Jansen S, Goode BL. TIRF microscopy analysis of human Cof1, Cof2, and ADF effects on actin filament severing and turnover. J Mol Biol 2016; 428:1604-16. [PMID: 26996939 DOI: 10.1016/j.jmb.2016.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 10/24/2022]
Abstract
Dynamic remodeling and turnover of cellular actin networks requires actin filament severing by actin-depolymerizing factor (ADF)/Cofilin proteins. Mammals express three different ADF/Cofilins (Cof1, Cof2, and ADF), and genetic studies suggest that in vivo they perform both overlapping and unique functions. To gain mechanistic insights into their different roles, we directly compared their G-actin and F-actin binding affinities, and quantified the actin filament severing activities of human Cof1, Cof2, and ADF using in vitro total internal reflection fluorescence microscopy. All three ADF/Cofilins had similar affinities for G-actin and F-actin. However, Cof2 and ADF severed filaments much more efficiently than Cof1 at both lower and higher concentrations and using either muscle or platelet actin. Furthermore, Cof2 and ADF were more effective than Cof1 in promoting "enhanced disassembly" when combined with actin disassembly co-factors Coronin-1B and actin-interacting protein 1 (AIP1), and these differences were observed on both preformed and actively growing filaments. To probe the mechanism underlying these differences, we used multi-wavelength total internal reflection fluorescence microscopy to directly observe Cy3-Cof1 and Cy3-Cof2 interacting with actin filaments in real time during severing. Cof1 and Cof2 each bound to filaments with similar kinetics, yet Cof2 induced severing much more rapidly than Cof1, decreasing the time interval between initial binding on a filament and severing at the same location. These differences in ADF/Cofilin activities and mechanisms may be used in cells to tune filament turnover rates, which can vary widely for different actin structures.
Collapse
Affiliation(s)
- Samantha M Chin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA
| | - Silvia Jansen
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
70
|
Van Audenhove I, Denert M, Boucherie C, Pieters L, Cornelissen M, Gettemans J. Fascin Rigidity and L-plastin Flexibility Cooperate in Cancer Cell Invadopodia and Filopodia. J Biol Chem 2016; 291:9148-60. [PMID: 26945069 DOI: 10.1074/jbc.m115.706937] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Indexed: 01/15/2023] Open
Abstract
Invadopodia and filopodia are dynamic, actin-based protrusions contributing to cancer cell migration, invasion, and metastasis. The force of actin bundles is essential for their protrusive activity. The bundling protein fascin is known to play a role in both invadopodia and filopodia. As it is more and more acknowledged that functionally related proteins cooperate, it is unlikely that only fascin bundles actin in these protrusions. Another interesting candidate is L-plastin, normally expressed in hematopoietic cells, but considered a common marker of many cancer types. We identified L-plastin as a new component of invadopodia, where it contributes to degradation and invasiveness. By means of specific, high-affinity nanobodies inhibiting bundling of fascin or L-plastin, we further unraveled their cooperative mode of action. We show that the bundlers cannot compensate for each other due to strikingly different bundling characteristics: L-plastin bundles are much thinner and less tightly packed. Composite bundles adopt an intermediate phenotype, with fascin delivering the rigidity and strength for protrusive force and structural stability, whereas L-plastin accounts for the flexibility needed for elongation. Consistent with this, elevated L-plastin expression promotes elongation and reduces protrusion density in cells with relatively lower L-plastin than fascin levels.
Collapse
Affiliation(s)
| | | | | | - Leen Pieters
- Basic Medical Science, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Maria Cornelissen
- Basic Medical Science, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | | |
Collapse
|
71
|
Eskin JA, Rankova A, Johnston AB, Alioto SL, Goode BL. Common formin-regulating sequences in Smy1 and Bud14 are required for the control of actin cable assembly in vivo. Mol Biol Cell 2016; 27:828-37. [PMID: 26764093 PMCID: PMC4803308 DOI: 10.1091/mbc.e15-09-0639] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Formins comprise a large family of proteins with diverse roles in remodeling the actin cytoskeleton. However, the spatiotemporal mechanisms used by cells to control formin activities are only beginning to be understood. Here we dissected Smy1, which has dual roles in regulating formins and myosin. Using mutagenesis, we identified specific sequences in Smy1 critical for its in vitro inhibitory effects on the FH2 domain of the formin Bnr1. By integrating smy1 alleles targeting those sequences, we genetically uncoupled Smy1's functions in regulating formins and myosin. Quantitative imaging analysis further demonstrated that the ability of Smy1 to directly control Bnr1 activity is crucial in vivo for proper actin cable length, shape, and velocity and, in turn, efficient secretory vesicle transport. A Smy1-like sequence motif was also identified in a different Bnr1 regulator, Bud14, and found to be essential for Bud14 functions in regulating actin cable architecture and function in vivo. Together these observations reveal unanticipated mechanistic ties between two distinct formin regulators. Further, they emphasize the importance of tightly controlling formin activities in vivo to generate specialized geometries and dynamics of actin structures tailored to their physiological roles.
Collapse
Affiliation(s)
- Julian A Eskin
- Department of Biology, Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Aneliya Rankova
- Department of Biology, Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Adam B Johnston
- Department of Biology, Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Salvatore L Alioto
- Department of Biology, Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| |
Collapse
|
72
|
Skau CT, Waterman CM. Specification of Architecture and Function of Actin Structures by Actin Nucleation Factors. Annu Rev Biophys 2016; 44:285-310. [PMID: 26098516 DOI: 10.1146/annurev-biophys-060414-034308] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The actin cytoskeleton is essential for diverse processes in mammalian cells; these processes range from establishing cell polarity to powering cell migration to driving cytokinesis to positioning intracellular organelles. How these many functions are carried out in a spatiotemporally regulated manner in a single cytoplasm has been the subject of much study in the cytoskeleton field. Recent work has identified a host of actin nucleation factors that can build architecturally diverse actin structures. The biochemical properties of these factors, coupled with their cellular location, likely define the functional properties of actin structures. In this article, we describe how recent advances in cell biology and biochemistry have begun to elucidate the role of individual actin nucleation factors in generating distinct cellular structures. We also consider how the localization and orientation of actin nucleation factors, in addition to their kinetic properties, are critical to their ability to build a functional actin cytoskeleton.
