151
|
Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 2019; 572:62-66. [PMID: 31341278 DOI: 10.1038/s41586-019-1419-5] [Citation(s) in RCA: 486] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Recent work has shown that meningeal lymphatic vessels (mLVs), mainly in the dorsal part of the skull, are involved in the clearance of cerebrospinal fluid (CSF), but the precise route of CSF drainage is still unknown. Here we reveal the importance of mLVs in the basal part of the skull for this process by visualizing their distinct anatomical location and characterizing their specialized morphological features, which facilitate the uptake and drainage of CSF. Unlike dorsal mLVs, basal mLVs have lymphatic valves and capillaries located adjacent to the subarachnoid space in mice. We also show that basal mLVs are hotspots for the clearance of CSF macromolecules and that both mLV integrity and CSF drainage are impaired with ageing. Our findings should increase the understanding of how mLVs contribute to the neuropathophysiological processes that are associated with ageing.
Collapse
|
152
|
A novel mouse tail lymphedema model for observing lymphatic pump failure during lymphedema development. Sci Rep 2019; 9:10405. [PMID: 31320677 PMCID: PMC6639358 DOI: 10.1038/s41598-019-46797-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
It has been suggested that many forms of secondary lymphedema in humans are driven by a progressive loss of lymphatic pump function after an initial risk-inducing event. However, the link between pump failure and disease progression has remained elusive due to experimental challenges in the clinical setting and a lack of adequate animal models. Using a novel surgical model of lymphatic injury, we track the adaptation and functional decline of the lymphatic network in response to surgery. This model mimics the histological hallmarks of the typical mouse tail lymphedema model while leaving an intact collecting vessel for analysis of functional changes during disease progression. Lymphatic function in the intact collecting vessel negatively correlated with swelling, while a loss of pumping pressure generation remained even after resolution of swelling. By using this model to study the role of obesity in lymphedema development, we show that obesity exacerbates acquired lymphatic pump failure following lymphatic injury, suggesting one mechanism through which obesity may worsen lymphedema. This lymphatic injury model will allow for future studies investigating the molecular mechanisms leading to lymphedema development.
Collapse
|
153
|
Yun JW, Cvek U, Kilgore PCSR, Tsunoda I, Omura S, Sato F, Zivadinov R, Ramanathan M, Minagar A, Alexander JS. Neurolymphatic biomarkers of brain endothelial inflammatory activation: Implications for multiple sclerosis diagnosis. Life Sci 2019; 229:116-123. [PMID: 31082401 DOI: 10.1016/j.lfs.2019.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/01/2019] [Accepted: 05/09/2019] [Indexed: 02/08/2023]
Abstract
AIMS Multiple sclerosis (MS) is the leading cause of non-traumatic neurological disability in young adults, and its diagnosis is often delayed due to the lack of diagnostic markers. Initiation of disease -modifying therapy in the early stages of MS is especially critical because currently available therapy mostly target relapsing-remitting MS, and is less effective as disease progresses into the more chronic form of secondary-progressive MS. Therefore, exploring specific and sensitive biomarkers will facilitate an expedited and more accurate diagnosis to allow currently available therapies to be more effective. MAIN METHODS Western blotting was conducted to detect the expression of neurolymphatic proteins in human brain endothelial cells in culture. Additionally, using a cohort of 150 patients with relapsing remitting MS, 26 with secondary progressive MS, and 60 healthy control samples, neurolymphatic protein expression was detected in serum samples using dot blot analysis. KEY FINDINGS Human brain microvascular endothelial cells express neurolymphatic markers. Neurolymphatic protein abundance increases with tumor necrosis factor (TNF)-α stimulation but decreases with interferon (IFN)- γ or combined (TNF + IFN) treatment. Circulating neurolymphatic protein levels is significantly lower in MS patients. Further, one of the markers, FOXC2, is associated with the clinical stages of MS, with significantly lower expression in secondary progressive MS compared to relapsing remitting MS. SIGNIFICANCE Our findings describe brain endothelial expression of neurolymphatic proteins, which is altered under inflammatory stress, and provide a possibility of using a collective pool of circulating neurolymphatic proteins as a diagnostic and prognostic biomarker of MS.
Collapse
MESH Headings
- Adult
- Biomarkers/blood
- Brain/metabolism
- Case-Control Studies
- Cells, Cultured
- Endothelial Cells/cytology
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Female
- Humans
- Inflammation/blood
- Inflammation/diagnosis
- Inflammation/immunology
- Male
- Middle Aged
- Multiple Sclerosis/blood
- Multiple Sclerosis/diagnosis
- Multiple Sclerosis/immunology
- Multiple Sclerosis, Chronic Progressive/blood
- Multiple Sclerosis, Chronic Progressive/diagnosis
- Multiple Sclerosis, Chronic Progressive/immunology
- Multiple Sclerosis, Relapsing-Remitting/blood
- Multiple Sclerosis, Relapsing-Remitting/diagnosis
- Multiple Sclerosis, Relapsing-Remitting/immunology
Collapse
Affiliation(s)
- J W Yun
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - U Cvek
- Laboratory for Advanced Biomedical Informatics, Louisiana State University Shreveport, Shreveport, LA, 71115, USA
| | - P C S R Kilgore
- Laboratory for Advanced Biomedical Informatics, Louisiana State University Shreveport, Shreveport, LA, 71115, USA
| | - I Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan
| | - S Omura
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan
| | - F Sato
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka, Japan
| | - R Zivadinov
- Department of Neurology, State University of New York, Buffalo, NY, USA; Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - M Ramanathan
- Department of Neurology, State University of New York, Buffalo, NY, USA; Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | - A Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - J S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA; Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA.
| |
Collapse
|
154
|
Qu X, Violette K, Sewell-Loftin MK, Soslow J, Saint-Jean L, Hinton RB, Merryman WD, Baldwin HS. Loss of flow responsive Tie1 results in Impaired
Aortic valve remodeling. Dev Biol 2019; 455:73-84. [PMID: 31319059 DOI: 10.1016/j.ydbio.2019.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 10/26/2022]
Abstract
The mechanisms regulating endothelial cell response to hemodynamic forces required for heart valve development, especially valve remodeling, remain elusive. Tie1, an endothelial specific receptor tyrosine kinase, is up-regulated by oscillating shear stress and is required for lymphatic valve development. In this study, we demonstrate that valvular endothelial Tie1 is differentially expressed in a dynamic pattern predicted by disturbed flow during valve remodeling. Following valvular endocardial specific deletion of Tie1 in mice, we observed enlarged aortic valve leaflets, decreased valve stiffness and valvular insufficiency. Valve abnormalities were only detected in late gestation and early postnatal mutant animals and worsened with age. The mutant mice developed perturbed extracellular matrix (ECM) deposition and remodeling characterized by increased glycosaminoglycan and decreased collagen content, as well as increased valve interstitial cell expression of Sox9, a transcription factor essential for normal ECM maturation during heart valve development. This study provides the first evidence that Tie1 is involved in modulation of late valve remodeling and suggests that an important Tie1-Sox9 signaling axis exists through which disturbed flows are converted by endocardial cells to paracrine Sox9 signals to modulate normal matrix remodeling of the aortic valve.
Collapse
Affiliation(s)
- Xianghu Qu
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - Kate Violette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - M K Sewell-Loftin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonathan Soslow
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - LeShana Saint-Jean
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert B Hinton
- Division of Cardiology, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - H Scott Baldwin
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
155
|
Ramadan R, Vromans E, Anang DC, Decrock E, Mysara M, Monsieurs P, Baatout S, Leybaert L, Aerts A. Single and fractionated ionizing radiation induce alterations in endothelial connexin expression and channel function. Sci Rep 2019; 9:4643. [PMID: 31217426 PMCID: PMC6584668 DOI: 10.1038/s41598-019-39317-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is an effective treatment for most tumor types. However, emerging evidence indicates an increased risk for atherosclerosis after ionizing radiation exposure, initiated by endothelial cell dysfunction. Interestingly, endothelial cells express connexin (Cx) proteins that are reported to exert proatherogenic as well as atheroprotective effects. Furthermore, Cxs form channels, gap junctions and hemichannels, that are involved in bystander signaling that leads to indirect radiation effects in non-exposed cells. We here aimed to investigate the consequences of endothelial cell irradiation on Cx expression and channel function. Telomerase immortalized human Coronary Artery/Microvascular Endothelial cells were exposed to single and fractionated X-rays. Several biological endpoints were investigated at different time points after exposure: Cx gene and protein expression, gap junctional dye coupling and hemichannel function. We demonstrate that single and fractionated irradiation induce upregulation of proatherogenic Cx43 and downregulation of atheroprotective Cx40 gene and protein levels in a dose-dependent manner. Single and fractionated irradiation furthermore increased gap junctional communication and induced hemichannel opening. Our findings indicate alterations in Cx expression that are typically observed in endothelial cells covering atherosclerotic plaques. The observed radiation-induced increase in Cx channel function may promote bystander signaling thereby exacerbating endothelial cell damage and atherogenesis.
