1
|
Thanh DD, Bich-Ngoc N, Paques C, Christian A, Herkenne S, Struman I, Muller M. The food dye Tartrazine disrupts vascular formation both in zebrafish larvae and in human primary endothelial cells. Sci Rep 2024; 14:30367. [PMID: 39639097 PMCID: PMC11621646 DOI: 10.1038/s41598-024-82076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Tartrazine (E102) is a controversial coloring agent whose potential impacts on human health are not fully understood. Our study reveals the vascular disrupting effects of tartrazine (TTZ) on developing zebrafish embryos in vivo and on human umbilical vein endothelial cells in vitro. The dye was shown to cause dose-dependent hemorrhages in zebrafish embryos. Analyzing transgenic zebrafish harboring fluorescent endothelial cells revealed that TTZ treatment disrupted cell organization into vessels in both the sub-intestinal vein and the brain area. Assays on human umbilical vein endothelial cells demonstrated that TTZ inhibited endothelial proliferation, tube formation, and migration in a dose-dependent manner. Taken together, our results indicate for the first time that TTZ can affect endothelial cell properties, possibly by disrupting Rho family GTPase pathways which control the cytoskeleton. Our finding provides a credible explanation for many reported human health impacts and offers prospective applications for biomedicine.
Collapse
Affiliation(s)
- Dinh Duy Thanh
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
- Department of Cell Biology, Faculty of Biology, VNU University of Science, Hanoi, 100000, Vietnam
| | - Nguyen Bich-Ngoc
- VNU School of Interdisciplinary Sciences and Arts, Vietnam National University, Hanoi, 100000, Vietnam
| | - Cécile Paques
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Aurélie Christian
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Stéphanie Herkenne
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Ingrid Struman
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Marc Muller
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium.
| |
Collapse
|
2
|
Yadunandanan Nair N, Samuel V, Ramesh L, Marib A, David DT, Sundararaman A. Actin cytoskeleton in angiogenesis. Biol Open 2022; 11:bio058899. [PMID: 36444960 PMCID: PMC9729668 DOI: 10.1242/bio.058899] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Actin, one of the most abundant intracellular proteins in mammalian cells, is a critical regulator of cell shape and polarity, migration, cell division, and transcriptional response. Angiogenesis, or the formation of new blood vessels in the body is a well-coordinated multi-step process. Endothelial cells lining the blood vessels acquire several new properties such as front-rear polarity, invasiveness, rapid proliferation and motility during angiogenesis. This is achieved by changes in the regulation of the actin cytoskeleton. Actin remodelling underlies the switch between the quiescent and angiogenic state of the endothelium. Actin forms endothelium-specific structures that support uniquely endothelial functions. Actin regulators at endothelial cell-cell junctions maintain the integrity of the blood-tissue barrier while permitting trans-endothelial leukocyte migration. This review focuses on endothelial actin structures and less-recognised actin-mediated endothelial functions. Readers are referred to other recent reviews for the well-recognised roles of actin in endothelial motility, barrier functions and leukocyte transmigration. Actin generates forces that are transmitted to the extracellular matrix resulting in vascular matrix remodelling. In this review, we attempt to synthesize our current understanding of the roles of actin in vascular morphogenesis. We speculate on the vascular bed specific differences in endothelial actin regulation and its role in the vast heterogeneity in endothelial morphology and function across the various tissues of our body.
Collapse
Affiliation(s)
- Nidhi Yadunandanan Nair
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Victor Samuel
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Lariza Ramesh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Areeba Marib
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Deena T. David
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| |
Collapse
|
3
|
Zhang Q, Nie H, Pan J, Xu H, Zhan Q. FMNL3 is Overexpressed in Tumor Tissues and Predicts an Immuno-Hot Phenotype in Pancreatic Cancer. Int J Gen Med 2022; 15:8285-8298. [DOI: 10.2147/ijgm.s384195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
|
4
|
Ahangar P, Cowin AJ. Reforming the Barrier: The Role of Formins in Wound Repair. Cells 2022; 11:cells11182779. [PMID: 36139355 PMCID: PMC9496773 DOI: 10.3390/cells11182779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
The restoration of an intact epidermal barrier after wound injury is the culmination of a highly complex and exquisitely regulated physiological process involving multiple cells and tissues, overlapping dynamic events and protein synthesis and regulation. Central to this process is the cytoskeleton, a system of intracellular proteins that are instrumental in regulating important processes involved in wound repair including chemotaxis, cytokinesis, proliferation, migration, and phagocytosis. One highly conserved family of cytoskeletal proteins that are emerging as major regulators of actin and microtubule nucleation, polymerization, and stabilization are the formins. The formin family includes 15 different proteins categorized into seven subfamilies based on three formin homology domains (FH1, FH2, and FH3). The formins themselves are regulated in different ways including autoinhibition, activation, and localization by a range of proteins, including Rho GTPases. Herein, we describe the roles and effects of the formin family of cytoskeletal proteins on the fundamental process of wound healing and highlight recent advances relating to their important functions, mechanisms, and regulation at the molecular and cellular levels.
Collapse
|
5
|
Pan MH, Wan X, Wang HH, Pan ZN, Zhang Y, Sun SC. FMNL3 regulates FASCIN for actin-mediated spindle migration and cytokinesis in mouse oocytes†. Biol Reprod 2021; 102:1203-1212. [PMID: 32167535 DOI: 10.1093/biolre/ioaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/23/2020] [Accepted: 03/12/2020] [Indexed: 11/14/2022] Open
Abstract
Formin-like 3 (FMNL3) is a member of the formin-likes (FMNLs), which belong to the formin family. As an F-actin nucleator, FMNL3 is essential for several cellular functions, such as polarity control, invasion, and migration. However, the roles of FMNL3 during oocytes meiosis remain unclear. In this study, we investigated the functions of FMNL3 during mouse oocyte maturation. Our results showed that FMNL3 mainly concentrated in the oocyte cortex and spindle periphery. Depleting FMNL3 led to the failure of polar body extrusion, and we also found large polar bodies in the FMNL3-deleted oocytes, indicating the occurrence of symmetric meiotic division. There was no effect of FMNL3 on spindle organization; however, we observed spindle migration defects at late metaphase I, which might be due to the decreased cytoplasmic actin. Microinjecting Fmnl3-EGFP mRNA into Fmnl3-depleted oocytes significantly rescued these defects. In addition, the results of co-immunoprecipitation and the perturbation of protein expression experiments suggested that FMNL3 interacted with the actin-binding protein FASCIN for the regulation of actin filaments in oocytes. Thus, our results provide the evidence that FMNL3 regulates FASCIN for actin-mediated spindle migration and cytokinesis during mouse oocyte meiosis.