Collapse
Affiliation(s)
- Colleen T Skau
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892; ,
| | | |
Collapse
|
73
|
Lehtimäki J, Hakala M, Lappalainen P. Actin Filament Structures in Migrating Cells. Handb Exp Pharmacol 2016; 235:123-152. [PMID: 27469496 DOI: 10.1007/164_2016_28] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell migration is necessary for several developmental processes in multicellular organisms. Furthermore, many physiological processes such as wound healing and immunological events in adult animals are dependent on cell migration. Consequently, defects in cell migration are linked to various diseases including immunological disorders as well as cancer progression and metastasis formation. Cell migration is driven by specific protrusive and contractile actin filament structures, but the types and relative contributions of these actin filament arrays vary depending on the cell type and the environment of the cell. In this chapter, we introduce the most important actin filament structures that contribute to mesenchymal and amoeboid cell migration modes and discuss the mechanisms by which the assembly and turnover of these structures are controlled by various actin-binding proteins.
Collapse
Affiliation(s)
- Jaakko Lehtimäki
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Markku Hakala
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland.
| |
Collapse
|
74
|
Steffen A, Stradal TEB, Rottner K. Signalling Pathways Controlling Cellular Actin Organization. Handb Exp Pharmacol 2016; 235:153-178. [PMID: 27757765 DOI: 10.1007/164_2016_35] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The actin cytoskeleton is essential for morphogenesis and virtually all types of cell shape changes. Reorganization is per definition driven by continuous disassembly and re-assembly of actin filaments, controlled by major, ubiquitously operating machines. These are specifically employed by the cell to tune its activities in accordance with respective environmental conditions or to satisfy specific needs.Here we sketch some fundamental signalling pathways established to contribute to the reorganization of specific actin structures at the plasma membrane. Rho-family GTPases are at the core of these pathways, and dissection of their precise contributions to actin reorganization in different cell types and tissues will thus continue to improve our understanding of these important signalling nodes. Furthermore, we will draw your attention to the emerging theme of actin reorganization on intracellular membranes, its functional relation to Rho-GTPase signalling, and its relevance for the exciting phenomenon autophagy.
Collapse
Affiliation(s)
- Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.,Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
| |
Collapse
|
75
|
Actin-Dependent Regulation of Borrelia burgdorferi Phagocytosis by Macrophages. Curr Top Microbiol Immunol 2016; 399:133-154. [DOI: 10.1007/82_2016_26] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
76
|
Sahasrabudhe A, Ghate K, Mutalik S, Jacob A, Ghose A. Formin 2 regulates the stabilization of filopodial tip adhesions in growth cones and affects neuronal outgrowth and pathfinding in vivo. Development 2015; 143:449-60. [PMID: 26718007 DOI: 10.1242/dev.130104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/23/2015] [Indexed: 12/28/2022]
Abstract
Growth cone filopodia are actin-based mechanosensory structures that are essential for chemoreception and the generation of contractile forces necessary for directional motility. However, little is known about the influence of filopodial actin structures on substrate adhesion and filopodial contractility. Formin 2 (Fmn2) localizes along filopodial actin bundles and its depletion does not affect filopodia initiation or elongation. However, Fmn2 activity is required for filopodial tip adhesion maturation and the ability of filopodia to generate traction forces. Dysregulation of filopodia in Fmn2-depleted neurons leads to compromised growth cone motility. Additionally, in mouse fibroblasts, Fmn2 regulates ventral stress fiber assembly and affects the stability of focal adhesions. In the developing chick spinal cord, Fmn2 activity is required cell-autonomously for the outgrowth and pathfinding of spinal commissural neurons. Our results reveal an unanticipated function for Fmn2 in neural development. Fmn2 regulates structurally diverse bundled actin structures, parallel filopodial bundles in growth cones and anti-parallel stress fibers in fibroblasts, in turn modulating the stability of substrate adhesions. We propose Fmn2 as a mediator of actin bundle integrity, enabling efficient force transmission to the adhesion sites.
Collapse
Affiliation(s)
- Abhishek Sahasrabudhe
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhaba Road, Pune 411008, India
| | - Ketakee Ghate
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhaba Road, Pune 411008, India
| | - Sampada Mutalik
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhaba Road, Pune 411008, India
| | - Ajesh Jacob
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhaba Road, Pune 411008, India
| | - Aurnab Ghose
- Indian Institute of Science Education and Research (IISER) Pune, Dr Homi Bhaba Road, Pune 411008, India
| |
Collapse
|
77
|
Tsai FC, Koenderink GH. Shape control of lipid bilayer membranes by confined actin bundles. SOFT MATTER 2015; 11:8834-8847. [PMID: 26395896 DOI: 10.1039/c5sm01583a] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In living cells, lipid membranes and biopolymers determine each other's conformation in a delicate force balance. Cellular polymers such as actin filaments are strongly confined by the plasma membrane in cell protrusions such as lamellipodia and filopodia. Conversely, protrusion formation is facilitated by actin-driven membrane deformation and these protrusions are maintained by dense actin networks or bundles of actin filaments. Here we investigate the mechanical interplay between actin bundles and lipid bilayer membranes by reconstituting a minimal model system based on cell-sized liposomes with encapsulated actin filaments bundled by fascin. To address the competition between the deformability of the membrane and the enclosed actin bundles, we tune the bundle stiffness (through the fascin-to-actin molar ratio) and the membrane rigidity (through protein decoration). Using confocal microscopy and quantitative image analysis, we show that actin bundles deform the liposomes into a rich set of morphologies. For liposomes having a small membrane bending rigidity, the actin bundles tend to generate finger-like membrane protrusions that resemble cellular filopodia. Stiffer bundles formed at high crosslink density stay straight in the liposome body, whereas softer bundles formed at low crosslink density are bent and kinked. When the membrane has a large bending rigidity, membrane protrusions are suppressed. In this case, membrane enclosure forces the actin bundles to organize into cortical rings, to minimize the energy cost associated with filament bending. Our results highlight the importance of taking into account mechanical interactions between the actin cytoskeleton and the membrane to understand cell shape control.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- FOM Institute AMOLF, Systems Biophysics Department, Science Park 104, 1098 XG Amsterdam, The Netherlands.