Collapse
Affiliation(s)
- Raghda Ramadan
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
- Department of Basic and Applied Medical Sciences, Physiology group, Ghent University, Ghent, Belgium
| | - Els Vromans
- Centre for Environmental Health Sciences, Hasselt University, Hasselt, Belgium
| | - Dornatien Chuo Anang
- Biomedical Research Institute and transnational university of Limburg, Hasselt University, Hasselt, Belgium
| | - Elke Decrock
- Department of Basic and Applied Medical Sciences, Physiology group, Ghent University, Ghent, Belgium
| | - Mohamed Mysara
- Microbiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Pieter Monsieurs
- Microbiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences, Physiology group, Ghent University, Ghent, Belgium
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium.
| |
Collapse
|
156
|
Inman KE, Caiaffa CD, Melton KR, Sandell LL, Achilleos A, Kume T, Trainor PA. Foxc2 is required for proper cardiac neural crest cell migration, outflow tract septation, and ventricle expansion. Dev Dyn 2019; 247:1286-1296. [PMID: 30376688 DOI: 10.1002/dvdy.24684] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Proper development of the great vessels of the heart and septation of the cardiac outflow tract requires cardiac neural crest cells. These cells give rise to the parasympathetic cardiac ganglia, the smooth muscle layer of the great vessels, some cardiomyocytes, and the conotruncal cushions and aorticopulmonary septum of the outflow tract. Ablation of cardiac neural crest cells results in defective patterning of each of these structures. Previous studies have shown that targeted deletion of the forkhead transcription factor C2 (Foxc2), results in cardiac phenotypes similar to that derived from cardiac neural crest cell ablation. RESULTS We report that Foxc2-/- embryos on the 129s6/SvEv inbred genetic background display persistent truncus arteriosus and hypoplastic ventricles before embryonic lethality. Foxc2 loss-of-function resulted in perturbed cardiac neural crest cell migration and their reduced contribution to the outflow tract as evidenced by lineage tracing analyses together with perturbed expression of the neural crest cell markers Sox10 and Crabp1. Foxc2 loss-of-function also resulted in alterations in PlexinD1, Twist1, PECAM1, and Hand1/2 expression in association with vascular and ventricular defects. CONCLUSIONS Our data indicate Foxc2 is required for proper migration of cardiac neural crest cells, septation of the outflow tract, and development of the ventricles. Developmental Dynamics 247:1286-1296, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kimberly E Inman
- Department of Natural Sciences, Shawnee State University, Portsmouth, Ohio
| | | | - Kristin R Melton
- Section of Neonatology, Pulmonary and Perinatal Biology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Lisa L Sandell
- Department of Oral Immunology & Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky
| | - Annita Achilleos
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri.,Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas
| |
Collapse
|
157
|
Chang CW, Seibel AJ, Song JW. Application of microscale culture technologies for studying lymphatic vessel biology. Microcirculation 2019; 26:e12547. [PMID: 30946511 DOI: 10.1111/micc.12547] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/04/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022]
Abstract
Immense progress in microscale engineering technologies has significantly expanded the capabilities of in vitro cell culture systems for reconstituting physiological microenvironments that are mediated by biomolecular gradients, fluid transport, and mechanical forces. Here, we examine the innovative approaches based on microfabricated vessels for studying lymphatic biology. To help understand the necessary design requirements for microfluidic models, we first summarize lymphatic vessel structure and function. Next, we provide an overview of the molecular and biomechanical mediators of lymphatic vessel function. Then we discuss the past achievements and new opportunities for microfluidic culture models to a broad range of applications pertaining to lymphatic vessel physiology. We emphasize the unique attributes of microfluidic systems that enable the recapitulation of multiple physicochemical cues in vitro for studying lymphatic pathophysiology. Current challenges and future outlooks of microscale technology for studying lymphatics are also discussed. Collectively, we make the assertion that further progress in the development of microscale models will continue to enrich our mechanistic understanding of lymphatic biology and physiology to help realize the promise of the lymphatic vasculature as a therapeutic target for a broad spectrum of diseases.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Alex J Seibel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio.,The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
158
|
Janardhan HP, Trivedi CM. Establishment and maintenance of blood-lymph separation. Cell Mol Life Sci 2019; 76:1865-1876. [PMID: 30758642 PMCID: PMC6482084 DOI: 10.1007/s00018-019-03042-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/15/2019] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
Hippocratic Corpus, a collection of Greek medical literature, described the functional anatomy of the lymphatic system in the fifth century B.C. Subsequent studies in cadavers and surgical patients firmly established that lymphatic vessels drain extravasated interstitial fluid, also known as lymph, into the venous system at the bilateral lymphovenous junctions. Recent advances revealed that lymphovenous valves and platelet-mediated hemostasis at the lymphovenous junctions maintain life-long separation of the blood and lymphatic vascular systems. Here, we review murine models that exhibit failure of blood-lymph separation to highlight the novel mechanisms and molecular targets for the modulation of lymphatic disorders. Specifically, we focus on the transcription factors, cofactors, and signaling pathways that regulate lymphovenous valve development and platelet-mediated lymphovenous hemostasis, which cooperate to maintain blood-lymph separation.
Collapse
Affiliation(s)
- Harish P Janardhan
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, The Albert Sherman Center, AS7-1047, 368 Plantation St, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, The Albert Sherman Center, AS7-1047, 368 Plantation St, Worcester, MA, 01605, USA.
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- The Li-Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
159
|
Choi D, Park E, Jung E, Cha B, Lee S, Yu J, Kim PM, Lee S, Hong YJ, Koh CJ, Cho CW, Wu Y, Li Jeon N, Wong AK, Shin L, Kumar SR, Bermejo-Moreno I, Srinivasan RS, Cho IT, Hong YK. Piezo1 incorporates mechanical force signals into the genetic program that governs lymphatic valve development and maintenance. JCI Insight 2019; 4:125068. [PMID: 30676326 DOI: 10.1172/jci.insight.125068] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/17/2019] [Indexed: 01/05/2023] Open
Abstract
The lymphatic system plays crucial roles in tissue homeostasis, lipid absorption, and immune cell trafficking. Although lymphatic valves ensure unidirectional lymph flows, the flow itself controls lymphatic valve formation. Here, we demonstrate that a mechanically activated ion channel Piezo1 senses oscillating shear stress (OSS) and incorporates the signal into the genetic program controlling lymphatic valve development and maintenance. Time-controlled deletion of Piezo1 using a pan-endothelial Cre driver (Cdh5[PAC]-CreERT2) or lymphatic-specific Cre driver (Prox1-CreERT2) equally inhibited lymphatic valve formation in newborn mice. Furthermore, Piezo1 deletion in adult lymphatics caused substantial lymphatic valve degeneration. Piezo1 knockdown in cultured lymphatic endothelial cells (LECs) largely abrogated the OSS-induced upregulation of the lymphatic valve signature genes. Conversely, ectopic Piezo1 overexpression upregulated the lymphatic valve genes in the absence of OSS. Remarkably, activation of Piezo1 using chemical agonist Yoda1 not only accelerated lymphatic valve formation in animals, but also triggered upregulation of some lymphatic valve genes in cultured LECs without exposure to OSS. In summary, our studies together demonstrate that Piezo1 is the force sensor in the mechanotransduction pathway controlling lymphatic valve development and maintenance, and Piezo1 activation is a potentially novel therapeutic strategy for congenital and surgery-associated lymphedema.
Collapse
Affiliation(s)
- Dongwon Choi
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Eunkyung Park
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Eunson Jung
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Somin Lee
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - James Yu
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Paul M Kim
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Sunju Lee
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Yeo Jin Hong
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Chester J Koh
- Division of Pediatric Urology, Texas Children's Hospital, Baylor Collexge of Medicine, Houston, Texas, USA
| | - Chang-Won Cho
- Department of Surgery, and.,Traditional Food Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, South Korea
| | - Yifan Wu
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Noo Li Jeon
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | | | | | | | - Ivan Bermejo-Moreno
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, California, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | - Young-Kwon Hong
- Department of Surgery, and.,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
160
|
Teuwen LA, Geldhof V, Carmeliet P. How glucose, glutamine and fatty acid metabolism shape blood and lymph vessel development. Dev Biol 2019; 447:90-102. [DOI: 10.1016/j.ydbio.2017.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/26/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022]
|
161
|
Surya VN, Michalaki E, Fuller GG, Dunn AR. Lymphatic endothelial cell calcium pulses are sensitive to spatial gradients in wall shear stress. Mol Biol Cell 2019; 30:923-931. [PMID: 30811261 PMCID: PMC6589782 DOI: 10.1091/mbc.e18-10-0618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cytosolic calcium (Ca2+) is a ubiquitous second messenger that influences numerous aspects of cellular function. In many cell types, cytosolic Ca2+ concentrations are characterized by periodic pulses, whose dynamics can influence downstream signal transduction. Here, we examine the general question of how cells use Ca2+ pulses to encode input stimuli in the context of the response of lymphatic endothelial cells (LECs) to fluid flow. Previous work shows that fluid flow regulates Ca2+ dynamics in LECs and that Ca2+-dependent signaling plays a key role in regulating lymphatic valve formation during embryonic development. However, how fluid flow might influence the Ca2+ pulse dynamics of individual LECs has remained, to our knowledge, little explored. We used live-cell imaging to characterize Ca2+ pulse dynamics in LECs exposed to fluid flow in an in vitro flow device that generates spatial gradients in wall shear stress (WSS), such as are found at sites of valve formation. We found that the frequency of Ca2+ pulses was sensitive to the magnitude of WSS, while the duration of individual Ca2+ pulses increased in the presence of spatial gradients in WSS. These observations provide an example of how cells can separately modulate Ca2+ pulse frequency and duration to encode distinct forms of information, a phenomenon that could extend to other cell types.