Collapse
Affiliation(s)
- Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Hong-Hui Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| |
Collapse
|
6
|
Salanga CM, Salanga MC. Genotype to Phenotype: CRISPR Gene Editing Reveals Genetic Compensation as a Mechanism for Phenotypic Disjunction of Morphants and Mutants. Int J Mol Sci 2021; 22:ijms22073472. [PMID: 33801686 PMCID: PMC8036752 DOI: 10.3390/ijms22073472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/25/2022] Open
Abstract
Forward genetic screens have shown the consequences of deleterious mutations; however, they are best suited for model organisms with fast reproductive rates and large broods. Furthermore, investigators must faithfully identify changes in phenotype, even if subtle, to realize the full benefit of the screen. Reverse genetic approaches also probe genotype to phenotype relationships, except that the genetic targets are predefined. Until recently, reverse genetic approaches relied on non-genomic gene silencing or the relatively inefficient, homology-dependent gene targeting for loss-of-function generation. Fortunately, the flexibility and simplicity of the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system has revolutionized reverse genetics, allowing for the precise mutagenesis of virtually any gene in any organism at will. The successful integration of insertions/deletions (INDELs) and nonsense mutations that would, at face value, produce the expected loss-of-function phenotype, have been shown to have little to no effect, even if other methods of gene silencing demonstrate robust loss-of-function consequences. The disjunction between outcomes has raised important questions about our understanding of genotype to phenotype and highlights the capacity for compensation in the central dogma. This review describes recent studies in which genomic compensation appears to be at play, discusses the possible compensation mechanisms, and considers elements important for robust gene loss-of-function studies.
Collapse
Affiliation(s)
- Cristy M. Salanga
- Office of the Vice President for Research, Northern Arizona University, Flagstaff, AZ 86011, USA;
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Matthew C. Salanga
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
- Correspondence:
| |
Collapse
|
7
|
Biber G, Ben-Shmuel A, Sabag B, Barda-Saad M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:131-196. [PMID: 33066873 DOI: 10.1016/bs.ircmb.2020.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.
Collapse
Affiliation(s)
- G Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - A Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - B Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
8
|
Fonseca CG, Barbacena P, Franco CA. Endothelial cells on the move: dynamics in vascular morphogenesis and disease. VASCULAR BIOLOGY 2020; 2:H29-H43. [PMID: 32935077 PMCID: PMC7487603 DOI: 10.1530/vb-20-0007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
The vascular system is a hierarchically organized network of blood vessels that play crucial roles in embryogenesis, homeostasis and disease. Blood vessels are built by endothelial cells – the cells lining the interior of blood vessels – through a process named vascular morphogenesis. Endothelial cells react to different biomechanical signals in their environment by adjusting their behavior to: (1) invade, proliferate and fuse to form new vessels (angiogenesis); (2) remodel, regress and establish a hierarchy in the network (patterning); and (3) maintain network stability (quiescence). Each step involves the coordination of endothelial cell differentiation, proliferation, polarity, migration, rearrangements and shape changes to ensure network integrity and an efficient barrier between blood and tissues. In this review, we highlighted the relevance and the mechanisms involving endothelial cell migration during different steps of vascular morphogenesis. We further present evidence on how impaired endothelial cell dynamics can contribute to pathology.
Collapse
Affiliation(s)
- Catarina G Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Barbacena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Claudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
9
|
Vaeyens MM, Jorge-Peñas A, Barrasa-Fano J, Shapeti A, Roeffaers M, Van Oosterwyck H. Actomyosin-dependent invasion of endothelial sprouts in collagen. Cytoskeleton (Hoboken) 2020; 77:261-276. [PMID: 32588525 DOI: 10.1002/cm.21624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
During sprouting angiogenesis-the growth of blood vessels from the existing vasculature-endothelial cells (ECs) adopt an elongated invasive form and exert forces at cell-cell and cell-matrix interaction sites. These cell shape changes and cellular tractions require extensive reorganizations of the actomyosin network. However, the respective roles of actin and myosin for endothelial sprouting are not fully elucidated. In this study, we further investigate these roles by treating 2D-migrating and 3D-sprouting ECs with chemical compounds targeting either myosin or actin. These treatments affected the endothelial cytoskeleton drastically and reduced the invasive response in a compound-specific manner; pointing toward a tight control of the actin and myosin activity during sprouting. Clusters in the data further illustrate that endothelial sprout morphology is sensitive to the in vitro model mechanical microenvironment and directs future research toward mechanical substrate guidance as a strategy for promoting engineered tissue vascularization. In summary, our results add to a growing corpus of research highlighting a key role of the cytoskeleton for sprouting angiogenesis.
Collapse
Affiliation(s)
- Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jorge Barrasa-Fano
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Apeksha Shapeti
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Roeffaers
- Department of Microbial and Molecular Systems (M2S), Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Liu J, Chen S, Chen Y, Geng N, Feng C. High expression of FMNL3 associates with cancer cell migration, invasion, and unfavorable prognosis in tongue squamous cell carcinoma. J Oral Pathol Med 2019; 48:459-467. [PMID: 30955218 DOI: 10.1111/jop.12857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/22/2019] [Accepted: 03/17/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Jiameng Liu
- Department of Oral and Maxillofacial Surgery The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Shan Chen
- Department of Oral and Maxillofacial Surgery The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Yang Chen
- Department of Oral and Maxillofacial Surgery The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Ningbo Geng
- Department of Oral and Maxillofacial Surgery The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Chongjin Feng
- Department of Oral and Maxillofacial Surgery The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| |
Collapse
|
11
|
Ludwinski FE, Patel AS, Damodaran G, Cho J, Furmston J, Xu Q, Jayasinghe SN, Smith A, Modarai B. Encapsulation of macrophages enhances their retention and angiogenic potential. NPJ Regen Med 2019; 4:6. [PMID: 30911410 PMCID: PMC6426993 DOI: 10.1038/s41536-019-0068-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/19/2018] [Indexed: 02/02/2023] Open
Abstract
Cell therapies to treat critical limb ischaemia have demonstrated only modest results in clinical trials, and this has been partly attributed to poor cell retention following their delivery directly into the ischaemic limb. The aim of this study was to determine whether alginate encapsulation of therapeutic pro-angio/arteriogenic macrophages enhances their retention and ultimately improves limb perfusion. A reproducible GMP-compliant method for generating 300 µm alginate capsules was developed to encapsulate pro-angio/arteriogenic macrophages. Longitudinal analysis revealed no detrimental effect of encapsulation on cell number or viability in vitro, and macrophages retained their pro-angio/arteriogenic phenotype. Intramuscular delivery of encapsulated macrophages into the murine ischaemic hindlimb demonstrated increased cell retention compared with injection of naked cells (P = 0.0001), and that this was associated both enhanced angiogenesis (P = 0.02) and arteriogenesis (P = 0.03), and an overall improvement in limb perfusion (P = 0.0001). Alginate encapsulation of pro-angio/arteriogenic macrophages enhances cell retention and subsequent limb reperfusion in vivo. Encapsulation may therefore represent a means of improving the efficacy of cell-based therapies currently under investigation for the treatment of limb ischaemia. Blood vessel-promoting immune cells stay longer in the body and help promote blood flow to the feet and toes of mice with critical limb ischemia when the therapeutic cells are packaged inside tiny bubbles of a biocompatible seaweed derivative called alginate. A team led by Bijan Modarai from King’s College London, UK, developed a reliable method for placing artery-stimulating macrophage cells inside alginate capsules measuring 300 micrometres in diameter, about the thickness of a postcard. In culture, the alginate coating had no effect on the macrophage viability; and when injected into the muscles of mice with artery blockages to their hindlimbs, the encapsulated cells were retained longer and offered greater therapeutic benefit than uncoated cells. This encapsulation strategy may improve the efficacy of comparable cell-based therapies for humans with limb ischemia.
