51
|
Chen J, He J, Ni R, Yang Q, Zhang Y, Luo L. Cerebrovascular Injuries Induce Lymphatic Invasion into Brain Parenchyma to Guide Vascular Regeneration in Zebrafish. Dev Cell 2019; 49:697-710.e5. [PMID: 31006646 DOI: 10.1016/j.devcel.2019.03.022] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022]
Abstract
Damage to regional cerebrovascular networks and neuronal tissues occurs during acute cerebrovascular diseases, such as ischemic stroke. The promotion of vascular regeneration is the most promising therapeutic approach. To understand the cellular and molecular mechanisms underlying brain vascular regeneration, we developed two zebrafish cerebrovascular injury models using genetic ablation and photochemical thrombosis. Although brain parenchyma is physiologically devoid of lymphatic vasculature, we found that cerebrovascular injuries induce rapid ingrowth of meningeal lymphatics into the injured parenchyma. The ingrown lymphatics on one hand become lumenized to drain interstitial fluid to resolve brain edema and on the other hand act as "growing tracks" for nascent blood vessels. The ingrown lymphatic vessels undergo apoptosis and clearance after cerebrovascular regeneration. This study reveals a pathological function of meningeal lymphatics, through previously unexpected ingrowth into brain parenchyma and a newly identified lymphatic function as vascular "growing tracks."
Collapse
Affiliation(s)
- Jingying Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Jianbo He
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Rui Ni
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Qifen Yang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Yaoguang Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
52
|
Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN, Stacker SA. The Interplay Between Lymphatic Vessels and Chemokines. Front Immunol 2019; 10:518. [PMID: 31105685 PMCID: PMC6499173 DOI: 10.3389/fimmu.2019.00518] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 12/21/2022] Open
Abstract
Chemokines are a family of small protein cytokines that act as chemoattractants to migrating cells, in particular those of the immune system. They are categorized functionally as either homeostatic, constitutively produced by tissues for basal levels of cell migration, or inflammatory, where they are generated in association with a pathological inflammatory response. While the extravasation of leukocytes via blood vessels is a key step in cells entering the tissues, the lymphatic vessels also serve as a conduit for cells that are recruited and localized through chemoattractant gradients. Furthermore, the growth and remodeling of lymphatic vessels in pathologies is influenced by chemokines and their receptors expressed by lymphatic endothelial cells (LECs) in and around the pathological tissue. In this review we summarize the diverse role played by specific chemokines and their receptors in shaping the interaction of lymphatic vessels, immune cells, and other pathological cell types in physiology and disease.
Collapse
Affiliation(s)
- Rae H Farnsworth
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Tara Karnezis
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Simon J Maciburko
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Melbourne, VIC, Australia
| | - Steven A Stacker
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
53
|
Zhao Y, Dang Z, Wei R, Gui W, Zhang Y, Chong S. The effects of CCR7 and related signaling pathways on Leishmania major -infected human dendritic cells. J Cell Physiol 2018; 234:13145-13156. [PMID: 30584667 DOI: 10.1002/jcp.27985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVES In our research, we aimed to investigate the roles of CC-chemokine receptor 7 (CCR7) and relevant signaling pathways in Leishmania major-infected human dendritic cells (DCs). METHODS Differentially expressed genes (DEGs) in L. major-infected human DCs were selected out and visualized using R program. Kyoto Encyclopedia of Genes and Genomes pathway analysis was conducted for investigation of significantly enriched signaling pathways and Gene Ontology enrichment analysis was carried out for the unveiling of enriched Molecular Functions and Biological Processes in L. major-infected human DCs. Besides, Hub gene was screened out using weighted gene coexpression network analysis and Cytoscape. In addition, enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction were used for detection of relative expression of CCR7, interleukin-12 (IL-12), and interferon-γ (IFN-γ) in L. major-infected human DCs and western blot analysis was used for detection of relative expression of CCR7 and other proteins in JAK-STAT signaling pathway in L. major-infected human DCs. RESULTS CCR7 was upregulated and both chemokine and JAK-STAT signaling pathway were activated in L. major-infected human DCs. During the L. major infection, total number of L. major-infected human DCs were increased, as well as the relative expression levels of CCR7, IL-12, and IFN-γ and proteins in the JAK-STAT signaling pathway. Overexpression of CCR7 not only increased expression levels of IL-12 and IFN-γ but also activated the JAK-STAT signaling pathway to affect the leishmaniasis progression. CONCLUSION L. major infection-induced activation of CCR7, as well as JAK2 and STAT1, might well upregulate the expression of BAX yet suppress the expression of both Bcl2 and c-Jun to affect leishmaniasis progression.
Collapse
Affiliation(s)
- Yumin Zhao
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Zhisheng Dang
- Key Laboratory on Biology of Parasite and Vector, Parasitic Disease Control Center of China Center for Disease Control and Prevention, Ministry of Health, Shanghai, China
| | - Riming Wei
- Collge of Biotechnology, Guilin Medical University, Guilin, China
| | - Weifeng Gui
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Yishu Zhang
- Department of Parasitology, Guilin Medical University, Guilin, China
| | - Shigui Chong
- School of Nursing, Guilin Medical University, Guilin, China
| |
Collapse
|
54
|
Abstract
Receptor tyrosine kinases (RTKs) are essential components of cell communication pathways utilized from the embryonic to adult stages of life. These transmembrane receptors bind polypeptide ligands, such as growth factors, inducing signalling cascades that control cellular processes such as proliferation, survival, differentiation, motility and inflammation. Many viruses have acquired homologs of growth factors encoded by the hosts that they infect. Production of growth factors during infection allows viruses to exploit RTKs for entry and replication in cells, as well as for host and environmental dissemination. This review describes the genetic diversity amongst virus-derived growth factors and the mechanisms by which RTK exploitation enhances virus survival, then highlights how viral ligands can be used to further understanding of RTK signalling and function during embryogenesis, homeostasis and disease scenarios.
Collapse
Affiliation(s)
- Zabeen Lateef
- a Department of Pharmacology and Toxicology, School of Biomedical Sciences , University of Otago , Dunedin , New Zealand
| | - Lyn M Wise
- a Department of Pharmacology and Toxicology, School of Biomedical Sciences , University of Otago , Dunedin , New Zealand
| |
Collapse
|
55
|
Okuda KS, Lee HM, Velaithan V, Ng MF, Patel V. Utilizing Zebrafish to Identify Anti-(Lymph)Angiogenic Compounds for Cancer Treatment: Promise and Future Challenges. Microcirculation 2018; 23:389-405. [PMID: 27177346 DOI: 10.1111/micc.12289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/11/2016] [Indexed: 12/13/2022]
Abstract
Cancer metastasis which predominantly occurs through blood and lymphatic vessels, is the leading cause of death in cancer patients. Consequently, several anti-angiogenic agents have been approved as therapeutic agents for human cancers such as metastatic renal cell carcinoma. Also, anti-lymphangiogenic drugs such as monoclonal antibodies VGX-100 and IMC-3C5 have undergone phase I clinical trials for advanced and metastatic solid tumors. Although anti-tumor-associated angiogenesis has proven to be a promising therapeutic strategy for human cancers, this approach is fraught with toxicities and development of drug resistance. This emphasizes the need for alternative anti-(lymph)angiogenic drugs. The use of zebrafish has become accepted as an established model for high-throughput screening, vascular biology, and cancer research. Importantly, various zebrafish transgenic lines have now been generated that can readily discriminate different vascular compartments. This now enables detailed in vivo studies that are relevant to both human physiological and tumor (lymph)angiogenesis to be conducted in zebrafish. This review highlights recent advancements in the zebrafish anti-vascular screening platform and showcases promising new anti-(lymph)angiogenic compounds that have been derived from this model. In addition, this review discusses the promises and challenges of the zebrafish model in the context of anti-(lymph)angiogenic compound discovery for cancer treatment.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Hui Mei Lee
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Vithya Velaithan
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Mei Fong Ng
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Vyomesh Patel
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
56
|
Antila S, Karaman S, Nurmi H, Airavaara M, Voutilainen MH, Mathivet T, Chilov D, Li Z, Koppinen T, Park JH, Fang S, Aspelund A, Saarma M, Eichmann A, Thomas JL, Alitalo K. Development and plasticity of meningeal lymphatic vessels. J Exp Med 2017; 214:3645-3667. [PMID: 29141865 PMCID: PMC5716035 DOI: 10.1084/jem.20170391] [Citation(s) in RCA: 311] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 12/25/2022] Open
Abstract
The recent discovery of meningeal lymphatic vessels (LVs) has raised interest in their possible involvement in neuropathological processes, yet little is known about their development or maintenance. We show here that meningeal LVs develop postnatally, appearing first around the foramina in the basal parts of the skull and spinal canal, sprouting along the blood vessels and cranial and spinal nerves to various parts of the meninges surrounding the central nervous system (CNS). VEGF-C, expressed mainly in vascular smooth muscle cells, and VEGFR3 in lymphatic endothelial cells were essential for their development, whereas VEGF-D deletion had no effect. Surprisingly, in adult mice, the LVs showed regression after VEGF-C or VEGFR3 deletion, administration of the tyrosine kinase inhibitor sunitinib, or expression of VEGF-C/D trap, which also compromised the lymphatic drainage function. Conversely, an excess of VEGF-C induced meningeal lymphangiogenesis. The plasticity and regenerative potential of meningeal LVs should allow manipulation of cerebrospinal fluid drainage and neuropathological processes in the CNS.