| | - Gijsje Hendrika Koenderink
- FOM Institute AMOLF, Systems Biophysics Department, Science Park 104, 1098 XG Amsterdam, The Netherlands.
| |
Collapse
|
78
|
Wang X, Shi L, Deng Y, Qu M, Mao S, Xu L, Xu W, Fang C. Inhibition of leucine aminopeptidase 3 suppresses invasion of ovarian cancer cells through down-regulation of fascin and MMP-2/9. Eur J Pharmacol 2015; 768:116-22. [PMID: 26526349 DOI: 10.1016/j.ejphar.2015.10.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/19/2015] [Accepted: 10/21/2015] [Indexed: 02/06/2023]
Abstract
Leucine aminopeptidase 3 (LAP3) is a cell surface aminopeptidase that catalyzes the hydrolysis of leucine residues from the amino termini of protein or peptide substrates. The over-expression of LAP3 correlates with prognosis and malignant development of several human cell carcinomas. However, the molecular mechanism remains unknown. In this study, we used ES-2 ovarian cancer cell line as a model system to explore the role of LAP3 in regulation of cancer cell invasion by employing a natural LAP3 inhibitor bestatin and LAP3 siRNA. Bestatin inhibited tumor cell migration and invasion in a dose-dependent manner. More interestingly, bestatin down-regulated expression of fascin protein and inhibited activity of fascin promoter luciferase reporter. Both proteome profiler array and Western blot assay showed that bestatin up-regulated the phosphorylation of Hsp27. Furthermore, LAP3 siRNA could up-regulate the phosphorylation of Hsp27 and down-regulate the expression of fascin. Meanwhile, LAP3 siRNA could also down-regulate the phosphorylation of Akt and the expression of MMP-2/9. Taken together, LAP3 could affect the expression of fascin and MMP-2/9 and may act as a potential anti-metastasis therapeutic target.
Collapse
Affiliation(s)
- Xuejian Wang
- Key Laboratory of Applied Pharmacology in Universities of Sh andong, Department of Pharmacology, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China
| | - Lihong Shi
- Key Laboratory of Applied Pharmacology in Universities of Sh andong, Department of Pharmacology, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China
| | - Yilin Deng
- Tender Office, Wei Fang People's Hospital, Weifang 261041, Shandong, China
| | - Meihua Qu
- Key Laboratory of Applied Pharmacology in Universities of Sh andong, Department of Pharmacology, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China
| | - Shumei Mao
- Key Laboratory of Applied Pharmacology in Universities of Sh andong, Department of Pharmacology, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China
| | - Liyan Xu
- Institute of Oncologic Pathology, The Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wenfang Xu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Chunyan Fang
- Key Laboratory of Applied Pharmacology in Universities of Sh andong, Department of Pharmacology, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
79
|
Ma Y, Machesky LM. Fascin1 in carcinomas: Its regulation and prognostic value. Int J Cancer 2015; 137:2534-44. [PMID: 25302416 DOI: 10.1002/ijc.29260] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/01/2014] [Indexed: 01/06/2023]
Abstract
Previous cell biological studies demonstrate that the actin bundling protein fascin1 regulates cell motility, migration and invasion. Human studies demonstrate that fascin1 is upregulated in many epithelial cancers. This review gives a brief overview of the role of fascin1 in cell migration and invasion, but focuses mainly on the regulation and clinical relevance of fascin1 in epithelial cancers. Here, we propose fascin1 as a potent prognostic biomarker for breast, colorectal, esophageal cancers and head and neck squamous cell carcinomas. Fascin1 may also be an attractive drug target against these carcinomas in the future, but more studies are needed.
Collapse
Affiliation(s)
- Yafeng Ma
- School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Medical Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, NSW2170, New South Wales, Australia
| | - Laura M Machesky
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, Scotland, United Kingdom
| |
Collapse
|
80
|
Single-molecule visualization of a formin-capping protein 'decision complex' at the actin filament barbed end. Nat Commun 2015; 6:8707. [PMID: 26566078 PMCID: PMC4660045 DOI: 10.1038/ncomms9707] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/22/2015] [Indexed: 01/01/2023] Open
Abstract
Precise control of actin filament length is essential to many cellular processes. Formins processively elongate filaments, whereas capping protein (CP) binds to barbed ends and arrests polymerization. While genetic and biochemical evidence has indicated that these two proteins function antagonistically, the mechanism underlying the antagonism has remained unresolved. Here we use multi-wavelength single-molecule fluorescence microscopy to observe the fully reversible formation of a long-lived 'decision complex' in which a CP dimer and a dimer of the formin mDia1 simultaneously bind the barbed end. Further, mDia1 displaced from the barbed end by CP can randomly slide along the filament and later return to the barbed end to re-form the complex. Quantitative kinetic analysis reveals that the CP-mDia1 antagonism that we observe in vitro occurs through the decision complex. Our observations suggest new molecular mechanisms for the control of actin filament length and for the capture of filament barbed ends in cells.