Collapse
Affiliation(s)
- Francesca E Ludwinski
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Ashish S Patel
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Gopinath Damodaran
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Jun Cho
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Joanna Furmston
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Qingbo Xu
- 2King's College London, Vascular Biology Section, School of Cardiovascular Medicine & Sciences, BHF Centre of Excellence, King's College London, London, UK
| | - Suwan N Jayasinghe
- 3BioPhysics Group, UCL Centre for Stem Cells and Regenerative Medicine, UCL Department of Mechanical Engineering and UCL Institute of Healthcare Engineering, University College London, Torrington Place, London, WC1E 7JE UK
| | - Alberto Smith
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Bijan Modarai
- 1King's College London, Academic Department of Vascular Surgery, School of Cardiovascular Medicine & Sciences, BHF Centre for Regenerative Medicine and BHF Centre of Excellence and the Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
12
|
Multiple roles of the actin and microtubule-regulating formins in the developing brain. Neurosci Res 2019; 138:59-69. [DOI: 10.1016/j.neures.2018.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
|
13
|
Morikawa S, Iribar H, Gutiérrez-Rivera A, Ezaki T, Izeta A. Pericytes in Cutaneous Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:1-63. [DOI: 10.1007/978-3-030-16908-4_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Detection and Classification of Hard and Soft Sweeps from Unphased Genotypes by Multilocus Genotype Identity. Genetics 2018; 210:1429-1452. [PMID: 30315068 DOI: 10.1534/genetics.118.301502] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 10/08/2018] [Indexed: 11/18/2022] Open
Abstract
Positive natural selection can lead to a decrease in genomic diversity at the selected site and at linked sites, producing a characteristic signature of elevated expected haplotype homozygosity. These selective sweeps can be hard or soft. In the case of a hard selective sweep, a single adaptive haplotype rises to high population frequency, whereas multiple adaptive haplotypes sweep through the population simultaneously in a soft sweep, producing distinct patterns of genetic variation in the vicinity of the selected site. Measures of expected haplotype homozygosity have previously been used to detect sweeps in multiple study systems. However, these methods are formulated for phased haplotype data, typically unavailable for nonmodel organisms, and some may have reduced power to detect soft sweeps due to their increased genetic diversity relative to hard sweeps. To address these limitations, we applied the H12 and H2/H1 statistics proposed in 2015 by Garud et al., which have power to detect both hard and soft sweeps, to unphased multilocus genotypes, denoting them as G12 and G2/G1. G12 (and the more direct expected homozygosity analog to H12, denoted G123) has comparable power to H12 for detecting both hard and soft sweeps. G2/G1 can be used to classify hard and soft sweeps analogously to H2/H1, conditional on a genomic region having high G12 or G123 values. The reason for this power is that, under random mating, the most frequent haplotypes will yield the most frequent multilocus genotypes. Simulations based on parameters compatible with our recent understanding of human demographic history suggest that expected homozygosity methods are best suited for detecting recent sweeps, and increase in power under recent population expansions. Finally, we find candidates for selective sweeps within the 1000 Genomes CEU, YRI, GIH, and CHB populations, which corroborate and complement existing studies.
Collapse
|
15
|
Zeng YF, Xiao YS, Liu Y, Luo XJ, Wen LD, Liu Q, Chen M. Formin-like 3 regulates RhoC/FAK pathway and actin assembly to promote cell invasion in colorectal carcinoma. World J Gastroenterol 2018; 24:3884-3897. [PMID: 30228782 PMCID: PMC6141330 DOI: 10.3748/wjg.v24.i34.3884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/16/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To clarify the underlying mechanism of formin-like 3 (FMNL3) in the promotion of colorectal carcinoma (CRC) cell invasion.
METHODS The in vitro biological function analyses of FMNL3 were performed by gain- and loss-of function approaches. Changes in the F-actin cytoskeleton were detected by the technologies of phalloidin-TRITC labeling and confocal microscopy. The signaling pathway mediated by FMNL3 was explored by western blot, gelatin zymograph assay, co-immunoprecipitation (co-IP), immunofluorescence co-localization, and glutathione S-transferase (GST) pull-down assay.
RESULTS The in vitro experimental results showed that FMNL3 significantly promoted the proliferation, invasion, and migration of CRC cells (P < 0.05 and P < 0.01). Moreover, FMNL3 regulated the remodeling of actin-based protrusions such as filopodia and lamellipodia in a RhoC-dependent manner. The western blot and gelatin zymograph assay results indicated that FMNL3 was involved in the RhoC/ focal adhesion kinase (FAK) pathway and acted as an effector of RhoC to activate the downstream signaling of p-FAK as well as p-MAPK and p-AKT. This resulted in the increased expression of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9) and vascular endothelial growth factor (VEGF), and the subsequent promotion of CRC cell invasion. The results of TAE226, U0126 or Ly294002 treatment confirmed an essential role of FMNL3 in activation of the RhoC/FAK pathway and the subsequent promotion of CRC invasion. Co-IP, co-localization and GST pull-down assays showed the direct interaction of FMNL3 with RhoC in vivo and in vitro.
CONCLUSION FMNL3 regulates the RhoC/FAK signaling pathway and RhoC-dependent remodeling of actin-based protrusions to promote CRC invasion.