Collapse
Affiliation(s)
- Vinay N Surya
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | | | - Gerald G Fuller
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
162
|
Lupu M, Caruntu C, Popa MI, Voiculescu VM, Zurac S, Boda D. Vascular patterns in basal cell carcinoma: Dermoscopic, confocal and histopathological perspectives. Oncol Lett 2019; 17:4112-4125. [PMID: 30944604 PMCID: PMC6444327 DOI: 10.3892/ol.2019.10070] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Basal cell carcinoma (BCC) is the most prevalent skin cancer in the Caucasian population. A variety of different phenotypic presentations of BCC are possible. Although BCCs rarely metastasize, these tumors commonly destroy underlying tissues and should therefore be treated promptly. As vascular formation and angiogenesis are indicators of tumor development and progression, the presence of blood vessels, their morphology and architecture are important markers in skin lesions, providing critical information towards pathogenesis and diagnosis. BCC commonly lacks pigmentation, therefore it is important to emphasize the usefulness of vascular feature detection, recognition, quantification and interpretation. To answer the question of whether vascular patterns observed on dermoscopy, reflectance confocal microscopy (RCM) and histopathology might reflect the biologic behavior of BCCs, we undertook this review article. Several studies have sought, by various means, to identify vascular features associated with the more aggressive BCC phenotypes. Dermoscopic vascular pattern assessment can facilitate diagnostic discrimination between BCC subtypes, more aggressive BCCs displaying less or no pink coloration and a relative absence of central tumor vessels. RCM, a novel, non-invasive imaging technique, allows for the quantification of blood vessel size, density, and flow intensity in BCCs. BCCs are distinguished on RCM chiefly by vessels that branch and intertwine between neoplastic aggregates, a pattern strongly reflecting tumor neo-angiogenesis. The analysis of these vascular morphological and distribution patterns can provide further support in the diagnosis, assessment, or monitoring of BCCs. Histopathology shows significantly higher microvessel densities in the peritumoral stroma of BCCs, when compared to normal skin or benign tumors. This angiogenic response in the stroma is associated with local aggressiveness, therefore the quantification of peritumoralmicrovessels may further assist with tumor evaluation. How dermoscopy and RCM vascular patterns in BCC correlate with histopathological subtype and thus help in discriminating aggressive subtypes definitely deserves further investigation.
Collapse
Affiliation(s)
- Mihai Lupu
- Department of Dermatology, MEDAS Medical Center, 030442 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Dermatology, 'Prof. N. Paulescu' National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Maria Iris Popa
- Department of Plastic and Reconstructive Surgery, 'Bagdasar Arseni' Clinical Emergency Hospital, 041915 Bucharest, Romania
| | - Vlad Mihai Voiculescu
- Department of Dermatology, 'Elias' University Emergency Hospital, 011461 Bucharest, Romania
| | - Sabina Zurac
- Department of Pathology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050653 Bucharest, Romania.,Department of Pathology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Daniel Boda
- Department of Dermatology, 'Prof. N. Paulescu' National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania.,Dermatology Research Laboratory, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
163
|
Narayanan SA, Ford J, Zawieja DC. Impairment of lymphatic endothelial barrier function by X-ray irradiation. Int J Radiat Biol 2019; 95:562-570. [PMID: 30570385 DOI: 10.1080/09553002.2019.1562253] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE Although the microvascular system is a significant target for radiation-induced effects, the lymphatic response to radiation has not been extensively investigated. This is one of the first investigations characterizing the lymphatic endothelial response to ionizing radiation. MATERIALS AND METHODS Rat mesenteric lymphatic endothelial cells (RMLECs) were exposed to X-ray doses of 0, 0.5, 1, 1.5, and 2 Gy. RMLEC cellular response was assessed 24 and 72-h post-irradiation via measures of cellular morphometry and junctional adhesion markers. RMLEC functional response was characterized through permeability experiments. RESULTS Cell morphometry showed radiation sensitivity at all doses. Notably, there was a loss of cell-to-cell adhesion with irradiated cells increasing in size and cellular roundness. This was coupled with decreased β-catenin and VE-cadherin intensity and altered F-actin anisotropy, leading to a loss of intercellular contact. RMLEC monolayers demonstrated increased permeability at all doses 24 h post-irradiation and at 2-Gy 72 h post-irradiation. CONCLUSIONS In summary, lymphatics show radiation sensitivity in the context of these cell culture experiments. Our results may have functional implications of lymphatics in tissue, with endothelial barrier dysfunction due to loss of cell-cell adhesion leading to leaky vessels and lymphedema. These preliminary experiments will build the framework for future investigations towards lymphatic radiation exposure response.
Collapse
Affiliation(s)
- S Anand Narayanan
- a Department of Medical Physiology , Texas A&M University -- College of Medicine , Temple , TX , USA
| | - John Ford
- b Department of Nuclear Engineering , Texas A&M University College of Engineering , College Station , TX , USA
| | - David C Zawieja
- a Department of Medical Physiology , Texas A&M University -- College of Medicine , Temple , TX , USA
| |
Collapse
|
164
|
Cho H, Kim J, Ahn JH, Hong YK, Mäkinen T, Lim DS, Koh GY. YAP and TAZ Negatively Regulate Prox1 During Developmental and Pathologic Lymphangiogenesis. Circ Res 2019; 124:225-242. [DOI: 10.1161/circresaha.118.313707] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Hyunsoo Cho
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Jaeryung Kim
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea (J.K., G.Y.K.)
| | - Ji Hoon Ahn
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Young-Kwon Hong
- Department of Surgery (Y.-K.H.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Biology (Y.-K.H.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Taija Mäkinen
- Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (T.M.)
| | - Dae-Sik Lim
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
| | - Gou Young Koh
- From the Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon (H.C., J.H.A., D.-S.L., G.Y.K.)
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea (J.K., G.Y.K.)
| |
Collapse
|
165
|
Trincot CE, Xu W, Zhang H, Kulikauskas MR, Caranasos TG, Jensen BC, Sabine A, Petrova TV, Caron KM. Adrenomedullin Induces Cardiac Lymphangiogenesis After Myocardial Infarction and Regulates Cardiac Edema Via Connexin 43. Circ Res 2019; 124:101-113. [PMID: 30582443 PMCID: PMC6318063 DOI: 10.1161/circresaha.118.313835] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Cardiac lymphangiogenesis contributes to the reparative process post-myocardial infarction, but the factors and mechanisms regulating it are not well understood. OBJECTIVE To determine if epicardial-secreted factor AM (adrenomedullin; Adm=gene) improves cardiac lymphangiogenesis post-myocardial infarction via lateralization of Cx43 (connexin 43) in cardiac lymphatic vasculature. METHODS AND RESULTS Firstly, we identified sex-dependent differences in cardiac lymphatic numbers in uninjured mice using light-sheet microscopy. Using a mouse model of Adm hi/hi ( Adm overexpression) and permanent left anterior descending ligation to induce myocardial infarction, we investigated cardiac lymphatic structure, growth, and function in injured murine hearts. Overexpression of Adm increased lymphangiogenesis and cardiac function post-myocardial infarction while suppressing cardiac edema and correlated with changes in Cx43 localization. Lymphatic function in response to AM treatment was attenuated in mice with a lymphatic-specific Cx43 deletion. In vitro experiments in cultured human lymphatic endothelial cells identified a novel mechanism to improve gap junction coupling by pharmaceutically targeting Cx43 with verapamil. Finally, we show that connexin protein expression in cardiac lymphatics is conserved between mouse and human. CONCLUSIONS AM is an endogenous, epicardial-derived factor that drives reparative cardiac lymphangiogenesis and function via Cx43, and this represents a new therapeutic pathway for improving myocardial edema after injury.
Collapse
Affiliation(s)
- Claire E. Trincot
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill
| | - Wenjing Xu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Molly R. Kulikauskas
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Thomas G. Caranasos
- Department of Surgery, Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina at Chapel Hill
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill
- McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Amelie Sabine
- Department of Oncology, University of Lausanne and Lausanne University Hospital and Ludwig Institute for Cancer Research Lausanne, Chemin de Boveresses 155, CH-1066, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Lausanne University Hospital and Ludwig Institute for Cancer Research Lausanne, Chemin de Boveresses 155, CH-1066, Switzerland
- Division of Experimental Pathlogy, Lausanne University Hospital
| | - Kathleen M. Caron
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill
- McAllister Heart Institute, University of North Carolina at Chapel Hill
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill , 111 Mason Farm Rd, MBRB 6312B, CB 7545, Chapel Hill, NC 27599
| |
Collapse
|
166
|
Hendrikx S, Coso S, Prat-Luri B, Wetterwald L, Sabine A, Franco CA, Nassiri S, Zangger N, Gerhardt H, Delorenzi M, Petrova TV. Endothelial Calcineurin Signaling Restrains Metastatic Outgrowth by Regulating Bmp2. Cell Rep 2019; 26:1227-1241.e6. [DOI: 10.1016/j.celrep.2019.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 11/22/2018] [Accepted: 01/04/2019] [Indexed: 01/02/2023] Open
|
167
|
Rodriguez-Laguna L, Agra N, Ibañez K, Oliva-Molina G, Gordo G, Khurana N, Hominick D, Beato M, Colmenero I, Herranz G, Torres Canizalez JM, Rodríguez Pena R, Vallespín E, Martín-Arenas R, Del Pozo Á, Villaverde C, Bustamante A, Ayuso C, Lapunzina P, Lopez-Gutierrez JC, Dellinger MT, Martinez-Glez V. Somatic activating mutations in PIK3CA cause generalized lymphatic anomaly. J Exp Med 2018; 216:407-418. [PMID: 30591517 PMCID: PMC6363432 DOI: 10.1084/jem.20181353] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/10/2018] [Accepted: 11/29/2018] [Indexed: 11/11/2022] Open
Abstract
Generalized lymphatic anomaly (GLA) is a vascular disorder characterized by diffuse or multifocal lymphatic malformations (LMs). Here, Rodriguez-Laguna et al. report that somatic activating PIK3CA mutations can cause GLA, and we provide preclinical and clinical evidence to support the use of rapamycin for the treatment of GLA. Generalized lymphatic anomaly (GLA) is a vascular disorder characterized by diffuse or multifocal lymphatic malformations (LMs). The etiology of GLA is poorly understood. We identified four distinct somatic PIK3CA variants (Glu542Lys, Gln546Lys, His1047Arg, and His1047Leu) in tissue samples from five out of nine patients with GLA. These same PIK3CA variants occur in PIK3CA-related overgrowth spectrum and cause hyperactivation of the PI3K–AKT–mTOR pathway. We found that the mTOR inhibitor, rapamycin, prevented lymphatic hyperplasia and dysfunction in mice that expressed an active form of PIK3CA (His1047Arg) in their lymphatics. We also found that rapamycin reduced pain in patients with GLA. In conclusion, we report that somatic activating PIK3CA mutations can cause GLA, and we provide preclinical and clinical evidence to support the use of rapamycin for the treatment of this disabling and deadly disease.