Collapse
Affiliation(s)
- Salli Antila
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Sinem Karaman
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Harri Nurmi
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Program in Developmental Biology, Institute of Biotechnology, HiLIFE Unit, University of Helsinki, Helsinki, Finland
| | - Merja H Voutilainen
- Program in Developmental Biology, Institute of Biotechnology, HiLIFE Unit, University of Helsinki, Helsinki, Finland
| | - Thomas Mathivet
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France
| | - Dmitri Chilov
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Zhilin Li
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Tapani Koppinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Jun-Hee Park
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Shentong Fang
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Aleksanteri Aspelund
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Program in Developmental Biology, Institute of Biotechnology, HiLIFE Unit, University of Helsinki, Helsinki, Finland
| | - Anne Eichmann
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Jean-Léon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT
- Sorbonne Universités, UPMC Université Paris 06, Institut National de la Santé et de la Recherche Médicale U1127, Centre National de la Recherche Scientifique, AP-HP, Institut du Cerveau et de la Moelle Epinière, Hôpital Pitié-Salpêtrière, Paris, France
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| |
Collapse
|
57
|
Vaahtomeri K, Karaman S, Mäkinen T, Alitalo K. Lymphangiogenesis guidance by paracrine and pericellular factors. Genes Dev 2017; 31:1615-1634. [PMID: 28947496 PMCID: PMC5647933 DOI: 10.1101/gad.303776.117] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This review by Vaahtomeri et al. discusses the mechanisms by which the lymphatic vasculature network is formed, remodeled, and adapted to physiological and pathological challenges. It describes how the lymphatic vasculature network is controlled by an intricate balance of growth factors and biomechanical cues. Lymphatic vessels are important for tissue fluid homeostasis, lipid absorption, and immune cell trafficking and are involved in the pathogenesis of several human diseases. The mechanisms by which the lymphatic vasculature network is formed, remodeled, and adapted to physiological and pathological challenges are controlled by an intricate balance of growth factor and biomechanical cues. These transduce signals for the readjustment of gene expression and lymphatic endothelial migration, proliferation, and differentiation. In this review, we describe several of these cues and how they are integrated for the generation of functional lymphatic vessel networks.
Collapse
Affiliation(s)
- Kari Vaahtomeri
- Wihuri Research Institute, Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| | - Sinem Karaman
- Wihuri Research Institute, Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| | - Taija Mäkinen
- Department of Immunology, Genetics, and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Kari Alitalo
- Wihuri Research Institute, Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
58
|
Hogan BM, Schulte-Merker S. How to Plumb a Pisces: Understanding Vascular Development and Disease Using Zebrafish Embryos. Dev Cell 2017; 42:567-583. [PMID: 28950100 DOI: 10.1016/j.devcel.2017.08.015] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/01/2017] [Accepted: 08/21/2017] [Indexed: 01/09/2023]
Abstract
Our vasculature plays diverse and critical roles in homeostasis and disease. In recent decades, the use of zebrafish has driven our understanding of vascular development into new areas, identifying new genes and mechanisms controlling vessel formation and allowing unprecedented observation of the cellular and molecular events that shape the developing vasculature. Here, we highlight key mechanisms controlling formation of the zebrafish vasculature and investigate how knowledge from this highly tractable model system has informed our understanding of vascular disease in humans.
Collapse
Affiliation(s)
- Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, St Lucia, Brisbane, QLD 4072, Australia.
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster 48149, Germany; Cells-in-Motion Cluster of Excellence (EXC-1003), WWU Münster, 48149 Münster, Germany.
| |
Collapse
|
59
|
Gβ1 is required for neutrophil migration in zebrafish. Dev Biol 2017; 428:135-147. [PMID: 28554852 DOI: 10.1016/j.ydbio.2017.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 11/20/2022]
Abstract
Signaling mediated by G protein-coupled receptors (GPCRs) is essential for the migration of cells toward chemoattractants. The recruitment of neutrophils to injured tissues in zebrafish larvae is a useful model for studying neutrophil migration and trafficking in vivo. Indeed, the study of this process led to the discovery that PI3Kγ is required for the polarity and motility of neutrophils, features that are necessary for the directed migration of these cells to wounds. However, the mechanism by which PI3Kγ is activated remains to be determined. Here we show that signaling by specifically the heterotrimeric G protein subunit Gβ1 is critical for neutrophil migration in response to wounding. In embryos treated with small-molecule inhibitors of Gβγ signaling, neutrophils failed to migrate to wound sites. Although both the Gβ1 and Gβ4 isoforms are expressed in migrating neutrophils, only deficiency for the former (morpholino-based knockdown) interfered with the directed migration of neutrophils towards wounds. The Gβ1 deficiency also impaired the ability of cells to change cell shape and reduced their general motility, defects that are similar to those in neutrophils deficient for PI3Kγ. Transplantation assays showed that the requirement for Gβ1 in neutrophil migration is cell autonomous. Finally, live imaging revealed that Gβ1 is required for polarized activation of PI3K, and for the actin dynamics that enable neutrophil migration. Collectively, our data indicate that Gβ1 signaling controls proper neutrophil migration by activating PI3K and modulating actin dynamics. Moreover, they illustrate a role for a specific Gβ isoform in chemotaxis in vivo.
Collapse
|
60
|
Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cell Biol 2017; 18:477-494. [PMID: 28537573 DOI: 10.1038/nrm.2017.36] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Blood and lymphatic vessels pervade almost all body tissues and have numerous essential roles in physiology and disease. The inner lining of these networks is formed by a single layer of endothelial cells, which is specialized according to the needs of the tissue that it supplies. Whereas the general mechanisms of blood and lymphatic vessel development are being defined with increasing molecular precision, studies of the processes of endothelial specialization remain mostly descriptive. Recent insights from genetic animal models illuminate how endothelial cells interact with each other and with their tissue environment, providing paradigms for vessel type- and organ-specific endothelial differentiation. Delineating these governing principles will be crucial for understanding how tissues develop and maintain, and how their function becomes abnormal in disease.
Collapse
|
61
|
Jung HM, Castranova D, Swift MR, Pham VN, Venero Galanternik M, Isogai S, Butler MG, Mulligan TS, Weinstein BM. Development of the larval lymphatic system in zebrafish. Development 2017; 144:2070-2081. [PMID: 28506987 PMCID: PMC5482986 DOI: 10.1242/dev.145755] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/24/2017] [Indexed: 12/16/2022]
Abstract
The lymphatic vascular system is a hierarchically organized complex network essential for tissue fluid homeostasis, immune trafficking and absorption of dietary fats in the human body. Despite its importance, the assembly of the lymphatic network is still not fully understood. The zebrafish is a powerful model organism that enables study of lymphatic vessel development using high-resolution imaging and sophisticated genetic and experimental manipulation. Although several studies have described early lymphatic development in the fish, lymphatic development at later stages has not been completely elucidated. In this study, we generated a new Tg(mrc1a:egfp)y251 transgenic zebrafish that uses a mannose receptor, C type 1 (mrc1a) promoter to drive strong EGFP expression in lymphatic vessels at all stages of development and in adult zebrafish. We used this line to describe the assembly of the major vessels of the trunk lymphatic vascular network, including the later-developing collateral cardinal, spinal, superficial lateral and superficial intersegmental lymphatics. Our results show that major trunk lymphatic vessels are conserved in the zebrafish, and provide a thorough and complete description of trunk lymphatic vessel assembly.