Collapse
Affiliation(s)
- Yuan-Feng Zeng
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Yi-Sheng Xiao
- Teaching and Researching Section of Morphology, College of Basic Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yong Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Xiao-Jiang Luo
- Department of General Surgery, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Li-Dan Wen
- Clinical Medical Sciences Institute, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Qian Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Min Chen
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
16
|
Fischer RS, Lam PY, Huttenlocher A, Waterman CM. Filopodia and focal adhesions: An integrated system driving branching morphogenesis in neuronal pathfinding and angiogenesis. Dev Biol 2018; 451:86-95. [PMID: 30193787 DOI: 10.1016/j.ydbio.2018.08.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/08/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022]
Abstract
Single cell branching during development in vertebrates is typified by neuronal branching to form neurites and vascular branches formed by sprouting angiogenesis. Neurons and endothelial tip cells possess subcellular protrusions that share many common features from the morphological to the molecular level. Both systems utilize filopodia as their cellular protrusion organelles and depend on specific integrin-mediated adhesions to the local extracellular matrix for guidance in their pathfinding. We discuss the similar molecular machineries involved in these two types of cell branch formation and use their analogy to propose a new mechanism for angiogenic filopodia function, namely as adhesion assembly sites. In support of this model we provide primary data of angiogenesis in zebrafish in vivo showing that the actin assembly factor VASP participates in both filopodia formation and adhesion assembly at the base of the filopodia, enabling forward progress of the tip cell. The use of filopodia and their associated adhesions provide a common mechanism for neuronal and endothelial pathfinding during development in response to extracellular matrix cues.
Collapse
Affiliation(s)
- Robert S Fischer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States
| | - Pui-Ying Lam
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, United States
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin, United States
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, United States.
| |
Collapse
|
17
|
LeCorgne H, Tudosie AM, Lavik K, Su R, Becker KN, Moore S, Walia Y, Wisner A, Koehler D, Alberts AS, Williams FE, Eisenmann KM. Differential Toxicity of mDia Formin-Directed Functional Agonists and Antagonists in Developing Zebrafish. Front Pharmacol 2018; 9:340. [PMID: 29692731 PMCID: PMC5902741 DOI: 10.3389/fphar.2018.00340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/23/2018] [Indexed: 12/16/2022] Open
Abstract
The mammalian Diaphanous-related (mDia) formins are cytoskeletal regulators that assemble and, in some cases, bundle filamentous actin (F-actin), as well as stabilize microtubules. The development of small molecule antagonists and agonists that interrogate mDia formin function has allowed us to investigate the roles of formins in disease states. A small molecule inhibitor of FH2 domain (SMIFH2) inhibits mDia-dependent actin dynamics and abrogates tumor cell migration and cell division in vitro and ex vivo tissue explants. mDia formin activation with small molecule intramimics IMM01/02 and mDia2-DAD peptides inhibited glioblastoma motility and invasion in vitro and ex vivo rat brain slices. However, SMIFH2, IMMs, and mDia2 DAD efficacy in vivo remains largely unexplored and potential toxicity across a range of developmental phenotypes has not been thoroughly characterized. In this study, we performed an in vivo screen of early life-stage toxicity in Danio rerio zebrafish embryos 2 days post-fertilization (dpf) in response to SMIFH2, IMM01/02, and mDia2 DAD. SMIFH2 at concentrations ≥5–10 μM induced significant defects in developing zebrafish, including shorter body lengths, tail curvature and defective tail cellularity, craniofacial malformations, pericardial edema, absent and/or compromised vasculature function and flow, depressed heart rates and increased mortality. Conversely, IMM and mDia2 DAD peptides were minimally toxic at concentrations up to 10–20 and 50 μM, respectively. SMIFH2's therapeutic potential may therefore be limited by its substantial in vivo toxicity at functional concentrations. mDia formin agonism with IMMs and mDia2 DADs may therefore be a more effective and less toxic anti-invasive therapeutic approach.
Collapse
Affiliation(s)
- Hunter LeCorgne
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Andrew M Tudosie
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Kari Lavik
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Robin Su
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Kathryn N Becker
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Sara Moore
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Yashna Walia
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| | - Alexander Wisner
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science, Toledo, OH, United States
| | - Daniel Koehler
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science, Toledo, OH, United States
| | - Arthur S Alberts
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Frederick E Williams
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science, Toledo, OH, United States
| | - Kathryn M Eisenmann
- Department of Cancer Biology, University of Toledo Health Science, Toledo, OH, United States
| |
Collapse
|
18
|
Huebner H, Knoerr B, Betzler A, Hartner A, Kehl S, Baier F, Wachter D, Strick R, Beckmann M, Fahlbusch F, Ruebner M. Detyrosinated tubulin is decreased in fetal vessels of preeclampsia placentas. Placenta 2018; 62:58-65. [DOI: 10.1016/j.placenta.2017.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 11/29/2022]
|
19
|
An CI, Ichihashi Y, Peng J, Sinha NR, Hagiwara N. Transcriptome Dynamics and Potential Roles of Sox6 in the Postnatal Heart. PLoS One 2016; 11:e0166574. [PMID: 27832192 PMCID: PMC5104335 DOI: 10.1371/journal.pone.0166574] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/31/2016] [Indexed: 01/20/2023] Open
Abstract
The postnatal heart undergoes highly coordinated developmental processes culminating in the complex physiologic properties of the adult heart. The molecular mechanisms of postnatal heart development remain largely unexplored despite their important clinical implications. To gain an integrated view of the dynamic changes in gene expression during postnatal heart development at the organ level, time-series transcriptome analyses of the postnatal hearts of neonatal through adult mice (P1, P7, P14, P30, and P60) were performed using a newly developed bioinformatics pipeline. We identified functional gene clusters by principal component analysis with self-organizing map clustering which revealed organized, discrete gene expression patterns corresponding to biological functions associated with the neonatal, juvenile and adult stages of postnatal heart development. Using weighted gene co-expression network analysis with bootstrap inference for each of these functional gene clusters, highly robust hub genes were identified which likely play key roles in regulating expression of co-expressed, functionally linked genes. Additionally, motivated by the role of the transcription factor Sox6 in the functional maturation of skeletal muscle, the role of Sox6 in the postnatal maturation of cardiac muscle was investigated. Differentially expressed transcriptome analyses between Sox6 knockout (KO) and control hearts uncovered significant upregulation of genes involved in cell proliferation at postnatal day 7 (P7) in the Sox6 KO heart. This result was validated by detecting mitotically active cells in the P7 Sox6 KO heart. The current report provides a framework for the complex molecular processes of postnatal heart development, thus enabling systematic dissection of the developmental regression observed in the stressed and failing adult heart.