Collapse
|
81
|
Young LE, Heimsath EG, Higgs HN. Cell type-dependent mechanisms for formin-mediated assembly of filopodia. Mol Biol Cell 2015; 26:4646-59. [PMID: 26446836 PMCID: PMC4678021 DOI: 10.1091/mbc.e15-09-0626] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/01/2015] [Indexed: 11/11/2022] Open
Abstract
Filopodia are finger-like protrusions from the plasma membrane and are of fundamental importance to cellular physiology, but the mechanisms governing their assembly are still in question. One model, called convergent elongation, proposes that filopodia arise from Arp2/3 complex-nucleated dendritic actin networks, with factors such as formins elongating these filaments into filopodia. We test this model using constitutively active constructs of two formins, FMNL3 and mDia2. Surprisingly, filopodial assembly requirements differ between suspension and adherent cells. In suspension cells, Arp2/3 complex is required for filopodial assembly through either formin. In contrast, a subset of filopodia remains after Arp2/3 complex inhibition in adherent cells. In adherent cells only, mDia1 and VASP also contribute to filopodial assembly, and filopodia are disproportionately associated with focal adhesions. We propose an extension of the existing models for filopodial assembly in which any cluster of actin filament barbed ends in proximity to the plasma membrane, either Arp2/3 complex dependent or independent, can initiate filopodial assembly by specific formins.
Collapse
Affiliation(s)
- Lorna E Young
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Ernest G Heimsath
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Henry N Higgs
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
82
|
Sun BO, Fang Y, Li Z, Chen Z, Xiang J. Role of cellular cytoskeleton in epithelial-mesenchymal transition process during cancer progression. Biomed Rep 2015; 3:603-610. [PMID: 26405532 DOI: 10.3892/br.2015.494] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023] Open
Abstract
Currently, cancer metastases remain a major clinical problem that highlights the importance of recognition of the metastatic process in cancer diagnosis and treatment. A critical process associated with the metastasis process is the transformation of epithelial cells toward the motile mesenchymal state, a process called epithelial-mesenchymal transition (EMT). Increasing evidence suggests the crucial role of the cytoskeleton in the EMT process. The cytoskeleton is composed of the actin cytoskeleton, the microtubule network and the intermediate filaments that provide structural design and mechanical strength that is necessary for the EMT. The dynamic reorganization of the actin cytoskeleton is a prerequisite for the morphology, migration and invasion of cancer cells. The microtubule network is the cytoskeleton that provides the driving force during cell migration. Intermediate filaments are significantly rearranged, typically switching from cytokeratin-rich to vimentin-rich networks during the EMT process, accompanied by a greatly enhanced cell motility capacity. In the present review, the recent novel insights into the different cytoskeleton underlying EMT are summarized. There are numerous advances in our understanding of the fundamental role of the cytoskeleton in cancer cell invasion and migration.
Collapse
Affiliation(s)
- B O Sun
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yantian Fang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhenyang Li
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zongyou Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jianbin Xiang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
83
|
Jansen S, Collins A, Chin SM, Ydenberg CA, Gelles J, Goode BL. Single-molecule imaging of a three-component ordered actin disassembly mechanism. Nat Commun 2015; 6:7202. [PMID: 25995115 PMCID: PMC4443854 DOI: 10.1038/ncomms8202] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 04/17/2015] [Indexed: 12/25/2022] Open
Abstract
The mechanisms by which cells destabilize and rapidly disassemble filamentous actin networks have remained elusive; however, Coronin, Cofilin and AIP1 have been implicated in this process. Here using multi-wavelength single-molecule fluorescence imaging, we show that mammalian Cor1B, Cof1 and AIP1 work in concert through a temporally ordered pathway to induce highly efficient severing and disassembly of actin filaments. Cor1B binds to filaments first, and dramatically accelerates the subsequent binding of Cof1, leading to heavily decorated, stabilized filaments. Cof1 in turn recruits AIP1, which rapidly triggers severing and remains bound to the newly generated barbed ends. New growth at barbed ends generated by severing was blocked specifically in the presence of all three proteins. This activity enabled us to reconstitute and directly visualize single actin filaments being rapidly polymerized by formins at their barbed ends while simultanteously being stochastically severed and capped along their lengths, and disassembled from their pointed ends. The roles of Coronin, Cofilin and AIP1 in promoting actin disassembly have not been well understood. Here using single-molecule fluorescence imaging, Jansen et al. show that the three proteins act together in a coordinated, temporal pathway to induce rapid severing and disassembly of actin filaments.
Collapse
Affiliation(s)
- Silvia Jansen
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| | - Agnieszka Collins
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| | - Samantha M Chin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| | - Casey A Ydenberg
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| | - Jeff Gelles
- Department of Biochemistry, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South street, Waltham, Massachusetts 02454, USA
| |
Collapse
|
84
|
Bosch PJ, Corrêa IR, Sonntag MH, Ibach J, Brunsveld L, Kanger JS, Subramaniam V. Evaluation of fluorophores to label SNAP-tag fused proteins for multicolor single-molecule tracking microscopy in live cells. Biophys J 2015; 107:803-14. [PMID: 25140415 DOI: 10.1016/j.bpj.2014.06.040] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/22/2014] [Accepted: 06/10/2014] [Indexed: 11/19/2022] Open
Abstract
Single-molecule tracking has become a widely used technique for studying protein dynamics and their organization in the complex environment of the cell. In particular, the spatiotemporal distribution of membrane receptors is an active field of study due to its putative role in the regulation of signal transduction. The SNAP-tag is an intrinsically monovalent and highly specific genetic tag for attaching a fluorescent label to a protein of interest. Little information is currently available on the choice of optimal fluorescent dyes for single-molecule microscopy utilizing the SNAP-tag labeling system. We surveyed 6 green and 16 red excitable dyes for their suitability in single-molecule microscopy of SNAP-tag fusion proteins in live cells. We determined the nonspecific binding levels and photostability of these dye conjugates when bound to a SNAP-tag fused membrane protein in live cells. We found that only a limited subset of the dyes tested is suitable for single-molecule tracking microscopy. The results show that a careful choice of the dye to conjugate to the SNAP-substrate to label SNAP-tag fusion proteins is very important, as many dyes suffer from either rapid photobleaching or high nonspecific staining. These characteristics appear to be unpredictable, which motivated the need to perform the systematic survey presented here. We have developed a protocol for evaluating the best dyes, and for the conditions that we evaluated, we find that Dy 549 and CF 640 are the best choices tested for single-molecule tracking. Using an optimal dye pair, we also demonstrate the possibility of dual-color single-molecule imaging of SNAP-tag fusion proteins. This survey provides an overview of the photophysical and imaging properties of a range of SNAP-tag fluorescent substrates, enabling the selection of optimal dyes and conditions for single-molecule imaging of SNAP-tagged fusion proteins in eukaryotic cell lines.