Collapse
Affiliation(s)
- Hyun Min Jung
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew R Swift
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sumio Isogai
- Department of Anatomy, School of Medicine, Iwate Medical University, Morioka 020-8505, Japan
| | - Matthew G Butler
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy S Mulligan
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
62
|
Wertheim KY, Roose T. A Mathematical Model of Lymphangiogenesis in a Zebrafish Embryo. Bull Math Biol 2017; 79:693-737. [PMID: 28233173 PMCID: PMC5501200 DOI: 10.1007/s11538-017-0248-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/19/2017] [Indexed: 01/18/2023]
Abstract
The lymphatic system of a vertebrate is important in health and diseases. We propose a novel mathematical model to elucidate the lymphangiogenic processes in zebrafish embryos. Specifically, we are interested in the period when lymphatic endothelial cells (LECs) exit the posterior cardinal vein and migrate to the horizontal myoseptum of a zebrafish embryo. We wonder whether vascular endothelial growth factor C (VEGFC) is a morphogen and a chemotactic factor for these LECs. The model considers the interstitial flow driving convection, the reactive transport of VEGFC, and the changing dynamics of the extracellular matrix in the embryo. Simulations of the model illustrate that VEGFC behaves very differently in diffusion and convection-dominant scenarios. In the former case, it must bind to the matrix to establish a functional morphogen gradient. In the latter case, the opposite is true and the pressure field is the key determinant of what VEGFC may do to the LECs. Degradation of collagen I, a matrix component, by matrix metallopeptidase 2 controls the spatiotemporal dynamics of VEGFC. It controls whether diffusion or convection is dominant in the embryo; it can create channels of abundant VEGFC and scarce collagen I to facilitate lymphangiogenesis; when collagen I is insufficient, VEGFC cannot influence the LECs at all. We predict that VEGFC is a morphogen for the migrating LECs, but it is not a chemotactic factor for them.
Collapse
Affiliation(s)
- Kenneth Y. Wertheim
- Faculty of Engineering and the Environment, University of Southampton, Highfield Campus, Southampton, SO17 1BJ UK
| | - Tiina Roose
- Faculty of Engineering and the Environment, University of Southampton, Highfield Campus, Southampton, SO17 1BJ UK
| |
Collapse
|
63
|
Torraca V, Tulotta C, Snaar-Jagalska BE, Meijer AH. The chemokine receptor CXCR4 promotes granuloma formation by sustaining a mycobacteria-induced angiogenesis programme. Sci Rep 2017; 7:45061. [PMID: 28332618 PMCID: PMC5362882 DOI: 10.1038/srep45061] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/20/2017] [Indexed: 12/14/2022] Open
Abstract
CXC chemokine receptor 4 plays a critical role in chemotaxis and leukocyte differentiation. Furthermore, there is increasing evidence that links this receptor to angiogenesis. Using the well-established zebrafish-Mycobacterium marinum model for tuberculosis, angiogenesis was recently found to be important for the development of cellular aggregates called granulomas that contain the mycobacteria and are the hallmark of tuberculosis disease. Here, we found that initiation of the granuloma-associated proangiogenic programme requires CXCR4 signalling. The nascent granulomas in cxcr4b-deficient zebrafish embryos were poorly vascularised, which in turn also delayed bacterial growth. Suppressed infection expansion in cxcr4b mutants could not be attributed to an overall deficient recruitment of leukocytes or to different intramacrophage bacterial growth rate, as cxcr4b mutants displayed similar microbicidal capabilities against initial mycobacterial infection and the cellular composition of granulomatous lesions was similar to wildtype siblings. Expression of vegfaa was upregulated to a similar extent in cxcr4b mutants and wildtypes, suggesting that the granuloma vascularisation phenotype of cxcr4b mutants is independent of vascular endothelial growth factor.
Collapse
|
64
|
Karpanen T, Padberg Y, van de Pavert SA, Dierkes C, Morooka N, Peterson-Maduro J, van de Hoek G, Adrian M, Mochizuki N, Sekiguchi K, Kiefer F, Schulte D, Schulte-Merker S. An Evolutionarily Conserved Role for Polydom/Svep1 During Lymphatic Vessel Formation. Circ Res 2017; 120:1263-1275. [PMID: 28179432 PMCID: PMC5389596 DOI: 10.1161/circresaha.116.308813] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 01/03/2023]
Abstract
Supplemental Digital Content is available in the text. Rationale: Lymphatic vessel formation and function constitutes a physiologically and pathophysiologically important process, but its genetic control is not well understood. Objective: Here, we identify the secreted Polydom/Svep1 protein as essential for the formation of the lymphatic vasculature. We analyzed mutants in mice and zebrafish to gain insight into the role of Polydom/Svep1 in the lymphangiogenic process. Methods and Results: Phenotypic analysis of zebrafish polydom/svep1 mutants showed a decrease in venous and lymphovenous sprouting, which leads to an increased number of intersegmental arteries. A reduced number of primordial lymphatic cells populated the horizontal myoseptum region but failed to migrate dorsally or ventrally, resulting in severe reduction of the lymphatic trunk vasculature. Corresponding mutants in the mouse Polydom/Svep1 gene showed normal egression of Prox-1+ cells from the cardinal vein at E10.5, but at E12.5, the tight association between the cardinal vein and lymphatic endothelial cells at the first lymphovenous contact site was abnormal. Furthermore, mesenteric lymphatic structures at E18.5 failed to undergo remodeling events in mutants and lacked lymphatic valves. In both fish and mouse embryos, the expression of the gene suggests a nonendothelial and noncell autonomous mechanism. Conclusions: Our data identify zebrafish and mouse Polydom/Svep1 as essential extracellular factors for lymphangiogenesis. Expression of the respective genes by mesenchymal cells in intimate proximity with venous and lymphatic endothelial cells is required for sprouting and migratory events in zebrafish and for remodeling events of the lymphatic intraluminal valves in mouse embryos.
Collapse
Affiliation(s)
- Terhi Karpanen
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.).
| | - Yvonne Padberg
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Serge A van de Pavert
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Cathrin Dierkes
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Nanami Morooka
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Josi Peterson-Maduro
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Glenn van de Hoek
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Max Adrian
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Naoki Mochizuki
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Kiyotoshi Sekiguchi
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Friedemann Kiefer
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Dörte Schulte
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.)
| | - Stefan Schulte-Merker
- From the Hubrecht Institute, KNAW and UMC Utrecht, Utrecht, the Netherlands (T.K., S.A.v.d.P., J.P.-M., G.v.d.H., M.A., S.S.-M.); Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Germany (Y.P., D.S., S.S.-M.); CiM Cluster of Excellence (EXC 1003-CiM), Münster, Germany (Y.P., D.S., S.S.-M.); Max Planck Institute for Molecular Biomedicine, Münster, Germany (C.D., F.K.); Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Suita, Japan (N.M., K.S.); and Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan (N.M.).
| |
Collapse
|
65
|
Thymosin β4 Improves Differentiation and Vascularization of EHTs. Stem Cells Int 2017; 2017:6848271. [PMID: 28191018 PMCID: PMC5278226 DOI: 10.1155/2017/6848271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/07/2016] [Indexed: 01/14/2023] Open
Abstract
Induced pluripotent stem cells (iPSC) constitute a powerful tool to study cardiac physiology and represents a promising treatment strategy to tackle cardiac disease. However, iPSCs remain relatively immature after differentiation. Additionally, engineered heart tissue (EHT) has been investigated as a therapy option in preclinical disease models with promising results, although their vascularization and functionality leave room for improvement. Thymosin β4 (Tβ4) has been shown to promote the differentiation of progenitor cell lines to cardiomyocytes while it also induces angiogenic sprouting and vascular maturation. We examined the potential impact of Tβ4 to enhance maturation of cardiomyocytes from iPSCs. Assessing the expression of transcription factors associated with cardiac differentiation, we were able to demonstrate the increased generation of cells displaying cardiomyocyte characteristics in vitro. Furthermore, we demonstrated, in a zebrafish model of embryonic vascular development, that Tβ4 is crucial for the proper execution of lymphatic and angiogenic vessel sprouting. Finally, utilizing Tβ4-transduced EHTs generated from mice genetically engineered to label endothelial cells in vitro, we show that treatment with Tβ4 promotes vascularization and contractility in EHTs, highlighting Tβ4 as a growth factor improving the formation of cardiomyocytes from iPSC and enhancing the performance of EHTs generated from neonatal cardiomyocytes.