Collapse
Affiliation(s)
- Chung-Il An
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Yasunori Ichihashi
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| | - Jie Peng
- Department of Statistics, University of California Davis, Davis, California, United States of America
| | - Neelima R. Sinha
- Department of Plant Biology, University of California Davis, Davis, California, United States of America
| | - Nobuko Hagiwara
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California Davis, Davis, California, United States of America
- * E-mail: (CA); (YI); (NH)
| |
Collapse
|
20
|
Péladeau C, Heibein A, Maltez MT, Copeland SJ, Copeland JW. A specific FMNL2 isoform is up-regulated in invasive cells. BMC Cell Biol 2016; 17:32. [PMID: 27578625 PMCID: PMC5006604 DOI: 10.1186/s12860-016-0110-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/23/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Formins are a highly conserved family of cytoskeletal remodeling proteins. A growing body of evidence suggests that formins play key roles in the progression and spread of a variety of cancers. There are 15 human formin proteins and of these the Diaphanous-Related Formins (DRFs) are the best characterized. Included in the DRFs are the Formin-Like proteins, FMNL1, 2 & 3, each of which have been strongly implicated in driving tumorigenesis and metastasis of specific tumors. In particular, increased FMNL2 expression correlates with increased invasiveness of colorectal cancer (CRC) in vivo and for a variety of CRC cell-lines in vitro. FMNL2 expression is also required for invasive cell motility in other cancer cell-lines. There are multiple alternatively spliced isoforms of FMNL2 and it is predicted that the encoded proteins will differ in their regulation, subcellular localization and in their ability to regulate cytoskeletal dynamics. RESULTS Using RT-PCR we identified four FMNL2 isoforms expressed in CRC and melanoma cell-lines. We find that a previously uncharacterized FMNL2 isoform is predominantly expressed in a variety of melanoma and CRC cell lines; this isoform is also more effective in driving 3D motility. Building on previous reports, we also show that FMNL2 is required for invasion in A375 and WM266.4 melanoma cells. CONCLUSIONS Taken together, these results suggest that FMNL2 is likely to be generally required in melanoma cells for invasion, that a specific isoform of FMNL2 is up-regulated in invasive CRC and melanoma cells and this isoform is the most effective at facilitating invasion.
Collapse
Affiliation(s)
- Christine Péladeau
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Allan Heibein
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Melissa T Maltez
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Sarah J Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - John W Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
21
|
Cell-cell junctional mechanotransduction in endothelial remodeling. Cell Mol Life Sci 2016; 74:279-292. [PMID: 27506620 PMCID: PMC5219012 DOI: 10.1007/s00018-016-2325-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/15/2016] [Accepted: 08/03/2016] [Indexed: 02/06/2023]
Abstract
The vasculature is one of the most dynamic tissues that encounter numerous mechanical cues derived from pulsatile blood flow, blood pressure, activity of smooth muscle cells in the vessel wall, and transmigration of immune cells. The inner layer of blood and lymphatic vessels is covered by the endothelium, a monolayer of cells which separates blood from tissue, an important function that it fulfills even under the dynamic circumstances of the vascular microenvironment. In addition, remodeling of the endothelial barrier during angiogenesis and trafficking of immune cells is achieved by specific modulation of cell-cell adhesion structures between the endothelial cells. In recent years, there have been many new discoveries in the field of cellular mechanotransduction which controls the formation and destabilization of the vascular barrier. Force-induced adaptation at endothelial cell-cell adhesion structures is a crucial node in these processes that challenge the vascular barrier. One of the key examples of a force-induced molecular event is the recruitment of vinculin to the VE-cadherin complex upon pulling forces at cell-cell junctions. Here, we highlight recent advances in the current understanding of mechanotransduction responses at, and derived from, endothelial cell-cell junctions. We further discuss their importance for vascular barrier function and remodeling in development, inflammation, and vascular disease.
Collapse
|
22
|
Rengarajan M, Hayer A, Theriot JA. Endothelial Cells Use a Formin-Dependent Phagocytosis-Like Process to Internalize the Bacterium Listeria monocytogenes. PLoS Pathog 2016; 12:e1005603. [PMID: 27152864 PMCID: PMC4859537 DOI: 10.1371/journal.ppat.1005603] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 04/06/2016] [Indexed: 01/11/2023] Open
Abstract
Vascular endothelial cells act as gatekeepers that protect underlying tissue from blood-borne toxins and pathogens. Nevertheless, endothelial cells are able to internalize large fibrin clots and apoptotic debris from the bloodstream, although the precise mechanism of such phagocytosis-like uptake is unknown. We show that cultured primary human endothelial cells (HUVEC) internalize both pathogenic and non-pathogenic Listeria bacteria comparably, in a phagocytosis-like process. In contrast with previously studied host cell types, including intestinal epithelial cells and hepatocytes, we find that endothelial internalization of Listeria is independent of all known pathogenic bacterial surface proteins. Consequently, we exploited the internalization and intracellular replication of L. monocytogenes to identify distinct host cell factors that regulate phagocytosis-like uptake in HUVEC. Using siRNA screening and subsequent genetic and pharmacologic perturbations, we determined that endothelial infectivity was modulated by cytoskeletal proteins that normally modulate global architectural changes, including phosphoinositide-3-kinase, focal adhesions, and the small GTPase Rho. We found that Rho kinase (ROCK) is acutely necessary for adhesion of Listeria to endothelial cells, whereas the actin-nucleating formins FHOD1 and FMNL3 specifically regulate internalization of bacteria as well as inert beads, demonstrating that formins regulate endothelial phagocytosis-like uptake independent of the specific cargo. Finally, we found that neither ROCK nor formins were required for macrophage phagocytosis of L. monocytogenes, suggesting that endothelial cells have distinct requirements for bacterial internalization from those of classical professional phagocytes. Our results identify a novel pathway for L. monocytogenes uptake by human host cells, indicating that this wily pathogen can invade a variety of tissues by using a surprisingly diverse suite of distinct uptake mechanisms that operate differentially in different host cell types.