Collapse
Affiliation(s)
- Lara Rodriguez-Laguna
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Noelia Agra
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Kristina Ibañez
- Bioinformatics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Gloria Oliva-Molina
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Gema Gordo
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Noor Khurana
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Devon Hominick
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - María Beato
- Department of Pathology, Hospital Universitario La Paz, Madrid, Spain
| | - Isabel Colmenero
- Department of Pathology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Gonzalo Herranz
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Elena Vallespín
- Structural and Functional Genomics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Rubén Martín-Arenas
- Structural and Functional Genomics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Ángela Del Pozo
- Bioinformatics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Cristina Villaverde
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Universidad Autónoma de Madrid, Madrid, Spain
| | - Ana Bustamante
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Ayuso
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Lapunzina
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Clinical Genetics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Juan C Lopez-Gutierrez
- Vascular Anomalies Center, Plastic Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Michael T Dellinger
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX .,Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Victor Martinez-Glez
- Vascular Malformations Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain .,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain.,Clinical Genetics Section, Institute of Medical and Molecular Genetics, Institute of Medical and Molecular Genetics-Instituto de Investigación PAZ, Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
168
|
Breslin JW, Yang Y, Scallan JP, Sweat RS, Adderley SP, Murfee WL. Lymphatic Vessel Network Structure and Physiology. Compr Physiol 2018; 9:207-299. [PMID: 30549020 PMCID: PMC6459625 DOI: 10.1002/cphy.c180015] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The lymphatic system is comprised of a network of vessels interrelated with lymphoid tissue, which has the holistic function to maintain the local physiologic environment for every cell in all tissues of the body. The lymphatic system maintains extracellular fluid homeostasis favorable for optimal tissue function, removing substances that arise due to metabolism or cell death, and optimizing immunity against bacteria, viruses, parasites, and other antigens. This article provides a comprehensive review of important findings over the past century along with recent advances in the understanding of the anatomy and physiology of lymphatic vessels, including tissue/organ specificity, development, mechanisms of lymph formation and transport, lymphangiogenesis, and the roles of lymphatics in disease. © 2019 American Physiological Society. Compr Physiol 9:207-299, 2019.
Collapse
Affiliation(s)
- Jerome W. Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Richard S. Sweat
- Department of Biomedical Engineering, Tulane University, New Orleans, LA
| | - Shaquria P. Adderley
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - W. Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, FL
| |
Collapse
|
169
|
Abstract
PIEZOs are mechanically activated cation channels. Recently, loss-of-function mutations of human PIEZO1 were found among patients with familial lymphedema, suggesting a requirement of PIEZO1 in the lymphatic system. In this paper, utilizing mouse models lacking PIEZO1 in endothelial cells, we show that this ion channel is required for the formation of lymphatic valves, a key structure for proper circulation of lymph in the body. The requirement of PIEZO1 in valve formation provides mechanistic insight on how PIEZO1 variants cause lymphatic dysfunction in patients. This study also extends the relevance of PIEZOs beyond acute signaling molecules (e.g., touch sensation) and highlights the importance of these ion channels in controlling morphological/structural specification during development. PIEZO1 is a cation channel that is activated by mechanical forces such as fluid shear stress or membrane stretch. PIEZO1 loss-of-function mutations in patients are associated with congenital lymphedema with pleural effusion. However, the mechanistic link between PIEZO1 function and the development or function of the lymphatic system is currently unknown. Here, we analyzed two mouse lines lacking PIEZO1 in endothelial cells (via Tie2Cre or Lyve1Cre) and found that they exhibited pleural effusion and died postnatally. Strikingly, the number of lymphatic valves was dramatically reduced in these mice. Lymphatic valves are essential for ensuring proper circulation of lymph. Mechanical forces have been implicated in the development of lymphatic vasculature and valve formation, but the identity of mechanosensors involved is unknown. Expression of FOXC2 and NFATc1, transcription factors known to be required for lymphatic valve development, appeared normal in Tie2Cre;Piezo1cKO mice. However, the process of protrusion in the valve leaflets, which is associated with collective cell migration, actin polymerization, and remodeling of cell–cell junctions, was impaired in Tie2Cre;Piezo1cKO mice. Consistent with these genetic findings, activation of PIEZO1 by Yoda1 in cultured lymphatic endothelial cells induced active remodeling of actomyosin and VE-cadherin+ cell–cell adhesion sites. Our analysis provides evidence that mechanically activated ion channel PIEZO1 is a key regulator of lymphatic valve formation.
Collapse
|
170
|
Bovay E, Sabine A, Prat-Luri B, Kim S, Son K, Willrodt AH, Olsson C, Halin C, Kiefer F, Betsholtz C, Jeon NL, Luther SA, Petrova TV. Multiple roles of lymphatic vessels in peripheral lymph node development. J Exp Med 2018; 215:2760-2777. [PMID: 30355615 PMCID: PMC6219737 DOI: 10.1084/jem.20180217] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/15/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022] Open
Abstract
This work shows how blood and lymphatic vessels contribute to lymph node organogenesis. Both vessel types transport lymphoid tissue inducer cells, while lymphatics also generate interstitial flow, important for mechanical stromal activation and further lymph node expansion. The mammalian lymphatic system consists of strategically located lymph nodes (LNs) embedded into a lymphatic vascular network. Mechanisms underlying development of this highly organized system are not fully understood. Using high-resolution imaging, we show that lymphoid tissue inducer (LTi) cells initially transmigrate from veins at LN development sites using gaps in venous mural coverage. This process is independent of lymphatic vasculature, but lymphatic vessels are indispensable for the transport of LTi cells that egress from blood capillaries elsewhere and serve as an essential LN expansion reservoir. At later stages, lymphatic collecting vessels ensure efficient LTi cell transport and formation of the LN capsule and subcapsular sinus. Perinodal lymphatics also promote local interstitial flow, which cooperates with lymphotoxin-β signaling to amplify stromal CXCL13 production and thereby promote LTi cell retention. Our data unify previous models of LN development by showing that lymphatics intervene at multiple points to assist LN expansion and identify a new role for mechanical forces in LN development.
Collapse
Affiliation(s)
- Esther Bovay
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Amélie Sabine
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Borja Prat-Luri
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Sudong Kim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Kyungmin Son
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | | | - Cecilia Olsson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Friedemann Kiefer
- Max Planck Institute for Molecular Biomedicine, Münster, Germany.,European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland .,Ludwig Institute for Cancer Research, Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Division of Experimental Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
171
|
Hägerling R, Hoppe E, Dierkes C, Stehling M, Makinen T, Butz S, Vestweber D, Kiefer F. Distinct roles of VE-cadherin for development and maintenance of specific lymph vessel beds. EMBO J 2018; 37:embj.201798271. [PMID: 30297530 DOI: 10.15252/embj.201798271] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells line blood and lymphatic vessels and form intercellular junctions, which preserve vessel structure and integrity. The vascular endothelial cadherin, VE-cadherin, mediates endothelial adhesion and is indispensible for blood vessel development and permeability regulation. However, its requirement for lymphatic vessels has not been addressed. During development, VE-cadherin deletion in lymphatic endothelial cells resulted in abortive lymphangiogenesis, edema, and prenatal death. Unexpectedly, inducible postnatal or adult deletion elicited vessel bed-specific responses. Mature dermal lymph vessels resisted VE-cadherin loss and maintained button junctions, which was associated with an upregulation of junctional molecules. Very different, mesenteric lymphatic collectors deteriorated and formed a strongly hyperplastic layer of lymphatic endothelial cells on the mesothelium. This massive hyperproliferation may have been favored by high mesenteric VEGF-C expression and was associated with VEGFR-3 phosphorylation and upregulation of the transcriptional activator TAZ Finally, intestinal lacteals fragmented into cysts or became highly distended possibly as a consequence of the mesenteric defects. Taken together, we demonstrate here the importance of VE-cadherin for lymphatic vessel development and maintenance, which is however remarkably vessel bed-specific.