Collapse
Affiliation(s)
- Peter J Bosch
- Nanobiophysics, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | - Michael H Sonntag
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jenny Ibach
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Johannes S Kanger
- Nanobiophysics, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Vinod Subramaniam
- Nanobiophysics, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
85
|
Wagh D, Terry-Lorenzo R, Waites CL, Leal-Ortiz SA, Maas C, Reimer RJ, Garner CC. Piccolo Directs Activity Dependent F-Actin Assembly from Presynaptic Active Zones via Daam1. PLoS One 2015; 10:e0120093. [PMID: 25897839 PMCID: PMC4405365 DOI: 10.1371/journal.pone.0120093] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/23/2015] [Indexed: 12/12/2022] Open
Abstract
The dynamic assembly of filamentous (F) actin plays essential roles in the assembly of presynaptic boutons, the fusion, mobilization and recycling of synaptic vesicles (SVs), and presynaptic forms of plasticity. However, the molecular mechanisms that regulate the temporal and spatial assembly of presynaptic F-actin remain largely unknown. Similar to other F-actin rich membrane specializations, presynaptic boutons contain a set of molecules that respond to cellular cues and trans-synaptic signals to facilitate activity-dependent assembly of F-actin. The presynaptic active zone (AZ) protein Piccolo has recently been identified as a key regulator of neurotransmitter release during SV cycling. It does so by coordinating the activity-dependent assembly of F-Actin and the dynamics of key plasticity molecules including Synapsin1, Profilin and CaMKII. The multidomain structure of Piccolo, its exquisite association with the AZ, and its ability to interact with a number of actin-associated proteins suggest that Piccolo may function as a platform to coordinate the spatial assembly of F-actin. Here we have identified Daam1, a Formin that functions with Profilin to drive F-actin assembly, as a novel Piccolo binding partner. We also found that within cells Daam1 activation promotes Piccolo binding, an interaction that can spatially direct the polymerization of F-Actin. Moreover, similar to Piccolo and Profilin, Daam1 loss of function impairs presynaptic-F-actin assembly in neurons. These data suggest a model in which Piccolo directs the assembly of presynaptic F-Actin from the AZ by scaffolding key actin regulatory proteins including Daam1.
Collapse
Affiliation(s)
- Dhananjay Wagh
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, California, United States of America
| | - Ryan Terry-Lorenzo
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, California, United States of America
| | - Clarissa L. Waites
- Department of Pathology and Cell Biology Columbia University New York, New York, United States of America
| | - Sergio A. Leal-Ortiz
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, California, United States of America
| | - Christoph Maas
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, California, United States of America
| | - Richard J. Reimer
- Department of Neurology and Neurological Sciences Stanford University and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Craig C. Garner
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
86
|
Groen CM, Jayo A, Parsons M, Tootle TL. Prostaglandins regulate nuclear localization of Fascin and its function in nucleolar architecture. Mol Biol Cell 2015; 26:1901-17. [PMID: 25808493 PMCID: PMC4436834 DOI: 10.1091/mbc.e14-09-1384] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/18/2015] [Indexed: 01/14/2023] Open
Abstract
Fascin, a conserved actin-bundling protein, is not only cytoplasmic but also localizes to the nucleus and nuclear periphery in both Drosophila and mammalian cell contexts. In Drosophila, prostaglandin signaling regulates this localization. In addition, Fascin plays a critical role in nucleolar architecture in both Drosophila and mammalian cells. Fascin, a highly conserved actin-bundling protein, localizes and functions at new cellular sites in both Drosophila and multiple mammalian cell types. During Drosophila follicle development, in addition to being cytoplasmic, Fascin is in the nuclei of the germline-derived nurse cells during stages 10B–12 (S10B–12) and at the nuclear periphery during stage 13 (S13). This localization is specific to Fascin, as other actin-binding proteins, Villin and Profilin, do not exhibit the same subcellular distribution. In addition, localization of fascin1 to the nucleus and nuclear periphery is observed in multiple mammalian cell types. Thus the regulation and function of Fascin at these new cellular locations is likely to be highly conserved. In Drosophila, loss of prostaglandin signaling causes a global reduction in nuclear Fascin and a failure to relocalize to the nuclear periphery. Alterations in nuclear Fascin levels result in defects in nucleolar morphology in both Drosophila follicles and cultured mammalian cells, suggesting that nuclear Fascin plays an important role in nucleolar architecture. Given the numerous roles of Fascin in development and disease, including cancer, our novel finding that Fascin has functions within the nucleus sheds new light on the potential roles of Fascin in these contexts.