Collapse
|
66
|
Bower NI, Vogrin AJ, Le Guen L, Chen H, Stacker SA, Achen MG, Hogan BM. Vegfd modulates both angiogenesis and lymphangiogenesis during zebrafish embryonic development. Development 2017; 144:507-518. [PMID: 28087639 DOI: 10.1242/dev.146969] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factors (VEGFs) control angiogenesis and lymphangiogenesis during development and in pathological conditions. In the zebrafish trunk, Vegfa controls the formation of intersegmental arteries by primary angiogenesis and Vegfc is essential for secondary angiogenesis, giving rise to veins and lymphatics. Vegfd has been largely thought of as dispensable for vascular development in vertebrates. Here, we generated a zebrafish vegfd mutant by genome editing. vegfd mutants display significant defects in facial lymphangiogenesis independent of vegfc function. Strikingly, we find that vegfc and vegfd cooperatively control lymphangiogenesis throughout the embryo, including during the formation of the trunk lymphatic vasculature. Interestingly, we find that vegfd and vegfc also redundantly drive artery hyperbranching phenotypes observed upon depletion of Flt1 or Dll4. Epistasis and biochemical binding assays suggest that, during primary angiogenesis, Vegfd influences these phenotypes through Kdr (Vegfr2) rather than Flt4 (Vegfr3). These data demonstrate that, rather than being dispensable during development, Vegfd plays context-specific indispensable and also compensatory roles during both blood vessel angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Neil I Bower
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Adam J Vogrin
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Ludovic Le Guen
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Huijun Chen
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marc G Achen
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
67
|
Padberg Y, Schulte-Merker S, van Impel A. The lymphatic vasculature revisited-new developments in the zebrafish. Methods Cell Biol 2016; 138:221-238. [PMID: 28129845 DOI: 10.1016/bs.mcb.2016.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The lymphatic system is lined by endothelial cells and part of the vasculature. It is essential for tissue fluid homeostasis, absorption of dietary fats, and immune surveillance in vertebrates. Misregulation of lymphatic vessel formation and dysfunction of the lymphatic system have been indicated in a number of pathological conditions including lymphedema formation, obesity or chronic inflammatory diseases such as rheumatoid arthritis. In zebrafish, lymphatics were discovered about 10years ago, and the underlying molecular pathways involved in its development have since been studied in detail. Due to its superior live cell imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study the development of the lymphatic system during early embryonic development. In the current review, we will focus on the key players during zebrafish lymphangiogenesis and compare the roles of these genes to their mammalian counterparts. In particular, we will focus on novel findings that shed new light on the molecular mechanisms of lymphatic cell fate specification, as well as sprouting and migration of lymphatic precursor cells.
Collapse
Affiliation(s)
- Y Padberg
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC M 1003-CiM), University of Münster, Münster, Germany
| | - S Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC M 1003-CiM), University of Münster, Münster, Germany
| | - A van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC M 1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
68
|
Munger SJ, Davis MJ, Simon AM. Defective lymphatic valve development and chylothorax in mice with a lymphatic-specific deletion of Connexin43. Dev Biol 2016; 421:204-218. [PMID: 27899284 DOI: 10.1016/j.ydbio.2016.11.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022]
Abstract
Lymphatic valves (LVs) are cusped luminal structures that permit the movement of lymph in only one direction and are therefore critical for proper lymphatic vessel function. Congenital valve aplasia or agenesis can, in some cases, be a direct cause of lymphatic disease. Knowledge about the molecular mechanisms operating during the development and maintenance of LVs may thus aid in the establishment of novel therapeutic approaches to treat lymphatic disorders. In this study, we examined the role of Connexin43 (Cx43), a gap junction protein expressed in lymphatic endothelial cells (LECs), during valve development. Mouse embryos with a null mutation in Cx43 (Gja1) were previously shown to completely lack mesenteric LVs at embryonic day 18. However, interpreting the phenotype of Cx43-/- mice was complicated by the fact that global deletion of Cx43 causes perinatal death due to heart defects during embryogenesis. We have now generated a mouse model (Cx43∆LEC) with a lymphatic-specific ablation of Cx43 and show that the absence of Cx43 in LECs causes a delay (rather than a complete block) in LV initiation, an increase in immature valves with incomplete leaflet elongation, a reduction in the total number of valves, and altered lymphatic capillary patterning. The physiological consequences of these lymphatic changes were leaky valves, insufficient lymph transport and reflux, and a high incidence of lethal chylothorax. These results demonstrate that the expression of Cx43 is specifically required in LECs for normal development of LVs.
Collapse
Affiliation(s)
| | - Michael J Davis
- Dept. of Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO, USA.
| | - Alexander M Simon
- Department of Physiology, University of Arizona, Tucson AZ 85724, USA.
| |
Collapse
|
69
|
Liu X, Uemura A, Fukushima Y, Yoshida Y, Hirashima M. Semaphorin 3G Provides a Repulsive Guidance Cue to Lymphatic Endothelial Cells via Neuropilin-2/PlexinD1. Cell Rep 2016; 17:2299-2311. [DOI: 10.1016/j.celrep.2016.11.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/12/2016] [Accepted: 10/28/2016] [Indexed: 11/24/2022] Open
|
70
|
Chen J, Zhu RF, Li FF, Liang YL, Wang C, Qin YW, Huang S, Zhao XX, Jing Q. MicroRNA-126a Directs Lymphangiogenesis Through Interacting With Chemokine and Flt4 Signaling in Zebrafish. Arterioscler Thromb Vasc Biol 2016; 36:2381-2393. [PMID: 27789478 DOI: 10.1161/atvbaha.116.308120] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/07/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE MicroRNA-126 (miR-126) is an endothelium-enriched miRNA and functions in vascular integrity and angiogenesis. The application of miRNA as potential biomarker and therapy target has been widely investigated in various pathological processes. However, its role in lymphatic diseases had not been widely explored. We aimed to reveal the role of miR-126 in lymphangiogenesis and the regulatory signaling pathways for potential targets of therapy. APPROACH AND RESULTS Loss-of-function studies using morpholino oligonucleotides and CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) system showed that silencing of miR-126a severely affected the formation of parachordal lymphangioblasts and thoracic duct in zebrafish embryos, although their development in miR-126b knockdown embryos was normal. Expression analyses by in situ hybridization and immunofluorescence indicated that miR-126a was expressed in lymphatic vessels, as well as in blood vessels. Time-lapse confocal imaging assay further revealed that knockdown of miR-126a blocked both lymphangiogenic sprouts budding from the posterior cardinal vein and lymphangioblasts extension along horizontal myoseptum. Bioinformatics analysis and in vivo report assay identified that miR-126a upregulated Cxcl12a by targeting its 5' untranslated region. Moreover, loss- and gain-of-function studies revealed that Cxcl12a signaling acted downstream of miR-126a during parachordal lymphangioblast extension, whereby Flt4 signaling acts as a cooperator of miR-126a, allowing it to modulate lymphangiogenic sprout formation. CONCLUSIONS These findings demonstrate that miR-126a directs lymphatic endothelial cell sprouting and extension by interacting with Cxcl12a-mediated chemokine signaling and Vegfc-Flt4 signal axis. Our results suggest that these key regulators of lymphangiogenesis may be involved in lymphatic pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Jian Chen
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Rong-Fang Zhu
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Fang-Fang Li
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Yu-Lai Liang
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Chen Wang
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Yong-Wen Qin
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Shuang Huang
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Xian-Xian Zhao
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.)
| | - Qing Jing
- From the Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, China (J.C., R.-F.Z., F.-F.L., Y.-L.L., C.W., Q.J.); and Department of Cardiology, Changhai Hospital, Shanghai, China (Y.-W.Q., S.H., X.-X.Z., Q.J.).