Collapse
Affiliation(s)
- René Hägerling
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Esther Hoppe
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Cathrin Dierkes
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Taija Makinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Butz
- Department Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- Department Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,DFG Cluster of Excellence 1003 "CiM - Cells in Motion", Münster, Germany
| | - Friedemann Kiefer
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany .,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,DFG Cluster of Excellence 1003 "CiM - Cells in Motion", Münster, Germany
| |
Collapse
|
172
|
Castorena-Gonzalez JA, Zawieja SD, Li M, Srinivasan RS, Simon AM, de Wit C, de la Torre R, Martinez-Lemus LA, Hennig GW, Davis MJ. Mechanisms of Connexin-Related Lymphedema. Circ Res 2018; 123:964-985. [PMID: 30355030 PMCID: PMC6771293 DOI: 10.1161/circresaha.117.312576] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Mutations in GJC2 and GJA1, encoding Cxs (connexins) 47 and 43, respectively, are linked to lymphedema, but the underlying mechanisms are unknown. Because efficient lymph transport relies on the coordinated contractions of lymphatic muscle cells (LMCs) and their electrical coupling through Cxs, Cx-related lymphedema is proposed to result from dyssynchronous contractions of lymphatic vessels. OBJECTIVE To determine which Cx isoforms in LMCs and lymphatic endothelial cells are required for the entrainment of lymphatic contraction waves and efficient lymph transport. METHODS AND RESULTS We developed novel methods to quantify the spatiotemporal entrainment of lymphatic contraction waves and used optogenetic techniques to analyze calcium signaling within and between the LMC and the lymphatic endothelial cell layers. Genetic deletion of the major lymphatic endothelial cell Cxs (Cx43, Cx47, or Cx37) revealed that none were necessary for the synchronization of the global calcium events that triggered propagating contraction waves. We identified Cx45 in human and mouse LMCs as the critical Cx mediating the conduction of pacemaking signals and entrained contractions. Smooth muscle-specific Cx45 deficiency resulted in 10- to 18-fold reduction in conduction speed, partial-to-severe loss of contractile coordination, and impaired lymph pump function ex vivo and in vivo. Cx45 deficiency resulted in profound inhibition of lymph transport in vivo, but only under an imposed gravitational load. CONCLUSIONS Our results (1) identify Cx45 as the Cx isoform mediating the entrainment of the contraction waves in LMCs; (2) show that major endothelial Cxs are dispensable for the entrainment of contractions; (3) reveal a lack of coupling between lymphatic endothelial cells and LMCs, in contrast to arterioles; (4) point to lymphatic valve defects, rather than contraction dyssynchrony, as the mechanism underlying GJC2- or GJA1-related lymphedema; and (5) show that a gravitational load exacerbates lymphatic contractile defects in the intact mouse hindlimb, which is likely critical for the development of lymphedema in the adult mouse.
Collapse
Affiliation(s)
| | - Scott D. Zawieja
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - Min Li
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City OK
| | | | - Cor de Wit
- Institute of Physiology, University of Luebeck, Luebeck Germany
| | | | - Luis A. Martinez-Lemus
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | | | - Michael J. Davis
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| |
Collapse
|
173
|
Investigating Effects of Fluid Shear Stress on Lymphatic Endothelial Cells. Methods Mol Biol 2018. [PMID: 30242762 DOI: 10.1007/978-1-4939-8712-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Recent studies using in vivo models have characterized lymph flow and demonstrated that lymph flow plays a key role in the later stages of lymphatic vascular development, including vascular remodeling, to create a hierarchical collecting vessel network and lymphatic valves (Sweet et al., J Clin Invest 125, 2995-3007, 2015). However, mechanistic insights into the response of lymphatic endothelial cells to fluid flow are difficult to obtain from in vivo studies because of the small size of lymphatic vessels and the technical challenge of lymphatic endothelial cell isolation. On the other hand, in vitro experiments can be tailored to isolate and test specific mechanotransduction pathways more cleanly than conditions in vivo. To measure in vitro the cellular response to flow, cultured primary lymphatic endothelial cells can be exposed to highly specific fluid forces like those believed to exist in vivo. Such in vitro studies have recently helped identify FOXC2 and GATA2 as important transcriptional regulators of lymphatic function during valve formation that are regulated by lymph flow dynamics. This chapter discusses the methods used to expose primary lymphatic endothelial cells (LECs) to lymph fluid dynamics and the relationship of these in vitro studies to in vivo lymphatic biology.
Collapse
|
174
|
Mackie DI, Al Mutairi F, Davis RB, Kechele DO, Nielsen NR, Snyder JC, Caron MG, Kliman HJ, Berg JS, Simms J, Poyner DR, Caron KM. h CALCRL mutation causes autosomal recessive nonimmune hydrops fetalis with lymphatic dysplasia. J Exp Med 2018; 215:2339-2353. [PMID: 30115739 PMCID: PMC6122977 DOI: 10.1084/jem.20180528] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/15/2018] [Accepted: 07/26/2018] [Indexed: 01/19/2023] Open
Abstract
We report the first case of nonimmune hydrops fetalis (NIHF) associated with a recessive, in-frame deletion of V205 in the G protein-coupled receptor, Calcitonin Receptor-Like Receptor (hCALCRL). Homozygosity results in fetal demise from hydrops fetalis, while heterozygosity in females is associated with spontaneous miscarriage and subfertility. Using molecular dynamic modeling and in vitro biochemical assays, we show that the hCLR(V205del) mutant results in misfolding of the first extracellular loop, reducing association with its requisite receptor chaperone, receptor activity modifying protein (RAMP), translocation to the plasma membrane and signaling. Using three independent genetic mouse models we establish that the adrenomedullin-CLR-RAMP2 axis is both necessary and sufficient for driving lymphatic vascular proliferation. Genetic ablation of either lymphatic endothelial Calcrl or nonendothelial Ramp2 leads to severe NIHF with embryonic demise and placental pathologies, similar to that observed in humans. Our results highlight a novel candidate gene for human congenital NIHF and provide structure-function insights of this signaling axis for human physiology.
Collapse
Affiliation(s)
- Duncan I Mackie
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Fuad Al Mutairi
- Department of Pediatrics, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Centre (KAIMRC), Riyadh, Saudi Arabia
| | - Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Daniel O Kechele
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - Joshua C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, NC
- Department of Surgery, Duke University Medical Center, Durham, NC
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC
| | - Harvey J Kliman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - John Simms
- School of Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, England, UK
| | - David R Poyner
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, England, UK
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
175
|
Tharp KM, Weaver VM. Modeling Tissue Polarity in Context. J Mol Biol 2018; 430:3613-3628. [PMID: 30055167 DOI: 10.1016/j.jmb.2018.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/27/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022]
Abstract
Polarity is critical for development and tissue-specific function. However, the acquisition and maintenance of tissue polarity is context dependent. Thus, cell and tissue polarity depend on cell adhesion which is regulated by the cytoskeleton and influenced by the biochemical composition of the extracellular microenvironment and modified by biomechanical cues within the tissue. These biomechanical cues include fluid flow induced shear stresses, cell-density and confinement-mediated compression, and cellular actomyosin tension intrinsic to the tissue or induced in response to morphogens or extracellular matrix stiffness. Here, we discuss how extracellular matrix stiffness and fluid flow influence cell-cell and cell-extracellular matrix adhesion and alter cytoskeletal organization to modulate cell and tissue polarity. We describe model systems that when combined with state of the art molecular screens and high-resolution imaging can be used to investigate how force modulates cell and tissue polarity.
Collapse
Affiliation(s)
- Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA; Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
176
|
Geng X, Cha B, Mahamud MR, Srinivasan RS. Intraluminal valves: development, function and disease. Dis Model Mech 2018; 10:1273-1287. [PMID: 29125824 PMCID: PMC5719258 DOI: 10.1242/dmm.030825] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The circulatory system consists of the heart, blood vessels and lymphatic vessels, which function in parallel to provide nutrients and remove waste from the body. Vascular function depends on valves, which regulate unidirectional fluid flow against gravitational and pressure gradients. Severe valve disorders can cause mortality and some are associated with severe morbidity. Although cardiac valve defects can be treated by valve replacement surgery, no treatment is currently available for valve disorders of the veins and lymphatics. Thus, a better understanding of valves, their development and the progression of valve disease is warranted. In the past decade, molecules that are important for vascular function in humans have been identified, with mouse studies also providing new insights into valve formation and function. Intriguing similarities have recently emerged between the different types of valves concerning their molecular identity, architecture and development. Shear stress generated by fluid flow has also been shown to regulate endothelial cell identity in valves. Here, we review our current understanding of valve development with an emphasis on its mechanobiology and significance to human health, and highlight unanswered questions and translational opportunities.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Md Riaj Mahamud
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
177
|
Ma W, Oliver G. Lymphatic Endothelial Cell Plasticity in Development and Disease. Physiology (Bethesda) 2018; 32:444-452. [PMID: 29021364 DOI: 10.1152/physiol.00015.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 11/22/2022] Open
Abstract
The lymphatic vasculature is crucial for maintaining tissue-fluid homeostasis, providing immune surveillance and mediating lipid absorption. The lymphatic vasculature is tightly associated with the blood vasculature, although it exhibits distinct morphological and functional features. Endothelial cells (ECs) lineage fate specification is determined during embryonic development; however, accumulating evidence suggests that differentiated ECs exhibit remarkable heterogeneity and plasticity. In this review, we provide an overview of the molecular mechanisms promoting lymphatic cell fate specification in the mammalian embryo. We also summarize available data suggesting that lymphatic EC fate is reprogrammable in normal and pathological settings. We further discuss the possible advantages of cell fate manipulation to treat certain disorders associated with lymphatic dysfunction.
Collapse
Affiliation(s)
- Wanshu Ma
- Center for Vascular & Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Guillermo Oliver
- Center for Vascular & Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
178
|
Wörsdörfer P, Wagner N, Ergün S. The role of connexins during early embryonic development: pluripotent stem cells, gene editing, and artificial embryonic tissues as tools to close the knowledge gap. Histochem Cell Biol 2018; 150:327-339. [PMID: 30039329 DOI: 10.1007/s00418-018-1697-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Since almost 4 decades, connexins have been discussed as important regulators of embryogenesis. Several different members of the gene family can be detected in the preimplantation embryo and during gastrulation. However, genetically engineered mice deficient for every connexin expressed during early development are available and even double-deficient mice were generated. Interestingly, all of these mice complete gastrulation without any abnormalities. This raises the question if the role of connexins has been overrated or if other gene family members compensate and mask their importance. To answer this question, embryos completely devoid of any gap junctional communication need to be investigated. This is challenging because a variety of connexin genes are co-expressed and some null mutations lead to a lethal phenotype. In addition, maternal connexin transcripts were described to persist until the blastocyst stage. In this review, we summarize the current knowledge about the role of connexins during preimplantation development and in embryonic stem cells. We propose that the use of pluripotent stem cells, trophoblast stem cells, as well as artificial embryo-like structures and organoid cultures in combination with multiplex CRISPR/Cas9-based genome editing provides a powerful platform to comprehensively readdress this issue and decipher the role of connexins during lineage decision, differentiation, and morphogenesis in a cell culture model for mouse and human development.