Collapse
Affiliation(s)
- Christopher M Groen
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Asier Jayo
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, United Kingdom
| | - Tina L Tootle
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
87
|
Scherer A, Kuhl S, Wessels D, Lusche DF, Hanson B, Ambrose J, Voss E, Fletcher E, Goldman C, Soll DR. A computer-assisted 3D model for analyzing the aggregation of tumorigenic cells reveals specialized behaviors and unique cell types that facilitate aggregate coalescence. PLoS One 2015; 10:e0118628. [PMID: 25790299 PMCID: PMC4366230 DOI: 10.1371/journal.pone.0118628] [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: 10/16/2014] [Accepted: 11/28/2014] [Indexed: 01/11/2023] Open
Abstract
We have developed a 4D computer-assisted reconstruction and motion analysis system, J3D-DIAS 4.1, and applied it to the reconstruction and motion analysis of tumorigenic cells in a 3D matrix. The system is unique in that it is fast, high-resolution, acquires optical sections using DIC microscopy (hence there is no associated photoxicity), and is capable of long-term 4D reconstruction. Specifically, a z-series at 5 μm increments can be acquired in less than a minute on tissue samples embedded in a 1.5 mm thick 3D Matrigel matrix. Reconstruction can be repeated at intervals as short as every minute and continued for 30 days or longer. Images are converted to mathematical representations from which quantitative parameters can be derived. Application of this system to cancer cells from established lines and fresh tumor tissue has revealed unique behaviors and cell types not present in non-tumorigenic lines. We report here that cells from tumorigenic lines and tumors undergo rapid coalescence in 3D, mediated by specific cell types that we have named “facilitators” and “probes.” A third cell type, the “dervish”, is capable of rapid movement through the gel and does not adhere to it. These cell types have never before been described. Our data suggest that tumorigenesis in vitro is a developmental process involving coalescence facilitated by specialized cells that culminates in large hollow spheres with complex architecture. The unique effects of select monoclonal antibodies on these processes demonstrate the usefulness of the model for analyzing the mechanisms of anti-cancer drugs.
Collapse
Affiliation(s)
- Amanda Scherer
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Spencer Kuhl
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Deborah Wessels
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Daniel F. Lusche
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Brett Hanson
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Joseph Ambrose
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Edward Voss
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Emily Fletcher
- Mercy Hospital System of Des Moines, Des Moines, Iowa, United States of America
| | - Charles Goldman
- Mercy Hospital System of Des Moines, Des Moines, Iowa, United States of America
| | - David R. Soll
- Monoclonal Antibody Research Institute, Developmental Studies Hybridoma Bank, Department of Biology, University of Iowa, Iowa City, Iowa, 52242, United States of America
- * E-mail:
| |
Collapse
|
88
|
Gungor-Ordueri NE, Cheng CY. Fascin - An actin binding and bundling protein in the testis and its role in ectoplasmic specialization dynamics. SPERMATOGENESIS 2015; 5:e1002733. [PMID: 26413410 DOI: 10.1080/21565562.2014.1002733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/19/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022]
Abstract
In the mammalian testis such as in rats, a unique actin-rich cell-cell adherens junction (AJ) known as ectoplasmic specialization (ES) is found in the seminiferous epithelium. ES is conspicuously found between Sertoli cells near the basement membrane known as the basal ES, which together with tight junction (TJ), gap junction, and desmosome constitute the blood-testis barrier (BTB). The BTB, in turn, anatomically divides the seminiferous epithelium into the basal and the adluminal (apical) compartment. On the other hand, ES is also found at the Sertoli-spermatid interface known as apical ES which is the only anchoring device for developing step 8-19 spermatids during spermiogenesis. One of the most typical features of the ES is the array of actin microfilament bundles that lie perpendicular to the Sertoli cell plasma membrane and are sandwiched in-between the cisternae of endoplasmic reticulum and the Sertoli cell plasma membrane. While these actin filament bundles confer the adhesive strength of Sertoli cells at the BTB and also spermatids in the adluminal compartment, they must be rapidly re-organized from their bundled to unbundled/branched configuration and vice versa to provide plasticity to the ES so that preleptotene spermatocytes and spermatids can be transported across the immunological barrier and the adluminal compartment, respectively, during the epithelial cycle of spermatogenesis. Fascin is a family of actin microfilament cross-linking and bundling proteins that is known to confer bundling of parallel actin microfilaments in mammalian cells. A recent report has illustrated the significance of a fascin protein called fascin 1 in actin microfilaments at the ES, pertinent to its role in spermatogenesis (Gungor-Ordueri et al. Am J Physiol Endocrinol Metab 307, E738-753, 2004 (DOI:10.1152/ajpendo.00113.2014). In this Commentary, we critically evaluate these findings in light of the role of fascin in other mammalian cells, providing some insightful information for future investigations.
Collapse
Affiliation(s)
- N Ece Gungor-Ordueri
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| |
Collapse
|
89
|
Fibroblasts probe substrate rigidity with filopodia extensions before occupying an area. Proc Natl Acad Sci U S A 2014; 111:17176-81. [PMID: 25404288 DOI: 10.1073/pnas.1412285111] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rigidity sensing and durotaxis are thought to be important elements in wound healing, tissue formation, and cancer treatment. It has been challenging, however, to study the underlying mechanism due to difficulties in capturing cells during the transient response to a rigidity interface. We have addressed this problem by developing a model experimental system that confines cells to a micropatterned area with a rigidity border. The system consists of a rigid domain of one large adhesive island, adjacent to a soft domain of small adhesive islands grafted on a nonadhesive soft gel. This configuration allowed us to test rigidity sensing away from the cell body during probing and spreading. NIH 3T3 cells responded to the micropatterned rigidity border similarly to cells at a conventional rigidity border, by showing a strong preference for staying on the rigid side. Furthermore, cells used filopodia extensions to probe substrate rigidity at a distance in front of the leading edge and regulated their responses based on the strain of the intervening substrate. Soft substrates inhibited focal adhesion maturation and promoted cell retraction, whereas rigid substrates allowed stable adhesions and cell spreading. Myosin II was required for not only the generation of probing forces but also the retraction in response to soft substrates. We suggest that a myosin II-driven, filopodia-based probing mechanism ahead of the leading edge allows cells to migrate efficiently, by sensing physical characteristics before moving over a substrate to avoid backtracking.