| |
Collapse
|
71
|
Nagasawa-Masuda A, Terai K. ERK activation in endothelial cells is a novel marker during neovasculogenesis. Genes Cells 2016; 21:1164-1175. [PMID: 27696620 DOI: 10.1111/gtc.12438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 08/16/2016] [Indexed: 01/01/2023]
Abstract
Vasculogenesis is essential during early development to construct networks transporting oxygen, blood and nutrients. Tip and stalk cells are specialized endothelial cells involved in novel vessel formation because of their behavior such as sprouting as a leading cell and following tip cell. However, the spatiotemporal details determining the emergence of these cells are unknown. Here, we first show that the ERK activity in endothelial cells represents the precursor of tip and stalk cells for vasculogenesis in zebrafish. We identified that tip and stalk cells for intersegmental vessel (ISV) formation were already specialized in the dorsal aorta (DA) before sprouting. Furthermore, similar specialization was observed in tip cells during parachordal vessel (PAV) formation in lymphangiogenesis. We also identified that the ERK activity was required for specialized cells to emerge from existing blood vessels. Our data show that the ERK activity is a novel marker for determining the emergence of cells in both angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Ayumi Nagasawa-Masuda
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kenta Terai
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-0032, Japan
| |
Collapse
|
72
|
Venero Galanternik M, Stratman AN, Jung HM, Butler MG, Weinstein BM. Building the drains: the lymphatic vasculature in health and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:689-710. [PMID: 27576003 DOI: 10.1002/wdev.246] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023]
Abstract
The lymphatic vasculature is comprised of a network of endothelial vessels found in close proximity to but separated from the blood vasculature. An essential tissue component of all vertebrates, lymphatics are responsible for the maintenance of fluid homeostasis, dissemination of immune cells, and lipid reabsorption under healthy conditions. When lymphatic vessels are impaired due to invasive surgery, genetic disorders, or parasitic infections, severe fluid build-up accumulates in the affected tissues causing a condition known as lymphedema. Malignant tumors can also directly activate lymphangiogenesis and use these vessels to promote the spread of metastatic cells. Although their first description goes back to the times of Hippocrates, with subsequent anatomical characterization at the beginning of the 20th-century, the lack of identifying molecular markers and tools to visualize these translucent vessels meant that investigation of lymphatic vessels fell well behind research of blood vessels. However, after years under the shadow of the blood vasculature, recent advances in imaging technologies and new genetic and molecular tools have accelerated the pace of research on lymphatic vessel development. These new tools have facilitated both work in classical mammalian models and the emergence of new powerful vertebrate models like zebrafish, quickly driving the field of lymphatic development back into the spotlight. In this review, we summarize the highlights of recent research on the development and function of the lymphatic vascular network in health and disease. WIREs Dev Biol 2016, 5:689-710. doi: 10.1002/wdev.246 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Marina Venero Galanternik
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Amber N Stratman
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hyun Min Jung
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Matthew G Butler
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Brant M Weinstein
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
73
|
Semo J, Nicenboim J, Yaniv K. Development of the lymphatic system: new questions and paradigms. Development 2016; 143:924-35. [PMID: 26980792 DOI: 10.1242/dev.132431] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The lymphatic system is a blind-ended network of vessels that plays important roles in mediating tissue fluid homeostasis, intestinal lipid absorption and the immune response. A profound understanding of the development of lymphatic vessels, as well as of the molecular cues governing their formation and morphogenesis, might prove essential for our ability to treat lymphatic-related diseases. The embryonic origins of lymphatic vessels have been debated for over a century, with a model claiming a venous origin for the lymphatic endothelium being predominant. However, recent studies have provided new insights into the origins of lymphatic vessels. Here, we review the molecular mechanisms controlling lymphatic specification and sprouting, and we discuss exciting findings that shed new light on previously uncharacterized sources of lymphatic endothelial cells.
Collapse
Affiliation(s)
- Jonathan Semo
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Julian Nicenboim
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
74
|
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are uniquely required to balance the formation of new blood vessels with the maintenance and remodelling of existing ones, during development and in adult tissues. Recent advances have greatly expanded our understanding of the tight and multi-level regulation of VEGFR2 signalling, which is the primary focus of this Review. Important insights have been gained into the regulatory roles of VEGFR-interacting proteins (such as neuropilins, proteoglycans, integrins and protein tyrosine phosphatases); the dynamics of VEGFR2 endocytosis, trafficking and signalling; and the crosstalk between VEGF-induced signalling and other endothelial signalling cascades. A clear understanding of this multifaceted signalling web is key to successful therapeutic suppression or stimulation of vascular growth.
Collapse
|
75
|
Betz C, Lenard A, Belting HG, Affolter M. Cell behaviors and dynamics during angiogenesis. Development 2016; 143:2249-60. [DOI: 10.1242/dev.135616] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022]
Abstract
Vascular networks are formed and maintained through a multitude of angiogenic processes, such as sprouting, anastomosis and pruning. Only recently has it become possible to study the behavior of the endothelial cells that contribute to these networks at a single-cell level in vivo. This Review summarizes what is known about endothelial cell behavior during developmental angiogenesis, focusing on the morphogenetic changes that these cells undergo.
Collapse
Affiliation(s)
- Charles Betz
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, Basel CH-4056, Switzerland
| | - Anna Lenard
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, Basel CH-4056, Switzerland
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, Basel CH-4056, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, Basel CH-4056, Switzerland
| |
Collapse
|
76
|
Buttler K, Lohrberg M, Gross G, Weich HA, Wilting J. Integration of CD45-positive leukocytes into newly forming lymphatics of adult mice. Histochem Cell Biol 2016; 145:629-36. [PMID: 26748643 PMCID: PMC4848334 DOI: 10.1007/s00418-015-1399-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 10/28/2022]
Abstract
The embryonic origin of lymphatic endothelial cells (LECs) has been a matter of controversy since more than a century. However, recent studies in mice have supported the concept that embryonic lymphangiogenesis is a complex process consisting of growth of lymphatics from specific venous segments as well as the integration of lymphangioblasts into the lymphatic networks. Similarly, the mechanisms of adult lymphangiogenesis are poorly understood and have rarely been studied. We have recently shown that endothelial progenitor cells isolated from the lung of adult mice have the capacity to form both blood vessels and lymphatics when grafted with Matrigel plugs into the skin of syngeneic mice. Here, we followed up on these experiments and studied the behavior of host leukocytes during lymphangiogenesis in the Matrigel plugs. We observed a striking co-localization of CD45(+) leukocytes with the developing lymphatics. Numerous CD45(+) cells expressed the LEC marker podoplanin and were obviously integrated into the lining of lymphatic capillaries. This indicates that, similar to inflammation-induced lymphangiogenesis in man, circulating CD45(+) cells of adult mice are capable of initiating lymphangiogenesis and of adopting a lymphvasculogenic cellular differentiation program. The data are discussed in the context of embryonic and inflammation-induced lymphangiogenesis.
Collapse
Affiliation(s)
- K Buttler
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| | - M Lohrberg
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| | - G Gross
- Department of Gene Regulation, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - H A Weich
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - J Wilting
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany.
| |
Collapse
|
77
|
Tulotta C, Stefanescu C, Beletkaia E, Bussmann J, Tarbashevich K, Schmidt T, Snaar-Jagalska BE. Inhibition of signaling between human CXCR4 and zebrafish ligands by the small molecule IT1t impairs the formation of triple-negative breast cancer early metastases in a zebrafish xenograft model. Dis Model Mech 2016; 9:141-53. [PMID: 26744352 PMCID: PMC4770151 DOI: 10.1242/dmm.023275] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/25/2015] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and recurrent type of breast carcinoma that is associated with poor patient prognosis. Because of the limited efficacy of current treatments, new therapeutic strategies need to be developed. The CXCR4-CXCL12 chemokine signaling axis guides cell migration in physiological and pathological processes, including breast cancer metastasis. Although targeted therapies to inhibit the CXCR4-CXCL12 axis are under clinical experimentation, still no effective therapeutic approaches have been established to block CXCR4 in TNBC. To unravel the role of the CXCR4-CXCL12 axis in the formation of TNBC early metastases, we used the zebrafish xenograft model. Importantly, we demonstrate that cross-communication between the zebrafish and human ligands and receptors takes place and human tumor cells expressing CXCR4 initiate early metastatic events by sensing zebrafish cognate ligands at the metastatic site. Taking advantage of the conserved intercommunication between human tumor cells and the zebrafish host, we blocked TNBC early metastatic events by chemical and genetic inhibition of CXCR4 signaling. We used IT1t, a potent CXCR4 antagonist, and show for the first time its promising anti-tumor effects. In conclusion, we confirm the validity of the zebrafish as a xenotransplantation model and propose a pharmacological approach to target CXCR4 in TNBC. Summary: CXCR4-expressing human tumor cells respond to zebrafish cognate ligands and initiate metastatic events in a zebrafish xenograft model. The CXCR4 antagonist IT1t has promising tumor inhibitory effects.