Collapse
Affiliation(s)
- Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany.
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstr.6, 97070, Würzburg, Germany
| |
Collapse
|
179
|
Function of Connexins in the Interaction between Glial and Vascular Cells in the Central Nervous System and Related Neurological Diseases. Neural Plast 2018; 2018:6323901. [PMID: 29983707 PMCID: PMC6015683 DOI: 10.1155/2018/6323901] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 02/05/2023] Open
Abstract
Neuronal signaling together with synapse activity in the central nervous system requires a precisely regulated microenvironment. Recently, the blood-brain barrier is considered as a “neuro-glia-vascular unit,” a structural and functional compound composed of capillary endothelial cells, glial cells, pericytes, and neurons, which plays a pivotal role in maintaining the balance of the microenvironment in and out of the brain. Tight junctions and adherens junctions, which function as barriers of the blood-brain barrier, are two well-known kinds in the endothelial cell junctions. In this review, we focus on the less-concerned contribution of gap junction proteins, connexins in blood-brain barrier integrity under physio-/pathology conditions. In the neuro-glia-vascular unit, connexins are expressed in the capillary endothelial cells and prominent in astrocyte endfeet around and associated with maturation and function of the blood-brain barrier through a unique signaling pathway and an interaction with tight junction proteins. Connexin hemichannels and connexin gap junction channels contribute to the physiological or pathological progress of the blood-brain barrier; in addition, the interaction with other cell-cell-adhesive proteins is also associated with the maintenance of the blood-brain barrier. Lastly, we explore the connexins and connexin channels involved in the blood-brain barrier in neurological diseases and any programme for drug discovery or delivery to target or avoid the blood-brain barrier.
Collapse
|
180
|
Molica F, Figueroa XF, Kwak BR, Isakson BE, Gibbins JM. Connexins and Pannexins in Vascular Function and Disease. Int J Mol Sci 2018; 19:ijms19061663. [PMID: 29874791 PMCID: PMC6032213 DOI: 10.3390/ijms19061663] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 12/24/2022] Open
Abstract
Connexins (Cxs) and pannexins (Panxs) are ubiquitous membrane channel forming proteins that are critically involved in many aspects of vascular physiology and pathology. The permeation of ions and small metabolites through Panx channels, Cx hemichannels and gap junction channels confers a crucial role to these proteins in intercellular communication and in maintaining tissue homeostasis. This review provides an overview of current knowledge with respect to the pathophysiological role of these channels in large arteries, the microcirculation, veins, the lymphatic system and platelet function. The essential nature of these membrane proteins in vascular homeostasis is further emphasized by the pathologies that are linked to mutations and polymorphisms in Cx and Panx genes.
Collapse
Affiliation(s)
- Filippo Molica
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland.
| | - Xavier F Figueroa
- Departamento de Fisiología, Faculdad de Ciencias Biológicas, Pontifica Universidad Católica de Chile, Santiago 8330025, Chile.
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland.
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Jonathan M Gibbins
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading RG6 6AS, UK.
| |
Collapse
|
181
|
Wong BW, Zecchin A, García-Caballero M, Carmeliet P. Emerging Concepts in Organ-Specific Lymphatic Vessels and Metabolic Regulation of Lymphatic Development. Dev Cell 2018; 45:289-301. [PMID: 29738709 DOI: 10.1016/j.devcel.2018.03.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/12/2017] [Accepted: 03/26/2018] [Indexed: 02/08/2023]
Abstract
The lymphatic system has been less well characterized than the blood vascular system; however, work in recent years has uncovered novel regulators and non-venous lineages that contribute to lymphatic formation in various organs. Further, the identification of organ-specific lymphatic beds underscores their potential interaction with organ development and function, and highlights the possibility of targeting these organ-specific lymphatics beds in disease. This review focuses on newly described metabolic and epigenetic regulators of lymphangiogenesis and the interplay between lymphatic development and function in a number of major organ systems.
Collapse
Affiliation(s)
- Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, KU Leuven, Herestraat 49 - B912, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven 3000, Belgium
| | - Annalisa Zecchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, KU Leuven, Herestraat 49 - B912, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven 3000, Belgium
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, KU Leuven, Herestraat 49 - B912, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Leuven Cancer Institute, KU Leuven, Herestraat 49 - B912, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven 3000, Belgium.
| |
Collapse
|
182
|
Boucher J, Simonneau C, Denet G, Clarhaut J, Balandre AC, Mesnil M, Cronier L, Monvoisin A. Pannexin-1 in Human Lymphatic Endothelial Cells Regulates Lymphangiogenesis. Int J Mol Sci 2018; 19:ijms19061558. [PMID: 29882918 PMCID: PMC6032340 DOI: 10.3390/ijms19061558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 12/23/2022] Open
Abstract
The molecular mechanisms governing the formation of lymphatic vasculature are not yet well understood. Pannexins are transmembrane proteins that form channels which allow for diffusion of ions and small molecules (<1 kDa) between the extracellular space and the cytosol. The expression and function of pannexins in blood vessels have been studied in the last few decades. Meanwhile, no studies have been conducted to evaluate the role of pannexins during human lymphatic vessel formation. Here we show, using primary human dermal lymphatic endothelial cells (HDLECs), pharmacological tools (probenecid, Brilliant Blue FCF, mimetic peptides [10Panx]) and siRNA-mediated knockdown that Pannexin-1 is necessary for capillary tube formation on Matrigel and for VEGF-C-induced invasion. These results newly identify Pannexin-1 as a protein highly expressed in HDLECs and its requirement during in vitro lymphangiogenesis.
Collapse
Affiliation(s)
- Jonathan Boucher
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Claire Simonneau
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Golthlay Denet
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Jonathan Clarhaut
- CNRS UMR 7285, Institut de Chimie des Milieux et des Matériaux de Poitiers (IC2MP), University of Poitiers, 86073 Poitiers, France.
- CHU de Poitiers, 86021 Poitiers, France.
| | - Annie-Claire Balandre
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Marc Mesnil
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Laurent Cronier
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| | - Arnaud Monvoisin
- CNRS ERL 7003, Laboratoire "Signalisation & Transports Ioniques Membranaires", University of Poitiers, 86073 Poitiers, France.
| |
Collapse
|
183
|
Osaki T, Serrano JC, Kamm RD. Cooperative Effects of Vascular Angiogenesis and Lymphangiogenesis. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:120-132. [PMID: 30417074 DOI: 10.1007/s40883-018-0054-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
In this study, we modeled lymphangiogenesis and vascular angiogenesis in a microdevice using a tissue engineering approach. Lymphatic vessels (LV) and blood vessels (BV) were fabricated by sacrificial molding with seeding human lymphatic endothelial cells and human umbilical vein endothelial cells into molded microchannels (600 μm diameter). During subsequent perfusion culture, lymphangiogenesis and vascular angiogenesis were induced by addition of phorbol 12-myristate 13-acetate (PMA) and VEGF-C or VEGF-A characterized by podoplanin and Prox-1 expression. The lymphatic capillaries formed button-like junctions treated with dexamethasone. To test the potential for screening anti-angiogenic (vascular and lymphatic) factors, antagonists of VEGF were introduced. We found that an inhibitor of VEGF-R3 did not completely suppress lymphatic angiogenesis with BVs present, although lymphatic angiogenesis was selectively prevented by addition of a VEGF-R3 inhibitor without BVs. To probe the mechanism of action, we focus on matrix metalloproteinase (MMP) secretion by vascular endothelial cells and lymphatic endothelial cells under monoculture or co-culture conditions. We found that vascular angiogenesis facilitated lymphangiogenesis via remodeling of the local microenvironment by the increased secretion of MMP, mainly by endothelial cells. Applications of this model include a drug screening assay for corneal disease and models for tumorigenesis including lymphatic angiogenesis and vascular angiogenesis.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jean C Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,BioSystems and Micromechanics (BioSyM), Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| |
Collapse
|
184
|
Frye M, Taddei A, Dierkes C, Martinez-Corral I, Fielden M, Ortsäter H, Kazenwadel J, Calado DP, Ostergaard P, Salminen M, He L, Harvey NL, Kiefer F, Mäkinen T. Matrix stiffness controls lymphatic vessel formation through regulation of a GATA2-dependent transcriptional program. Nat Commun 2018; 9:1511. [PMID: 29666442 PMCID: PMC5904183 DOI: 10.1038/s41467-018-03959-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 03/22/2018] [Indexed: 12/31/2022] Open
Abstract
Tissue and vessel wall stiffening alters endothelial cell properties and contributes to vascular dysfunction. However, whether extracellular matrix (ECM) stiffness impacts vascular development is not known. Here we show that matrix stiffness controls lymphatic vascular morphogenesis. Atomic force microscopy measurements in mouse embryos reveal that venous lymphatic endothelial cell (LEC) progenitors experience a decrease in substrate stiffness upon migration out of the cardinal vein, which induces a GATA2-dependent transcriptional program required to form the first lymphatic vessels. Transcriptome analysis shows that LECs grown on a soft matrix exhibit increased GATA2 expression and a GATA2-dependent upregulation of genes involved in cell migration and lymphangiogenesis, including VEGFR3. Analyses of mouse models demonstrate a cell-autonomous function of GATA2 in regulating LEC responsiveness to VEGF-C and in controlling LEC migration and sprouting in vivo. Our study thus uncovers a mechanism by which ECM stiffness dictates the migratory behavior of LECs during early lymphatic development.