Collapse
|
90
|
Adams JC. Fascin-1 as a biomarker and prospective therapeutic target in colorectal cancer. Expert Rev Mol Diagn 2014; 15:41-8. [DOI: 10.1586/14737159.2015.976557] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
91
|
Cellular control of cortical actin nucleation. Curr Biol 2014; 24:1628-1635. [PMID: 25017211 PMCID: PMC4110400 DOI: 10.1016/j.cub.2014.05.069] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/02/2014] [Accepted: 05/28/2014] [Indexed: 11/20/2022]
Abstract
The contractile actin cortex is a thin layer of actin, myosin, and actin-binding proteins that subtends the membrane of animal cells. The cortex is the main determinant of cell shape and plays a fundamental role in cell division [1-3], migration [4], and tissue morphogenesis [5]. For example, cortex contractility plays a crucial role in amoeboid migration of metastatic cells [6] and during division, where its misregulation can lead to aneuploidy [7]. Despite its importance, our knowledge of the cortex is poor, and even the proteins nucleating it remain unknown, though a number of candidates have been proposed based on indirect evidence [8-15]. Here, we used two independent approaches to identify cortical actin nucleators: a proteomic analysis using cortex-rich isolated blebs, and a localization/small hairpin RNA (shRNA) screen searching for phenotypes with a weakened cortex or altered contractility. This unbiased study revealed that two proteins generated the majority of cortical actin: the formin mDia1 and the Arp2/3 complex. Each nucleator contributed a similar amount of F-actin to the cortex but had very different accumulation kinetics. Electron microscopy examination revealed that each nucleator affected cortical network architecture differently. mDia1 depletion led to failure in division, but Arp2/3 depletion did not. Interestingly, despite not affecting division on its own, Arp2/3 inhibition potentiated the effect of mDia1 depletion. Our findings indicate that the bulk of the actin cortex is nucleated by mDia1 and Arp2/3 and suggest a mechanism for rapid fine-tuning of cortex structure and mechanics by adjusting the relative contribution of each nucleator.
Collapse
|
92
|
Kozlov MM, Campelo F, Liska N, Chernomordik LV, Marrink SJ, McMahon HT. Mechanisms shaping cell membranes. Curr Opin Cell Biol 2014; 29:53-60. [PMID: 24747171 DOI: 10.1016/j.ceb.2014.03.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 01/08/2023]
Abstract
Membranes of intracellular organelles are characterized by large curvatures with radii of the order of 10-30nm. While, generally, membrane curvature can be a consequence of any asymmetry between the membrane monolayers, generation of large curvatures requires the action of mechanisms based on specialized proteins. Here we discuss the three most relevant classes of such mechanisms with emphasis on the physical requirements for proteins to be effective in generation of membrane curvature. We provide new quantitative estimates of membrane bending by shallow hydrophobic insertions and compare the efficiency of the insertion mechanism with those of the protein scaffolding and crowding mechanisms.
Collapse
Affiliation(s)
- Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel.
| | - Felix Campelo
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Nicole Liska
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Leonid V Chernomordik
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Harvey T McMahon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| |
Collapse
|
93
|
Hoffmann AK, Naj X, Linder S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages. FASEB J 2014; 28:3075-89. [PMID: 24696301 DOI: 10.1096/fj.13-247049] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease, an infectious disease that primarily affects the skin, nervous system, and joints. Uptake of borreliae by immune cells is decisive for the course of the infection, and remodelling of the host actin cytoskeleton is crucial in this process. In this study, we showed that the actin-regulatory formin Daam1 is important in Borrelia phagocytosis by primary human macrophages. Uptake of borreliae proceeds preferentially through capture by filopodia and formation of coiling pseudopods that enwrap the spirochetes. Using immunofluorescence, we localized endogenous and overexpressed Daam1 to filopodia and to F-actin-rich uptake structures. Live-cell imaging further showed that Daam1 is enriched at coiling pseudopods that arise from the macrophage surface. This filopodia-independent step was corroborated by control experiments of phagocytic cup formation with latex beads. Moreover, siRNA-mediated knockdown of Daam1 led to a 65% reduction of borreliae-induced filopodia, and, as shown by the outside-inside staining technique, to a 50% decrease in phagocytic uptake of borreliae, as well as a 37% reduction in coiling pseudopod formation. Collectively, we showed that Daam1 plays a dual role in the phagocytic uptake of borreliae: first, as a regulator of filopodia, which are used for capturing spirochetes, and second, in the formation of the coiling pseudopod that enwraps the bacterial cell. These data identify Daam1 as a novel regulator of B. burgdorferi phagocytosis. At the same time, this is the first demonstration of a role for Daam1 in phagocytic processes in general.-Hoffmann, A.-K., Naj, X., Linder, S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages.
Collapse
Affiliation(s)
- Ann-Kathrin Hoffmann
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Xenia Naj
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
94
|
Ena/VASP Enabled is a highly processive actin polymerase tailored to self-assemble parallel-bundled F-actin networks with Fascin. Proc Natl Acad Sci U S A 2014; 111:4121-6. [PMID: 24591594 DOI: 10.1073/pnas.1322093111] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Filopodia are exploratory finger-like projections composed of multiple long, straight, parallel-bundled actin filaments that protrude from the leading edge of migrating cells. Drosophila melanogaster Enabled (Ena) is a member of the Ena/vasodilator-stimulated phosphoprotein protein family, which facilitates the assembly of filopodial actin filaments that are bundled by Fascin. However, the mechanism by which Ena and Fascin promote the assembly of uniformly thick F-actin bundles that are capable of producing coordinated protrusive forces without buckling is not well understood. We used multicolor evanescent wave fluorescence microscopy imaging to follow individual Ena molecules on both single and Fascin-bundled F-actin in vitro. Individual Ena tetramers increase the elongation rate approximately two- to threefold and inhibit capping protein by remaining processively associated with the barbed end for an average of ∼10 s in solution, for ∼60 s when immobilized on a surface, and for ∼110 s when multiple Ena tetramers are clustered on a surface. Ena also can gather and simultaneously elongate multiple barbed ends. Collectively, these properties could facilitate the recruitment of Fascin and initiate filopodia formation. Remarkably, we found that Ena's actin-assembly properties are tunable on Fascin-bundled filaments, facilitating the formation of filopodia-like F-actin networks without tapered barbed ends. Ena-associated trailing barbed ends in Fascin-bundled actin filaments have approximately twofold more frequent and approximately fivefold longer processive runs, allowing them to catch up with leading barbed ends efficiently. Therefore, Fascin and Ena cooperate to extend and maintain robust filopodia of uniform thickness with aligned barbed ends by a unique mechanistic cycle.