Collapse
Affiliation(s)
- Claudia Tulotta
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Cristina Stefanescu
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elena Beletkaia
- Physics of Life Processes, Kamerligh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, Leiden 2333 CA, The Netherlands
| | - Jeroen Bussmann
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | | | - Thomas Schmidt
- Physics of Life Processes, Kamerligh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, Leiden 2333 CA, The Netherlands
| | - B Ewa Snaar-Jagalska
- Institute of Biology, Animal Sciences and Health, Gorlaeus Laboratories, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
78
|
Astin JW, Crosier PS. Lymphatics, Cancer and Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:199-218. [DOI: 10.1007/978-3-319-30654-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
79
|
Bowers S, Norden P, Davis G. Molecular Signaling Pathways Controlling Vascular Tube Morphogenesis and Pericyte-Induced Tube Maturation in 3D Extracellular Matrices. ADVANCES IN PHARMACOLOGY 2016; 77:241-80. [DOI: 10.1016/bs.apha.2016.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
80
|
Koltowska K, Paterson S, Bower NI, Baillie GJ, Lagendijk AK, Astin JW, Chen H, Francois M, Crosier PS, Taft RJ, Simons C, Smith KA, Hogan BM. mafba is a downstream transcriptional effector of Vegfc signaling essential for embryonic lymphangiogenesis in zebrafish. Genes Dev 2015; 29:1618-30. [PMID: 26253536 PMCID: PMC4536310 DOI: 10.1101/gad.263210.115] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Koltowska et al. used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for lymphatic vessel development. Vegfc signaling increases mafba expression to control downstream transcription, and this relationship is SoxF transcription factor-dependent. The lymphatic vasculature plays roles in tissue fluid balance, immune cell trafficking, fatty acid absorption, cancer metastasis, and cardiovascular disease. Lymphatic vessels form by lymphangiogenesis, the sprouting of new lymphatics from pre-existing vessels, in both development and disease contexts. The apical signaling pathway in lymphangiogenesis is the VEGFC/VEGFR3 pathway, yet how signaling controls cellular transcriptional output remains unknown. We used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for lymphatic vessel development. We found that mafba is required for the migration of lymphatic precursors after their initial sprouting from the posterior cardinal vein. mafba expression is enriched in sprouts emerging from veins, and we show that mafba functions cell-autonomously during lymphatic vessel development. Mechanistically, Vegfc signaling increases mafba expression to control downstream transcription, and this regulatory relationship is dependent on the activity of SoxF transcription factors, which are essential for mafba expression in venous endothelium. Here we identify an indispensable Vegfc–SoxF–Mafba pathway in lymphatic development.
Collapse
Affiliation(s)
- Katarzyna Koltowska
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Scott Paterson
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Neil I Bower
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Gregory J Baillie
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Huijun Chen
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Mathias Francois
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Ryan J Taft
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Cas Simons
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Kelly A Smith
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
81
|
Chemokine-guided angiogenesis directs coronary vasculature formation in zebrafish. Dev Cell 2015; 33:442-54. [PMID: 26017769 DOI: 10.1016/j.devcel.2015.04.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 10/20/2014] [Accepted: 04/01/2015] [Indexed: 02/02/2023]
Abstract
Interruption of the coronary blood supply severely impairs heart function with often fatal consequences for patients. However, the formation and maturation of these coronary vessels is not fully understood. Here we provide a detailed analysis of coronary vessel development in zebrafish. We observe that coronary vessels form in zebrafish by angiogenic sprouting of arterial cells derived from the endocardium at the atrioventricular canal. Endothelial cells express the CXC-motif chemokine receptor Cxcr4a and migrate to vascularize the ventricle under the guidance of the myocardium-expressed ligand Cxcl12b. cxcr4a mutant zebrafish fail to form a vascular network, whereas ectopic expression of Cxcl12b ligand induces coronary vessel formation. Importantly, cxcr4a mutant zebrafish fail to undergo heart regeneration following injury. Our results suggest that chemokine signaling has an essential role in coronary vessel formation by directing migration of endocardium-derived endothelial cells. Poorly developed vasculature in cxcr4a mutants likely underlies decreased regenerative potential in adults.
Collapse
|
82
|
Omran DA, Zaki MM, Hasan SF, Shousha HI. Diagnostic value of vascular endothelial growth factor and interleukin-17 in association with molecular diagnosis of Wuchereria bancrofti infection. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2015; 5:600-603. [DOI: 10.1016/s2222-1808(15)60895-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
83
|
Kimura E, Isogai S, Hitomi J. Integration of vascular systems between the brain and spinal cord in zebrafish. Dev Biol 2015; 406:40-51. [PMID: 26234750 DOI: 10.1016/j.ydbio.2015.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/08/2015] [Accepted: 07/15/2015] [Indexed: 11/28/2022]
Abstract
Cerebral and spinal vascular systems are organized individually, and they then conjugate at their border, through the integration of basilar artery and vertebral arteries. Zebrafish (Danio rerio) is an ideal organism for studying early vascular development, and the precise procedure of cranial and truncal vascular formation has been previously demonstrated using this model. However, the stepwise process of the integration between the brain and spinal cord has not been clearly elucidated. In this study, we describe the integration of the independent vascular systems for the brain and spinal cord, using transgenic zebrafish expressing enhanced green fluorescent protein in endothelial cells. Initially, basilar artery and primordial hindbrain channels, into which internal carotid arteries supplied blood, were connected with dorsal longitudinal anastomose vessels, via the first intersegmental artery. This initial connection was not influenced by flow dynamics, suggesting that vascular integration in this region is controlled by genetic cues. Vertebral arteries were formed individually as longitudinal vessels beneath the spinal cord, and became integrated with the basilar artery during subsequent remodeling. Furthermore, we confirmed the basal vasculature was well conserved in adult zebrafish. Observations of vascular integration presented herein will contribute to an understanding of regulatory mechanisms behind this process.
Collapse
Affiliation(s)
- Eiji Kimura
- Department of Anatomy, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa, Iwate 028-3694, Japan.
| | - Sumio Isogai
- Department of Anatomy, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa, Iwate 028-3694, Japan
| | - Jiro Hitomi
- Department of Anatomy, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa, Iwate 028-3694, Japan
| |
Collapse
|
84
|
Bussmann J, Raz E. Chemokine-guided cell migration and motility in zebrafish development. EMBO J 2015; 34:1309-18. [PMID: 25762592 DOI: 10.15252/embj.201490105] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/04/2015] [Indexed: 12/29/2022] Open
Abstract
Chemokines are vertebrate-specific, structurally related proteins that function primarily in controlling cell movements by activating specific 7-transmembrane receptors. Chemokines play critical roles in a large number of biological processes and are also involved in a range of pathological conditions. For these reasons, chemokines are at the focus of studies in developmental biology and of clinically oriented research aimed at controlling cancer, inflammation, and immunological diseases. The small size of the zebrafish embryos, their rapid external development, and optical properties as well as the large number of eggs and the fast expansion in genetic tools available make this model an extremely useful one for studying the function of chemokines and chemokine receptors in an in vivo setting. Here, we review the findings relevant to the role that chemokines play in the context of directed single-cell migration, primarily in neutrophils and germ cells, and compare it to the collective cell migration of the zebrafish lateral line. We present the current knowledge concerning the formation of the chemokine gradient, its interpretation within the cell, and the molecular mechanisms underlying the cellular response to chemokine signals during directed migration.
Collapse
Affiliation(s)
- Jeroen Bussmann
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands Gorlaeus Laboratories, Department of Molecular Cell Biology, Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| |
Collapse
|
85
|
Yang Y, Enis D, Zheng H, Chia S, Yang J, Chen M, Dhillon V, Papayannapoulou T, Kahn ML. Cell Adhesion Mediated by VCAM-ITGα9 Interactions Enables Lymphatic Development. Arterioscler Thromb Vasc Biol 2015; 35:1179-89. [PMID: 25745057 DOI: 10.1161/atvbaha.114.304997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/22/2015] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Adhesive ligand-receptor interactions play key roles in blood vessel angiogenesis but remain poorly characterized during lymphatic vessel growth. In this study, we use genetic approaches in both fish and mice to address the roles of cell surface integrin ligand vascular cell adhesion molecule (VCAM) and its 2 receptors, integrins α9 and α4, during lymphatic vascular development. APPROACH AND RESULTS Conditional deletion of the Vcam gene was used to test VCAM function in lymphatic growth in midgestation mice. Morpholino knockdown and cRNA rescue of the 2 zebrafish vcam alleles, as well as integrins α9 and 4, were used to test the role of these ligands and receptors during lymphatic growth in the developing fish. We show that VCAM is essential for lymphatic development in the zebrafish embryo and that integrin α9 (Itgα9) rather than Itgα4 is the required VCAM receptor in the developing fish. VCAM is expressed along lines of lymphatic migration in the mouse intestine, but its loss only retards lymphatic growth. CONCLUSIONS These studies reveal an unexpected role for cell-cell adhesion mediated by Itgα9-VCAM interactions during lymphatic development in the fish but not in the mouse. We propose that the relative importance of cellular adhesive ligands is magnified under conditions of rapid tissue growth when the cell number increases faster than cell matrix, such as in the early zebrafish embryo.