Collapse
Affiliation(s)
- Maike Frye
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Andrea Taddei
- Immunity and Cancer Laboratory, The Francis Crick Institute, 1 Midland Road, NW11AT, London, UK
| | - Cathrin Dierkes
- Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Ines Martinez-Corral
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Matthew Fielden
- Department of Applied Physics, KTH Royal Institute of Technology, Albanova University Center, 106 91, Stockholm, Sweden
| | - Henrik Ortsäter
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Jan Kazenwadel
- Centre for Cancer Biology, University of South Australia and SA Pathology, SA5000, Adelaide, South Australia, Australia
| | - Dinis P Calado
- Immunity and Cancer Laboratory, The Francis Crick Institute, 1 Midland Road, NW11AT, London, UK
| | - Pia Ostergaard
- Lymphovascular Research Unit, Molecular and Clinical Sciences Institute, St George's University of London, SW170RE, London, UK
| | - Marjo Salminen
- Department of Veterinary Biosciences, University of Helsinki, 00014, Helsinki, Finland
| | - Liqun He
- Department of Neurosurgery, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, SA5000, Adelaide, South Australia, Australia
| | - Friedemann Kiefer
- Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden.
| |
Collapse
|
185
|
Morris CJ, Kameny RJ, Boehme J, Gong W, He Y, Zhu T, Maltepe E, Raff GW, Fineman JR, Datar SA. KLF2-mediated disruption of PPAR-γ signaling in lymphatic endothelial cells exposed to chronically increased pulmonary lymph flow. Am J Physiol Heart Circ Physiol 2018; 315:H173-H181. [PMID: 29631374 DOI: 10.1152/ajpheart.00635.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lymphatic abnormalities associated with congenital heart disease are well described, yet the underlying mechanisms remain poorly understood. Using a clinically relevant ovine model of congenital heart disease with increased pulmonary blood flow, we have previously demonstrated that lymphatic endothelial cells (LECs) exposed in vivo to chronically increased pulmonary lymph flow accumulate ROS and have decreased bioavailable nitric oxide (NO). Peroxisome proliferator-activated receptor-γ (PPAR-γ), which abrogates production of cellular ROS by NADPH oxidase, is inhibited by Krüppel-like factor 2 (KLF2), a flow-induced transcription factor. We hypothesized that chronically increased pulmonary lymph flow induces a KLF2-mediated decrease in PPAR-γ and an accumulation of cellular ROS, contributing to decreased bioavailable NO in LECs. To better understand the mechanisms that transduce the abnormal mechanical forces associated with chronically increased pulmonary lymph flow, LECs were isolated from the efferent vessel of the caudal mediastinal lymph node of control ( n = 5) and shunt ( n = 5) lambs. KLF2 mRNA and protein were significantly increased in shunt compared with control LECs, and PPAR-γ mRNA and protein were significantly decreased. In control LECs exposed to shear forces in vitro, we found similar alterations to KLF2 and PPAR-γ expression. In shunt LECs, NADPH oxidase subunit expression was increased, and bioavailable NO was significantly lower. Transfection of shunt LECs with KLF2 siRNA normalized PPAR-γ, ROS, and bioavailable NO. Conversely, pharmacological inhibition of PPAR-γ in control LECs increased ROS equivalent to levels in shunt LECs at baseline. Taken together, these data suggest that one mechanism by which NO-mediated lymphatic function is disrupted after chronic exposure to increased pulmonary lymph flow is through altered KLF2-dependent PPAR-γ signaling, resulting in increased NADPH oxidase activity, accumulation of ROS, and decreased bioavailable NO. NEW & NOTEWORTHY Lymphatic endothelial cells, when exposed in vivo to chronically elevated pulmonary lymph flow in a model of congenital heart disease with increased pulmonary blood flow, demonstrate Krüppel-like factor 2-dependent disrupted peroxisome proliferator-activated receptor-γ signaling that results in the accumulation of reactive oxygen species and decreased bioavailable nitric oxide.
Collapse
Affiliation(s)
- Catherine J Morris
- Department of Pediatrics, University of California , San Francisco, California
| | - Rebecca J Kameny
- Department of Pediatrics, University of California , San Francisco, California
| | - Jason Boehme
- Department of Pediatrics, University of California , San Francisco, California
| | - Wenhui Gong
- Department of Pediatrics, University of California , San Francisco, California
| | - Youping He
- Department of Pediatrics, University of California , San Francisco, California
| | - Terry Zhu
- Department of Pediatrics, University of California , San Francisco, California
| | - Emin Maltepe
- Department of Pediatrics, University of California , San Francisco, California
| | - Gary W Raff
- Department of Surgery, University of California , Davis, California
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California , San Francisco, California.,Cardiovascular Research Institute, University of California , San Francisco, California
| | - Sanjeev A Datar
- Department of Pediatrics, University of California , San Francisco, California
| |
Collapse
|
186
|
Dong J, Xin J, Shen W, Chen X, Wen T, Zhang C, Wang R. Unipedal Diagnostic Lymphangiography Followed by Sequential CT Examinations in Patients With Idiopathic Chyluria: A Retrospective Study. AJR Am J Roentgenol 2018; 210:792-798. [PMID: 29470154 DOI: 10.2214/ajr.17.18936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The objective of our study was to investigate the clinical value of diagnostic lymphangiography followed by sequential CT examinations in patients with idiopathic chyluria. MATERIALS AND METHODS Thirty-six patients with idiopathic chyluria underwent unipedal diagnostic lymphangiography and then underwent sequential CT examinations. The examinations were reviewed separately by two radiologists. Abnormal distribution of contrast medium, lymphourinary leakages, and retrograde flow were noted, and the range and distribution of lymphatic vessel lesions were recorded. The stage of idiopathic chyluria based on CT findings and the stage based on clinical findings were compared. Therapeutic management and follow-up were recorded. Statistical analyses were performed. RESULTS Compared with CT studies performed after lymphangiography, diagnostic lymphangiography showed a unique capability to depict lymphourinary leakages in three patients. Lymphourinary fistulas and abnormal dilated lymphatic vessels were found in and around kidney in all patients. CT depicted retrograde flow of lymph fluid in 47.2% of patients. The consistency in staging chyluria based on CT findings and clinical findings was fair (κ = 0.455). Twenty-nine patients underwent conservative therapy, and seven underwent surgery. Surgical therapy was superior to conservative management (no recurrence, 85.7% of patients who underwent surgery vs 62.1% of patients who underwent conservative therapy; p = 0.025). CONCLUSION From assessing the drainage of contrast medium on unipedal diagnostic lymphangiography and the redistribution of contrast medium on sequential CT examinations, it is possible to detect the existence of lymphourinary fistulas, the precise location of lymphatic anomalies, the distribution of collateral lymphatic vessels, and hydrodynamic pressure abnormality in the lymph circulation in patients with idiopathic chyluria. CT staging of chyluria provides additional information that can be used to guide therapeutic management.
Collapse
Affiliation(s)
- Jian Dong
- 1 Department of Radiology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi St, Haidian District, Beijing 100038, China
| | - Jianfeng Xin
- 2 Department of Lymphatic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Shen
- 2 Department of Lymphatic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaobai Chen
- 1 Department of Radiology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi St, Haidian District, Beijing 100038, China
| | - Tingguo Wen
- 1 Department of Radiology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi St, Haidian District, Beijing 100038, China
| | - Chunyan Zhang
- 1 Department of Radiology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi St, Haidian District, Beijing 100038, China
| | - Rengui Wang
- 1 Department of Radiology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi St, Haidian District, Beijing 100038, China
| |
Collapse
|
187
|
Martin-Almedina S, Mansour S, Ostergaard P. Human phenotypes caused by PIEZO1 mutations; one gene, two overlapping phenotypes? J Physiol 2018; 596:985-992. [PMID: 29331020 PMCID: PMC5851881 DOI: 10.1113/jp275718] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/03/2018] [Indexed: 12/22/2022] Open
Abstract
PIEZO1 is a large mechanosensitive ion channel protein. Diseases associated with PIEZO1 include autosomal recessive generalised lymphatic dysplasia of Fotiou (GLDF) and autosomal dominant dehydrated hereditary stomatocytosis with or without pseudohyperkalemia and/or perinatal oedema (DHS). The two disorders show overlapping features, fetal hydrops/perinatal oedema have been reported in both. Electrophysiological studies suggest opposite mechanisms of action: the mutations identified in GLDF patients cause a loss-of-function mechanism of disease and mutations in DHS patients cause gain of function. This raises the question: Is the pathogenic disease mechanism behind the fetal oedema the same in the two phenotypes? In this Symposium Review, we will discuss the two conditions and highlight key questions that remain to be answered. For instance, the perinatal oedema often resolves soon after birth and we are still at a loss to understand why. Are there any mechanisms which could compensate for the faulty PIEZO1 in these patients? Are there physiological changes at birth that are less reliant on the function of PIEZO1? Thus, there is a clear need for further studies into the two disorders, in order to fully understand the role of PIEZO1 in health and disease.