Collapse
Affiliation(s)
- Michelle Rengarajan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Julie A. Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Abstract
During angiogenesis, endothelial cells invade into the stromal matrix: a complex, structured array of extracellular matrix proteins. This three-dimensional deformable substrate also contains a mixture of angiogenic factors as well as embedded stromal cells. Interactions between endothelial cells and the stromal tissue make complex and important contributions to the process of angiogenesis; however, the composition of the stromal matrix is hard to replicate in vitro. The coculture angiogenesis assay is a long-term assay that uses fibroblasts to secrete and condition a stromal matrix that more closely mimics tissue than a simple collagen gel. Like all in vitro assays of angiogenesis, it has both strengths and weaknesses. Here we give protocols for the two of the most useful applications of the assay: screening for regulators of angiogenesis and high-resolution imaging.
Collapse
Affiliation(s)
- Mark Richards
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Harry Mellor
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
24
|
Formins at the Junction. Trends Biochem Sci 2015; 41:148-159. [PMID: 26732401 DOI: 10.1016/j.tibs.2015.12.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 12/21/2022]
Abstract
The actin cytoskeleton and adhesion junctions are physically and functionally coupled at the cell-cell interface between epithelial cells. The actin regulatory complex Arp2/3 has an established role in the turnover of junctional actin; however, the role of formins, the largest group of actin regulators, is less clear. Formins dynamically shape the actin cytoskeleton and have various functions within cells. In this review we describe recent progress on how formins regulate actin dynamics at cell-cell contacts and highlight formin functions during polarized protein traffic necessary for epithelialization.
Collapse
|
25
|
The Formin FMNL3 Controls Early Apical Specification in Endothelial Cells by Regulating the Polarized Trafficking of Podocalyxin. Curr Biol 2015; 25:2325-31. [PMID: 26299518 DOI: 10.1016/j.cub.2015.07.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/27/2015] [Accepted: 07/16/2015] [Indexed: 01/11/2023]
Abstract
Angiogenesis is the fundamental process by which new blood vessels form from pre-existing vasculature. It plays a critical role in the formation of the vasculature during development and is triggered in response to tissue hypoxia in adult organisms. This process requires complex and coordinated regulation of the endothelial cell cytoskeleton to control cell shape and polarity. In our previous work, we showed that the cytoskeletal regulator FMNL3/FRL2 controls the alignment of stabilized microtubules during polarized endothelial cell elongation and that depletion of FMNL3 retards elongation of the intersegmental vessels in zebrafish. Recent work has shown that FMNL3 is also needed for vascular lumen formation, a critical element of the formation of functional vessels. Here, we show that FMNL3 interacts with Cdc42 and RhoJ, two Rho family GTPases known to be required for lumen formation. FMNL3 and RhoJ are concentrated at the early apical surface, or AMIS, and regulate the formation of radiating actin cables from this site. In diverse biological systems, formins mediate polarized trafficking through the generation of similar actin filaments tracks. We show that FMNL3 and RhoJ are required for polarized trafficking of podocalyxin to the early apical surface--an important event in vascular lumenogenesis.
Collapse
|
26
|
Abstract
The morpholino anti-sense technology has been used extensively to test gene function. The zebrafish model allows a detailed comparison of knockdown (anti-sense) and knockout (mutation) effects. Recent studies reveal that these two approaches can often lead to surprisingly different phenotypes, thus raising a number of important questions.
Collapse
|
27
|
Phng LK, Gebala V, Bentley K, Philippides A, Wacker A, Mathivet T, Sauteur L, Stanchi F, Belting HG, Affolter M, Gerhardt H. Formin-mediated actin polymerization at endothelial junctions is required for vessel lumen formation and stabilization. Dev Cell 2015; 32:123-32. [PMID: 25584798 DOI: 10.1016/j.devcel.2014.11.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 07/31/2014] [Accepted: 11/10/2014] [Indexed: 12/31/2022]
Abstract
During blood vessel formation, endothelial cells (ECs) establish cell-cell junctions and rearrange to form multicellular tubes. Here, we show that during lumen formation, the actin nucleator and elongation factor, formin-like 3 (fmnl3), localizes to EC junctions, where filamentous actin (F-actin) cables assemble. Fluorescent actin reporters and fluorescence recovery after photobleaching experiments in zebrafish embryos identified a pool of dynamic F-actin with high turnover at EC junctions in vessels. Knockdown of fmnl3 expression, chemical inhibition of formin function, and expression of dominant-negative fmnl3 revealed that formin activity maintains a stable F-actin content at EC junctions by continual polymerization of F-actin cables. Reduced actin polymerization leads to destabilized endothelial junctions and consequently to failure in blood vessel lumenization and lumen instability. Our findings highlight the importance of formin activity in blood vessel morphogenesis.
Collapse
Affiliation(s)
- Li-Kun Phng
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Véronique Gebala
- Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3LY, UK
| | - Katie Bentley
- Computational Biology Laboratory, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Philippides
- Centre for Computational Neuroscience and Robotics, Department of Informatics, University of Sussex, Brighton BN1 9QJ, UK
| | - Andrin Wacker
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Thomas Mathivet
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Loïc Sauteur
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Fabio Stanchi
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Holger Gerhardt
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3LY, UK.
| |
Collapse
|
28
|
Cdc42 mediates Bmp-induced sprouting angiogenesis through Fmnl3-driven assembly of endothelial filopodia in zebrafish. Dev Cell 2015; 32:109-22. [PMID: 25584797 DOI: 10.1016/j.devcel.2014.11.024] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 07/05/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
Abstract
During angiogenesis in vivo, endothelial cells (ECs) at the tips of vascular sprouts actively extend filopodia that are filled with bundles of linear actin filaments. To date, signaling pathways involved in the formation of endothelial filopodia have been studied using in-vitro-cultured ECs that behave differently from those in vivo. Herein, we have delineated a signaling pathway that governs the assembly of endothelial filopodia during angiogenic sprouting of the caudal vein plexus (CVP) in zebrafish. During CVP formation, bone morphogenetic protein induces the extension of endothelial filopodia and their migration via Arhgef9b-mediated activation of Cdc42. Active Cdc42 binds to and stimulates Formin-like 3, an actin-regulatory protein of the formin family, which, in turn, promotes the extension of endothelial filopodia to facilitate angiogenic sprouting of the CVP. Thus, this study has elucidated molecular mechanisms underlying the formation of endothelial filopodia and their role in angiogenesis in vivo.