Collapse
|
95
|
Rosado M, Barber CF, Berciu C, Feldman S, Birren SJ, Nicastro D, Goode BL. Critical roles for multiple formins during cardiac myofibril development and repair. Mol Biol Cell 2014; 25:811-27. [PMID: 24430873 PMCID: PMC3952851 DOI: 10.1091/mbc.e13-08-0443] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/13/2013] [Accepted: 01/09/2014] [Indexed: 12/31/2022] Open
Abstract
Cardiac and skeletal muscle function depends on the proper formation of myofibrils, which are tandem arrays of highly organized actomyosin contractile units called sarcomeres. How the architecture of these colossal molecular assemblages is established during development and maintained over the lifetime of an animal is poorly understood. We investigate the potential roles in myofibril formation and repair of formin proteins, which are encoded by 15 different genes in mammals. Using quantitative real-time PCR analysis, we find that 13 formins are differentially expressed in mouse hearts during postnatal development. Seven formins immunolocalize to sarcomeres in diverse patterns, suggesting that they have a variety of functional roles. Using RNA interference silencing, we find that the formins mDia2, DAAM1, FMNL1, and FMNL2 are required nonredundantly for myofibrillogenesis. Knockdown phenotypes include global loss of myofibril organization and defective sarcomeric ultrastructure. Finally, our analysis reveals an unanticipated requirement specifically for FMNL1 and FMNL2 in the repair of damaged myofibrils. Together our data reveal an unexpectedly large number of formins, with diverse localization patterns and nonredundant roles, functioning in myofibril development and maintenance, and provide the first evidence of actin assembly factors being required to repair myofibrils.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Animals, Newborn
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Differentiation
- Formins
- Gene Expression Regulation, Developmental
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/antagonists & inhibitors
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Microtubule-Associated Proteins/antagonists & inhibitors
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Muscle Development/genetics
- Myocardium/cytology
- Myocardium/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- NADPH Dehydrogenase/antagonists & inhibitors
- NADPH Dehydrogenase/genetics
- NADPH Dehydrogenase/metabolism
- Primary Cell Culture
- Protein Isoforms/antagonists & inhibitors
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Sarcomeres/metabolism
- Sarcomeres/ultrastructure
- Thiazolidines/pharmacology
- Wound Healing/genetics
- rho GTP-Binding Proteins/antagonists & inhibitors
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
| | | | - Cristina Berciu
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Steven Feldman
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Susan J. Birren
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Daniela Nicastro
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Bruce L. Goode
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| |
Collapse
|
96
|
Winterhoff M, Junemann A, Nordholz B, Linkner J, Schleicher M, Faix J. The Diaphanous-related formin dDia1 is required for highly directional phototaxis and formation of properly sized fruiting bodies in Dictyostelium. Eur J Cell Biol 2013; 93:212-24. [PMID: 24331584 DOI: 10.1016/j.ejcb.2013.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/12/2013] [Accepted: 11/14/2013] [Indexed: 11/17/2022] Open
Abstract
Diaphanous-related formins (DRFs) act as downstream effectors of Rho family GTPases and drive the formation and elongation of linear actin filaments in various cellular processes. Here we analyzed the DRF dDia1 from Dictyostelium cells. The biochemical characterization of recombinant dDia1-FH1FH2 by bulk polymerization assays and single filament TIRF microscopy revealed that dDia1 is a rather weak nucleator. Addition of any of the three Dictyostelium profilin isoforms, however, markedly accelerated formin-mediated actin filament barbed end elongation in TIRF assays. Interestingly, filament elongation was significantly faster in presence of DdPFN I (profilin I) when compared to the other two isoforms, suggesting selectivity of dDia1 for DdPFN I. Additionally, we frequently observed dissociation of the formin from growing barbed ends. These findings are consistent with dilution-induced depolymerization assays in presence of dDia1-FH1FH2 showing that dDia1 is a weak capper in comparison with heterodimeric capping protein. To study the physiological role of this formin, we created cell lines lacking dDia1 or overexpressing GFP-tagged dDia1. Of note, constitutively active dDia1 accumulated homogenously in the entire pseudopod suggesting that it controls microfilament architecture to regulate cell migration. Comparison of wild type and dDia1-null cells in random cell migration and chemotaxis toward a cAMP gradient revealed no major differences. By contrast, phototaxis of dDia1-deficient cells during the multicellular stage was markedly impaired. While wild type slugs moved with high directionality toward the light source, the trails of dDia1-null slugs displayed a characteristic V-shaped profile and deviated in angles between 50° and 60° from the path of the incident light. Possibly in conjunction with this defect, dDia1-null cells also formed substantially smaller fruiting bodies. These findings demonstrate dDia1 to be critically involved in collective cell migration during terminal differentiation.
Collapse
Affiliation(s)
- Moritz Winterhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - Alexander Junemann
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - Benjamin Nordholz
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - Jörn Linkner
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany
| | - Michael Schleicher
- Institute for Anatomy and Cell Biology, Ludwig-Maximilians-University, 80336 München, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg Straße 1, 30625 Hannover, Germany.
| |
Collapse
|