Collapse
Affiliation(s)
- Yiqing Yang
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - David Enis
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Hui Zheng
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Stephanie Chia
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Jisheng Yang
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Mei Chen
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Veerpal Dhillon
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Thalia Papayannapoulou
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Mark L Kahn
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.).
| |
Collapse
|
86
|
Mahadevan A, Welsh IC, Sivakumar A, Gludish DW, Shilvock AR, Noden DM, Huss D, Lansford R, Kurpios NA. The left-right Pitx2 pathway drives organ-specific arterial and lymphatic development in the intestine. Dev Cell 2014; 31:690-706. [PMID: 25482882 PMCID: PMC4326534 DOI: 10.1016/j.devcel.2014.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/29/2014] [Accepted: 10/31/2014] [Indexed: 01/16/2023]
Abstract
The dorsal mesentery (DM) is the major conduit for blood and lymphatic vessels in the gut. The mechanisms underlying their morphogenesis are challenging to study and remain unknown. Here we show that arteriogenesis in the DM begins during gut rotation and proceeds strictly on the left side, dependent on the Pitx2 target gene Cxcl12. Although competent Cxcr4-positive angioblasts are present on the right, they fail to form vessels and progressively emigrate. Surprisingly, gut lymphatics also initiate in the left DM and arise only after-and dependent on-arteriogenesis, implicating arteries as drivers of gut lymphangiogenesis. Our data begin to unravel the origin of two distinct vascular systems and demonstrate how early left-right molecular asymmetries are translated into organ-specific vascular patterns. We propose a dual origin of gut lymphangiogenesis in which prior arterial growth is required to initiate local lymphatics that only subsequently connect to the vascular system.
Collapse
Affiliation(s)
- Aparna Mahadevan
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ian C Welsh
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - David W Gludish
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Abigail R Shilvock
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Drew M Noden
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - David Huss
- Developmental Neuroscience Program and Department of Radiology, Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, 4661 Sunset Boulevard, Los Angeles, CA 90027, USA
| | - Rusty Lansford
- Developmental Neuroscience Program and Department of Radiology, Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, 4661 Sunset Boulevard, Los Angeles, CA 90027, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
87
|
Klein KR, Karpinich NO, Espenschied ST, Willcockson HH, Dunworth WP, Hoopes SL, Kushner EJ, Bautch VL, Caron KM. Decoy receptor CXCR7 modulates adrenomedullin-mediated cardiac and lymphatic vascular development. Dev Cell 2014; 30:528-40. [PMID: 25203207 DOI: 10.1016/j.devcel.2014.07.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/06/2014] [Accepted: 07/14/2014] [Indexed: 01/08/2023]
Abstract
Atypical 7-transmembrane receptors, often called decoy receptors, act promiscuously as molecular sinks to regulate ligand bioavailability and consequently temper the signaling of canonical G protein-coupled receptor (GPCR) pathways. Loss of mammalian CXCR7, the most recently described decoy receptor, results in postnatal lethality due to aberrant cardiac development and myocyte hyperplasia. Here, we provide the molecular underpinning for this proliferative phenotype by demonstrating that the dosage and signaling of adrenomedullin (Adm, gene; AM, protein)-a mitogenic peptide hormone required for normal cardiovascular development-is tightly controlled by CXCR7. To this end, Cxcr7(-/-) mice exhibit gain-of-function cardiac and lymphatic vascular phenotypes that can be reversed upon genetic depletion of adrenomedullin ligand. In addition to identifying a biological ligand accountable for the phenotypes of Cxcr7(-/-) mice, these results reveal a previously underappreciated role for decoy receptors as molecular rheostats in controlling the timing and extent of GPCR-mediated cardiac and vascular development.
Collapse
Affiliation(s)
- Klara R Klein
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Natalie O Karpinich
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott T Espenschied
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Helen H Willcockson
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - William P Dunworth
- Department of Genetics, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Samantha L Hoopes
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Erich J Kushner
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria L Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
88
|
Abstract
Chemokines are a group of small, secreted molecules that signal through G protein-coupled receptors to promote cell survival and proliferation and to provide directional guidance to migrating cells. CXCL12 is one of the most evolutionary conserved chemokines and signals through the chemokine receptor CXCR4 to guide cell migration during embryogenesis, immune cell trafficking and cancer metastasis. Here and in the accompanying poster, we provide an overview of chemokine signaling, focusing on CXCL12, and we highlight some of the different chemokine-dependent strategies used to guide migrating cells.
Collapse
Affiliation(s)
- John Wang
- Developmental Genetics Program, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Holger Knaut
- Developmental Genetics Program, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA Kimmel Center for Stem Cell Biology, New York University Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
89
|
Nakagawara H, Tajima H, Miyashita T, Kitagawa H, Makino I, Sakai S, Hayashi H, Nakanuma S, Oyama K, Takamura H, Fushida S, Fujimura T, Ohta T. Lymph node spread of gallbladder cancer from the perspective of embryologically-based anatomy and significance of the lymphatic basin along the embryonic right hepatic artery. Mol Clin Oncol 2014; 2:963-967. [PMID: 25279182 DOI: 10.3892/mco.2014.342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/03/2014] [Indexed: 12/18/2022] Open
Abstract
Lymph node metastasis from gallbladder cancer is often found in the pericholedochal area; however, these regional lymph nodes are not typically accompanied by arteries. We hypothesized that the artery accompanying pericholedochal lymph nodes was either the regressed embryonic right hepatic artery (eRHA) or an aberrant right hepatic artery (aRHA) remaining without regression. This study aimed to determine the artery supplying pericholedochal lymph nodes. We obtained serial tissue sections of resected specimens from 10 patients who underwent pancreaticoduodenectomy with combined resection of the superior mesenteric artery and vein and investigated the association between the distribution of enlarged lymph nodes and the course of blood vessels in each section. In 2 cases with aRHA, enlarged lymph nodes were distributed in the posterosuperior area, pancreaticoduodenal region and retroportal area along this artery. By contrast, no blood vessels accompanied enlarged lymph nodes in 8 patients exhibiting a normal hepatic artery branching pattern, although these nodes exhibited a distribution pattern similar to that of patients with the aRHA. Thus, the artery supplying pericholedochal lymph nodes appears to be either the regressed eRHA or an aRHA persisting without regression.
Collapse
Affiliation(s)
- Hisatoshi Nakagawara
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Hidehiro Tajima
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Tomoharu Miyashita
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Hirohisa Kitagawa
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Isamu Makino
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Seisho Sakai
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Hironori Hayashi
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Shinichi Nakanuma
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Katsunobu Oyama
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Hiroyuki Takamura
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Sachio Fushida
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Takashi Fujimura
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Tetsuo Ohta
- Division of Cancer Medicine, Department of Gastroenterologic Surgery, Graduate School of Medicine Science, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| |
Collapse
|
90
|
Lee KM, Danuser R, Stein JV, Graham D, Nibbs RJB, Graham GJ. The chemokine receptors ACKR2 and CCR2 reciprocally regulate lymphatic vessel density. EMBO J 2014; 33:2564-80. [PMID: 25271254 PMCID: PMC4283412 DOI: 10.15252/embj.201488887] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Macrophages regulate lymphatic vasculature development; however, the molecular mechanisms regulating their recruitment to developing, and adult, lymphatic vascular sites are not known. Here, we report that resting mice deficient for the inflammatory chemokine-scavenging receptor, ACKR2, display increased lymphatic vessel density in a range of tissues under resting and regenerating conditions. This appears not to alter dendritic cell migration to draining lymph nodes but is associated with enhanced fluid drainage from peripheral tissues and thus with a hypotensive phenotype. Examination of embryonic skin revealed that this lymphatic vessel density phenotype is developmentally established. Further studies indicated that macrophages and the inflammatory CC-chemokine CCL2, which is scavenged by ACKR2, are associated with this phenotype. Accordingly, mice deficient for the CCL2 signalling receptor, CCR2, displayed a reciprocal phenotype of reduced lymphatic vessel density. Further examination revealed that proximity of pro-lymphangiogenic macrophages to developing lymphatic vessel surfaces is increased in ACKR2-deficient mice and reduced in CCR2-deficient mice. Therefore, these receptors regulate vessel density by reciprocally modulating pro-lymphangiogenic macrophage recruitment, and proximity, to developing, resting and regenerating lymphatic vessels.