Collapse
|
29
|
Zeng YF, Xiao YS, Lu MZ, Luo XJ, Hu GZ, Deng KY, Wu XM, Xin HB. Increased expression of formin-like 3 contributes to metastasis and poor prognosis in colorectal carcinoma. Exp Mol Pathol 2015; 98:260-7. [PMID: 25758200 DOI: 10.1016/j.yexmp.2015.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023]
Abstract
Formin-like 3 (FMNL3), a member of diaphanous-related formins subfamily, plays an important role in cytoskeleton reorganization, cell adhesion and cancer cell invasion in vitro. This study aimed to explore the expression of FMNL3 in colorectal carcinoma (CRC) cell-lines and tissues, and further evaluate its prognostic value and correlation with the clinicopathological parameters, and also investigate the effects of FMNL3 gene silencing on the growth and metastasis of CRC in vivo. Immunohistochemical analysis showed that FMNL3 protein was distributed in a punctuate aggregation pattern and located mainly in the cytoplasm of glandular cavity side, close to the nucleus of CRC cells. The positive rate of FMNL3 expression was 87.5% (84/96) in CRC, which was significantly higher than that in adjacent normal mucosa (30%, 9/30). Moreover, FMNL3 protein expressed far more in primary CRC with metastasis and corresponding lymph nodes metastatic CRC than in primary CRC without metastasis. Increased expression of FMNL3 was closely correlated with tumor size, differentiation, serosal invasion, and both lymph node metastasis and distant metastasis. However, it was not correlated with patients' age and gender. According to Kaplan-Meier survival analyses, patients with FMNL3 high expression level had lower overall survival rate than that with FMNL3 low expression level. Univariate and multivariate analyses revealed that high FMNL3 expression was a significant and independent prognostic predictor of patients with CRC. In addition, FMNL3 mRNA and protein levels were substantially up-regulated in CRC-metastasis-derived cell lines, as compared to those in primary-CRC-derived ones. FMNL3 gene silencing suppressed the growth and metastasis of CRC in vivo. In conclusion, FMNL3 plays an important role in the progression and metastasis of CRC and may be a novel potential prognostic predictor and therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Yuan-Feng Zeng
- Department of Pathology, Jiangxi Provincial People's Hospital, Nanchang, China; Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yi-Sheng Xiao
- Teaching and Researching Section of Morphology, College of Basic Medicine, Jiangxi University of Chinese Traditional Medicine, Nanchang, China
| | - Ming-Zhi Lu
- Department of Pathology, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Xiao-Jiang Luo
- Department of General Surgery, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Guo-Zhu Hu
- Institute of Clinical Medical Sciences, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Ke-Yu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Xiao-Mu Wu
- Jiangxi Institute of Neurology, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, China.
| |
Collapse
|
30
|
Abstract
Angiogenesis is a complex process involving the interactions of endothelial cells not only with pro-angiogenic factors but also with stromal cells and stromal matrix components. Modeling this process in vitro is challenging, and many different assays have been described, each with their own particular strengths and weaknesses. The coculture assay is a long-term assay of angiogenesis that uses fibroblasts to secrete and condition a stromal matrix that more closely mimics tissue than a simple collagen gel. The assay is particularly suited to screening for angiogenic regulators and also for high-resolution imaging of endothelial cells undergoing angiogenic morphogenesis.
Collapse
|
31
|
Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrafish. Dev Cell 2014; 32:97-108. [PMID: 25533206 DOI: 10.1016/j.devcel.2014.11.018] [Citation(s) in RCA: 562] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/19/2014] [Accepted: 11/10/2014] [Indexed: 12/16/2022]
Abstract
The widespread availability of programmable site-specific nucleases now enables targeted gene disruption in the zebrafish. In this study, we applied site-specific nucleases to generate zebrafish lines bearing individual mutations in more than 20 genes. We found that mutations in only a small proportion of genes caused defects in embryogenesis. Moreover, mutants for ten different genes failed to recapitulate published Morpholino-induced phenotypes (morphants). The absence of phenotypes in mutant embryos was not likely due to maternal effects or failure to eliminate gene function. Consistently, a comparison of published morphant defects with the Sanger Zebrafish Mutation Project revealed that approximately 80% of morphant phenotypes were not observed in mutant embryos, similar to our mutant collection. Based on these results, we suggest that mutant phenotypes become the standard metric to define gene function in zebrafish, after which Morpholinos that recapitulate respective phenotypes could be reliably applied for ancillary analyses.
Collapse
|
32
|
Gauvin TJ, Young LE, Higgs HN. The formin FMNL3 assembles plasma membrane protrusions that participate in cell-cell adhesion. Mol Biol Cell 2014; 26:467-77. [PMID: 25428984 PMCID: PMC4310738 DOI: 10.1091/mbc.e14-07-1247] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
FMNL3 localizes broadly over the plasma membrane as discrete puncta, with particular enrichment in filopodia and ruffles and at cell–cell contacts. In addition, a population of FMNL3-containing vesicles of endocytic origin can fuse with the plasma membrane. FMNL3 suppression causes reductions in filopodia and cell–cell adhesion. FMNL3 is a vertebrate-specific formin protein previously shown to play a role in angiogenesis and cell migration. Here we define the cellular localization of endogenous FMNL3, the dynamics of GFP-tagged FMNL3 during cell migration, and the effects of FMNL3 suppression in mammalian culture cells. The majority of FMNL3 localizes in a punctate pattern, with >95% of these puncta being indistinguishable from the plasma membrane by fluorescence microscopy. A small number of dynamic cytoplasmic FMNL3 patches also exist, which enrich near cell–cell contact sites and fuse with the plasma membrane at these sites. These cytoplasmic puncta appear to be part of larger membranes of endocytic origin. On the plasma membrane, FMNL3 enriches particularly in filopodia and membrane ruffles and at nascent cell–cell adhesions. FMNL3-containing filopodia occur both at the cell–substratum interface and at cell–cell contacts, with the latter being 10-fold more stable. FMNL3 suppression by siRNA has two major effects: decrease in filopodia and compromised cell–cell adhesion in cells migrating as a sheet. Overall our results suggest that FMNL3 functions in assembly of actin-based protrusions that are specialized for cell–cell adhesion.
Collapse
Affiliation(s)
- Timothy J Gauvin
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Lorna E Young
- 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
|
33
|
Pierick AR, McKane M, Wen KK, Bartlett HL. Aip1p dynamics are altered by the R256H mutation in actin. J Vis Exp 2014:e51551. [PMID: 25146730 DOI: 10.3791/51551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Mutations in actin cause a range of human diseases due to specific molecular changes that often alter cytoskeletal function. In this study, imaging of fluorescently tagged proteins using total internal fluorescence (TIRF) microscopy is used to visualize and quantify changes in cytoskeletal dynamics. TIRF microscopy and the use of fluorescent tags also allows for quantification of the changes in cytoskeletal dynamics caused by mutations in actin. Using this technique, quantification of cytoskeletal function in live cells valuably complements in vitro studies of protein function. As an example, missense mutations affecting the actin residue R256 have been identified in three human actin isoforms suggesting this amino acid plays an important role in regulatory interactions. The effects of the actin mutation R256H on cytoskeletal movements were studied using the yeast model. The protein, Aip1, which is known to assist cofilin in actin depolymerization, was tagged with green fluorescent protein (GFP) at the N-terminus and tracked in vivo using TIRF microscopy. The rate of Aip1p movement in both wild type and mutant strains was quantified. In cells expressing R256H mutant actin, Aip1p motion is restricted and the rate of movement is nearly half the speed measured in wild type cells (0.88 ± 0.30 μm/sec in R256H cells compared to 1.60 ± 0.42 μm/sec in wild type cells, p < 0.005).
Collapse
Affiliation(s)
- Alyson R Pierick
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Melissa McKane
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Kuo-Kuang Wen
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Heather L Bartlett
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa; Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa;
| |
Collapse
|
34
|
Abstract
Formin proteins were recognized as effectors of Rho GTPases some 15 years ago. They contribute to different cellular actin cytoskeleton structures by their ability to polymerize straight actin filaments at the barbed end. While not all formins necessarily interact with Rho GTPases, a subgroup of mammalian formins, termed Diaphanous-related formins or DRFs, were shown to be activated by small GTPases of the Rho superfamily. DRFs are autoinhibited in the resting state by an N- to C-terminal interaction that renders the central actin polymerization domain inactive. Upon the interaction with a GTP-bound Rho, Rac, or Cdc42 GTPase, the C-terminal autoregulation domain is displaced from its N-terminal recognition site and the formin becomes active to polymerize actin filaments. In this review we discuss the current knowledge on the structure, activation, and function of formin-GTPase interactions for the mammalian formin families Dia, Daam, FMNL, and FHOD. We describe both direct and indirect interactions of formins with GTPases, which lead to formin activation and cytoskeletal rearrangements. The multifaceted function of formins as effector proteins of Rho GTPases thus reflects the diversity of the actin cytoskeleton in cells.
Collapse
Affiliation(s)
- Sonja Kühn
- Center of Advanced European Studies and Research (caesar); Group Physical Biochemistry; Bonn, Germany
| | - Matthias Geyer
- Center of Advanced European Studies and Research (caesar); Group Physical Biochemistry; Bonn, Germany
| |
Collapse
|
35
|
Randall TS, Ehler E. A formin-g role during development and disease. Eur J Cell Biol 2014; 93:205-11. [PMID: 24342720 DOI: 10.1016/j.ejcb.2013.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 11/22/2022] Open
Abstract
Several different protein families were shown to be involved in the regulation of actin filament formation and have been studied extensively in processes such as cell migration. Among them are members of the formin family, which tend to promote the formation of linear actin filaments. Studies in recent years, often using loss of function animal models, have indicated that formin family members play roles beyond cell motility in vitro and are involved in processes ranging from tissue morphogenesis and cell differentiation to diseases such as cancer and cardiomyopathy. Therefore the aim of this review is to discuss these findings and to start putting them into a subcellular context.
Collapse
Affiliation(s)
- Thomas S Randall
- Randall Division of Cell and Molecular Biophysics, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Elisabeth Ehler
- Randall Division of Cell and Molecular Biophysics, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
36
|
Abu Taha A, Taha M, Seebach J, Schnittler HJ. ARP2/3-mediated junction-associated lamellipodia control VE-cadherin-based cell junction dynamics and maintain monolayer integrity. Mol Biol Cell 2013; 25:245-56. [PMID: 24227887 PMCID: PMC3890345 DOI: 10.1091/mbc.e13-07-0404] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The ARP2/3 complex controls junction-associated intermittent lamellipodia (JAIL), which trigger VE-cadherin adhesion and dynamics. JAIL formation maintains paraendothelial barrier function under physiological conditions and depends on the local VE-cadherin concentration. Maintenance and remodeling of endothelial cell junctions critically depend on the VE-cadherin/catenin complex and its interaction with the actin filament cytoskeleton. Here we demonstrate that local lack of vascular endothelial (VE)-cadherin at established cell junctions causes actin-driven and actin-related protein 2/3 complex (ARP2/3)–controlled lamellipodia to appear intermittently at those sites. Lamellipodia overlap the VE-cadherin–free adjacent plasma membranes and facilitate formation of new VE-cadherin adhesion sites, which quickly move into the junctions, driving VE-cadherin dynamics and remodeling. Inhibition of the ARP2/3 complex by expression of the N-WASP (V)CA domain or application of two ARP2/3 inhibitors, CK-548 and CK-666, blocks VE-cadherin dynamics and causes intercellular gaps. Furthermore, expression of carboxy-terminal–truncated VE-cadherin increases the number of ARP2/3-controlled lamellipodia, whereas overexpression of wild-type VE-cadherin largely blocks it and decreases cell motility. The data demonstrate a functional interrelationship between VE-cadherin–mediated cell adhesion and actin-driven, ARP2/3-controlled formation of new VE-cadherin adhesion sites via intermittently appearing lamellipodia at established cell junctions. This coordinated mechanism controls VE-cadherin dynamics and cell motility and maintains monolayer integrity, thus potentially being relevant in disease and angiogenesis.
Collapse
Affiliation(s)
- Abdallah Abu Taha
- Institute of Anatomy and Vascular Biology, WWU-Münster, 48149 Münster, Germany
| | | | | | | |
Collapse
|
37
|
Santos-Ledo A, Jenny A, Marlow FL. Comparative gene expression analysis of the fmnl family of formins during zebrafish development and implications for tissue specific functions. Gene Expr Patterns 2012; 13:30-7. [PMID: 23072729 DOI: 10.1016/j.gep.2012.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/14/2012] [Accepted: 09/06/2012] [Indexed: 10/27/2022]
Abstract
Fmlns belong to the Formin family, catalysts of linear actin polymerization with mostly unknown roles in vivo. In cell culture Fmnls are involved in cell migration and adhesion and the formation of different types of protrusions including filopodia and blebs, suggesting important roles during development. Moreover, Fmnls can act downstream of Rac and Cdc42, mediators of cytoskeletal changes as targets of important pathways required for shaping tissues. The zebrafish genome encodes five Fmnls. Here we report their tissue specific expression patterns during early development and pharyngula stages. The fmnls show overlapping and distinct expression patterns, which suggest that they could regulate similar processes during development, but may also have independent functions. In particular, we find a strong maternal contribution of all fmnls, but distinct expression patterns in the developing brain eye, ear, heart and vascular system.
Collapse
Affiliation(s)
- Adrián Santos-Ledo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | | | | |
Collapse
|