Collapse
Affiliation(s)
- Kit M Lee
- Institute of Infection, Immunity and Inflammation College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Renzo Danuser
- Theodor Kocher Institute University of Bern, Bern, Switzerland
| | - Jens V Stein
- Theodor Kocher Institute University of Bern, Bern, Switzerland
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Robert J B Nibbs
- Institute of Infection, Immunity and Inflammation College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| | - Gerard J Graham
- Institute of Infection, Immunity and Inflammation College of Medical, Veterinary and Life Sciences University of Glasgow, Glasgow, UK
| |
Collapse
|
91
|
Haematopoietic stem cell induction by somite-derived endothelial cells controlled by meox1. Nature 2014; 512:314-8. [DOI: 10.1038/nature13678] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 07/14/2014] [Indexed: 11/08/2022]
|
92
|
Pichol-Thievend C, Hogan BM, Francois M. Lymphatic vascular specification and its modulation during embryonic development. Microvasc Res 2014; 96:3-9. [PMID: 25107456 DOI: 10.1016/j.mvr.2014.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/29/2014] [Indexed: 11/17/2022]
Abstract
Despite its essential roles in development and disease, the lymphatic vascular system has been less studied than the blood vascular network. In recent years, significant advances have been made in understanding the mechanisms that regulate lymphatic vessel formation, both during development and in pathological conditions. Remarkably, lymphatic endothelial cells are specified as a subpopulation of pre-existing venous endothelial cells. Here, we summarize the current knowledge of the transcription factor pathways responsible for lymphatic specification and we also focus on the factors that promote or restrict this event.
Collapse
Affiliation(s)
- Cathy Pichol-Thievend
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Mathias Francois
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
93
|
Kim JD, Jin SW. A tale of two models: mouse and zebrafish as complementary models for lymphatic studies. Mol Cells 2014; 37:503-10. [PMID: 24854860 PMCID: PMC4132301 DOI: 10.14348/molcells.2014.0108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/02/2014] [Indexed: 11/27/2022] Open
Abstract
Lymphatic vessels provide essential roles in maintaining fluid homeostasis and lipid absorption. Dysfunctions of the lymphatic vessels lead to debilitating pathological conditions, collectively known as lymphedema. In addition, lymphatic vessels are a critical moderator for the onset and progression of diverse human diseases including metastatic cancer and obesity. Despite their clinical importance, there is no currently effective pharmacological therapy to regulate functions of lymphatic vessels. Recent efforts to manipulate the Vascular Endothelial Growth Factor-C (VEGFC) pathway, which is arguably the most important signaling pathway regulating lymphatic endothelial cells, to alleviate lymphedema yielded largely mixed results, necessitating identification of new targetable signaling pathways for therapeutic intervention for lymphedema. Zebrafish, a relatively new model system to investigate lymphatic biology, appears to be an ideal model to identify novel therapeutic targets for lymphatic biology. In this review, we will provide an overview of our current understanding of the lymphatic vessels in vertebrates, and discuss zebrafish as a promising in vivo model to study lymphatic vessels.
Collapse
Affiliation(s)
- Jun-Dae Kim
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Suk-Won Jin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
94
|
Angiogenesis in zebrafish. Semin Cell Dev Biol 2014; 31:106-14. [DOI: 10.1016/j.semcdb.2014.04.037] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/24/2014] [Accepted: 04/30/2014] [Indexed: 12/21/2022]
|
95
|
Mulligan TS, Weinstein BM. Emerging from the PAC: studying zebrafish lymphatic development. Microvasc Res 2014; 96:23-30. [PMID: 24928500 DOI: 10.1016/j.mvr.2014.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/03/2014] [Indexed: 12/30/2022]
Abstract
Recently the zebrafish has emerged as a promising vertebrate model of lymphatic vasculature development. The establishment of numerous transgenic lines that label the lymphatic endothelium in the zebrafish has allowed the fine examination of the developmental timing and the anatomy of their lymphatic vasculature. Although many questions remain, studying lymphatic development in the zebrafish has resulted in the identification and characterization of novel and established mediators of lymphatic development and lymphangiogenesis. Here, we review the main stages involved in the development of the lymphatic vasculature in the zebrafish from its origins in the embryonic veins to the formation of the primary lymphatic vessels and highlight some of the key molecules necessary for these stages.
Collapse
Affiliation(s)
- Timothy S Mulligan
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health (NIH), Building 6B, Room 309, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Brant M Weinstein
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health (NIH), Building 6B, Room 309, 9000 Rockville Pike, Bethesda, MD 20892, USA
| |
Collapse
|
96
|
Astin JW, Haggerty MJL, Okuda KS, Le Guen L, Misa JP, Tromp A, Hogan BM, Crosier KE, Crosier PS. Vegfd can compensate for loss of Vegfc in zebrafish facial lymphatic sprouting. Development 2014; 141:2680-90. [PMID: 24903752 DOI: 10.1242/dev.106591] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lymphangiogenesis is a dynamic process that involves the sprouting of lymphatic endothelial cells (LECs) from veins to form lymphatic vessels. Vegfr3 signalling, through its ligand Vegfc and the extracellular protein Ccbe1, is essential for the sprouting of LECs to form the trunk lymphatic network. In this study we determined whether Vegfr3, Vegfc and Ccbe1 are also required for development of the facial and intestinal lymphatic networks in the zebrafish embryo. Whereas Vegfr3 and Ccbe1 are required for the development of all lymphatic vessels, Vegfc is dispensable for facial lymphatic sprouting but not for the complete development of the facial lymphatic network. We show that zebrafish vegfd is expressed in the head, genetically interacts with ccbe1 and can rescue the lymphatic defects observed following the loss of vegfc. Finally, whereas knockdown of vegfd has no phenotype, double knockdown of both vegfc and vegfd is required to prevent facial lymphatic sprouting, suggesting that Vegfc is not essential for all lymphatic sprouting and that Vegfd can compensate for loss of Vegfc during lymphatic development in the zebrafish head.
Collapse
Affiliation(s)
- Jonathan W Astin
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Michael J L Haggerty
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Kazuhide S Okuda
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Ludovic Le Guen
- Division of Molecular Genetics and Development, Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - June P Misa
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Alisha Tromp
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Benjamin M Hogan
- Division of Molecular Genetics and Development, Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Kathryn E Crosier
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine & Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
97
|
Le Guen L, Karpanen T, Schulte D, Harris NC, Koltowska K, Roukens G, Bower NI, van Impel A, Stacker SA, Achen MG, Schulte-Merker S, Hogan BM. Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signaling during embryonic lymphangiogenesis. Development 2014; 141:1239-49. [PMID: 24523457 DOI: 10.1242/dev.100495] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.
Collapse
Affiliation(s)
- Ludovic Le Guen
- Division of Molecular Genetics and Development, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4073, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Wilkinson RN, van Eeden FJ. The Zebrafish as a Model of Vascular Development and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:93-122. [DOI: 10.1016/b978-0-12-386930-2.00005-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
99
|
van Impel A, Schulte-Merker S. A Fisheye View on Lymphangiogenesis. DEVELOPMENTAL ASPECTS OF THE LYMPHATIC VASCULAR SYSTEM 2014; 214:153-65. [DOI: 10.1007/978-3-7091-1646-3_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
100
|
Kwon HB, Fukuhara S, Asakawa K, Ando K, Kashiwada T, Kawakami K, Hibi M, Kwon YG, Kim KW, Alitalo K, Mochizuki N. The parallel growth of motoneuron axons with the dorsal aorta depends on Vegfc/Vegfr3 signaling in zebrafish. Development 2013; 140:4081-90. [PMID: 24046321 PMCID: PMC3913045 DOI: 10.1242/dev.091702] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Blood vessels and neurons grow often side by side. However, the molecular and cellular mechanisms underlying their parallel development remain unclear. Here, we report that a subpopulation of secondary motoneurons extends axons ventrally outside of the neural tubes and rostrocaudally as a fascicle beneath the dorsal aorta (DA) in zebrafish. We tried to clarify the mechanism by which these motoneuron axons grow beneath the DA and found that Vegfc in the DA and Vegfr3 in the motoneurons were essential for the axon growth. Forced expression of either Vegfc in arteries or Vegfr3 in motoneurons resulted in enhanced axon growth of motoneurons over the DA. Both vegfr3 morphants and vegfc morphants lost the alignment of motoneuron axons with DA. In addition, forced expression of two mutant forms of Vegfr3 in motoneurons, potentially trapping endogenous Vegfc, resulted in failure of growth of motoneuron axons beneath the DA. Finally, a vegfr3 mutant fish lacked the motoneuron axons beneath the DA. Collectively, Vegfc from the preformed DA guides the axon growth of secondary motoneurons.
Collapse
Affiliation(s)
- Hyouk-Bum Kwon
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|