Collapse
Affiliation(s)
- Silvia Martin-Almedina
- Molecular and Clinical Sciences Institute, St George's, University of London, London, UK
| | - Sahar Mansour
- South West Thames Regional Genetics Unit, St George's, University of London, London, UK
| | - Pia Ostergaard
- Molecular and Clinical Sciences Institute, St George's, University of London, London, UK
| |
Collapse
|
188
|
Zhang Y, Daubel N, Stritt S, Mäkinen T. Transient loss of venous integrity during developmental vascular remodeling leads to red blood cell extravasation and clearance by lymphatic vessels. Development 2018; 145:dev.156745. [PMID: 29361560 PMCID: PMC5818000 DOI: 10.1242/dev.156745] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Maintenance of blood vessel integrity is crucial for vascular homeostasis and is mainly controlled at the level of endothelial cell (EC) junctions. Regulation of endothelial integrity has largely been investigated in the mature quiescent vasculature. Less is known about how integrity is maintained during vascular growth and remodeling involving extensive junctional reorganization. Here, we show that embryonic mesenteric blood vascular remodeling is associated with a transient loss of venous integrity and concomitant extravasation of red blood cells (RBCs), followed by their clearance by the developing lymphatic vessels. In wild-type mouse embryos, we observed activated platelets extending filopodia at sites of inter-EC gaps. In contrast, embryos lacking the activatory C-type lectin domain family 1, member b (CLEC1B) showed extravascular platelets and an excessive number of RBCs associated with and engulfed by the first lymphatic EC clusters that subsequently form lumenized blood-filled vessels connecting to the lymphatic system. These results uncover novel functions of platelets in maintaining venous integrity and lymphatic vessels in clearing extravascular RBCs during developmental remodeling of the mesenteric vasculature. They further provide insight into how vascular abnormalities characterized by blood-filled lymphatic vessels arise.
Collapse
Affiliation(s)
- Yang Zhang
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Nina Daubel
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Simon Stritt
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| |
Collapse
|
189
|
Wang T, Zheng L, Wang Q, Hu YW. Emerging roles and mechanisms of FOXC2 in cancer. Clin Chim Acta 2018; 479:84-93. [PMID: 29341903 DOI: 10.1016/j.cca.2018.01.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Forkhead box protein C2 (FOXC2), a transcription factor of the forkhead/winged-helix family, is required for embryonic and prenatal development. FOXC2 acts as a crucial modulator during both angiogenesis and lymphangiogenesis via multiple angiogenic and lymphangiogenic pathways, respectively. Although recent studies have shed light on the emerging role of FOXC2 in cancer, very little is known about the precise underlying mechanisms. The purpose of this review is to summarize the current understanding of FOXC2 and provide potential mechanistic explanations of the relationship between FOXC2 and cancer, as well as discuss the prospect for future research in the promising prognostic value of FOXC2 in cancer.
Collapse
Affiliation(s)
- Teng Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
190
|
Sabine A, Davis MJ, Bovay E, Petrova TV. Characterization of Mouse Mesenteric Lymphatic Valve Structure and Function. Methods Mol Biol 2018; 1846:97-129. [PMID: 30242755 DOI: 10.1007/978-1-4939-8712-2_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intraluminal valves of collecting lymphatic vessels ensure unidirectional lymph transport against hydrostatic pressure gradient. Mouse mesentery harbors up to 800 valves and represents a convenient model for lymphatic valve quantification, high resolution imaging of different stages of valve development as well as for analysis of valve function. The protocol describes embryonic and postnatal mesenteric lymphatic vessel preparation for whole-mount immunofluorescent staining and visualization of valve organization, quantification of main morphological parameters such as valve size and leaflet length, and the quantitative assessment of functional properties of adult valves using back-leak and closure tests.
Collapse
Affiliation(s)
- Amélie Sabine
- Department of Oncology, Division of Experimental Pathology, CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Michael J Davis
- Medical Pharmacology & Physiology, University of Missouri, Columbia, MO, USA
| | - Esther Bovay
- Department of Oncology, Division of Experimental Pathology, CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Division of Experimental Pathology, CHUV, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
- Swiss Institute for Cancer Research, EPFL, Lausanne, Switzerland.
| |
Collapse
|
191
|
García-Cardeña G, Slegtenhorst BR. Hemodynamic Control of Endothelial Cell Fates in Development. Annu Rev Cell Dev Biol 2017; 32:633-648. [PMID: 27712101 DOI: 10.1146/annurev-cellbio-100814-125610] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biomechanical forces are emerging as critical regulators of embryogenesis, particularly in the developing cardiovascular system. From the onset of blood flow, the embryonic vasculature is continuously exposed to a variety of hemodynamic forces. These biomechanical stimuli are key determinants of vascular cell specification and remodeling and the establishment of vascular homeostasis. In recent years, major advances have been made in our understanding of mechano-activated signaling networks that control both spatiotemporal and structural aspects of vascular development. It has become apparent that a major site for mechanotransduction is situated at the interface of blood and the vessel wall and that this process is controlled by the vascular endothelium. In this review, we discuss the hemodynamic control of endothelial cell fates, focusing on arterial-venous specification, lymphatic development, and the endothelial-to-hematopoietic transition, and present some recent insights into the mechano-activated pathways driving these cell fate decisions in the developing embryo.
Collapse
Affiliation(s)
- Guillermo García-Cardeña
- Program in Developmental and Regenerative Biology, Harvard Medical School, Boston, Massachusetts 02115; .,Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Bendix R Slegtenhorst
- Program in Developmental and Regenerative Biology, Harvard Medical School, Boston, Massachusetts 02115; .,Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115.,Department of Surgery, Erasmus MC-University Medical Center, 3015 CE, Rotterdam, The Netherlands
| |
Collapse
|
192
|
Fang JS, Coon BG, Gillis N, Chen Z, Qiu J, Chittenden TW, Burt JM, Schwartz MA, Hirschi KK. Shear-induced Notch-Cx37-p27 axis arrests endothelial cell cycle to enable arterial specification. Nat Commun 2017; 8:2149. [PMID: 29247167 PMCID: PMC5732288 DOI: 10.1038/s41467-017-01742-7] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 10/13/2017] [Indexed: 01/26/2023] Open
Abstract
Establishment of a functional vascular network is rate-limiting in embryonic development, tissue repair and engineering. During blood vessel formation, newly generated endothelial cells rapidly expand into primitive plexi that undergo vascular remodeling into circulatory networks, requiring coordinated growth inhibition and arterial-venous specification. Whether the mechanisms controlling endothelial cell cycle arrest and acquisition of specialized phenotypes are interdependent is unknown. Here we demonstrate that fluid shear stress, at arterial flow magnitudes, maximally activates NOTCH signaling, which upregulates GJA4 (commonly, Cx37) and downstream cell cycle inhibitor CDKN1B (p27). Blockade of any of these steps causes hyperproliferation and loss of arterial specification. Re-expression of GJA4 or CDKN1B, or chemical cell cycle inhibition, restores endothelial growth control and arterial gene expression. Thus, we elucidate a mechanochemical pathway in which arterial shear activates a NOTCH-GJA4-CDKN1B axis that promotes endothelial cell cycle arrest to enable arterial gene expression. These insights will guide vascular regeneration and engineering.
Collapse
Affiliation(s)
- Jennifer S Fang
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Brian G Coon
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Noelle Gillis
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Zehua Chen
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE 55 Cambridge Parkway, 8th Floor, Cambridge, MA, 02142, USA
| | - Jingyao Qiu
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Thomas W Chittenden
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE 55 Cambridge Parkway, 8th Floor, Cambridge, MA, 02142, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, A-111, 25 Shattuck Street, Boston, MA, 02115, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street #56-651, Cambridge, MA, 02142, USA
| | - Janis M Burt
- Department of Physiology, College of Medicine, The University of Arizona, 1501 N. Campbell Road, Tucson, AZ, 85724, USA
| | - Martin A Schwartz
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Karen K Hirschi
- Department of Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Cardiovascular Research Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
193
|
Behringer EJ, Scallan JP, Jafarnejad M, Castorena‐Gonzalez JA, Zawieja SD, Moore JE, Davis MJ, Segal SS. Calcium and electrical dynamics in lymphatic endothelium. J Physiol 2017; 595:7347-7368. [PMID: 28994159 PMCID: PMC5730853 DOI: 10.1113/jp274842] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Endothelial cell function in resistance arteries integrates Ca2+ signalling with hyperpolarization to promote relaxation of smooth muscle cells and increase tissue blood flow. Whether complementary signalling occurs in lymphatic endothelium is unknown. Intracellular calcium and membrane potential were evaluated in endothelial cell tubes freshly isolated from mouse collecting lymphatic vessels of the popliteal fossa. Resting membrane potential measured using intracellular microelectrodes averaged ∼-70 mV. Stimulation of lymphatic endothelium by acetylcholine or a TRPV4 channel agonist increased intracellular Ca2+ with robust depolarization. Findings from Trpv4-/- mice and with computational modelling suggest that the initial mobilization of intracellular Ca2+ leads to influx of Ca2+ and Na+ through TRPV4 channels to evoke depolarization. Lymphatic endothelial cells lack the Ca2+ -activated K+ channels present in arterial endothelium to generate endothelium-derived hyperpolarization. Absence of this signalling pathway with effective depolarization may promote rapid conduction of contraction along lymphatic muscle during lymph propulsion. ABSTRACT Subsequent to a rise in intracellular Ca2+ ([Ca2+ ]i ), hyperpolarization of the endothelium coordinates vascular smooth muscle relaxation along resistance arteries during blood flow control. In the lymphatic vasculature, collecting vessels generate rapid contractions coordinated along lymphangions to propel lymph, but the underlying signalling pathways are unknown. We tested the hypothesis that lymphatic endothelial cells (LECs) exhibit Ca2+ and electrical signalling properties that facilitate lymph propulsion. To study electrical and intracellular Ca2+ signalling dynamics in lymphatic endothelium, we excised collecting lymphatic vessels from the popliteal fossa of mice and removed their muscle cells to isolate intact LEC tubes (LECTs). Intracellular recording revealed a resting membrane potential of ∼-70 mV. Acetylcholine (ACh) increased [Ca2+ ]i with a time course similar to that observed in endothelium of resistance arteries (i.e. rapid initial peak with a sustained 'plateau'). In striking contrast to the endothelium-derived hyperpolarization (EDH) characteristic of arteries, LECs depolarized (>15 mV) to either ACh or TRPV4 channel activation. This depolarization was facilitated by the absence of Ca2+ -activated K+ (KCa ) channels as confirmed with PCR, persisted in the absence of extracellular Ca2+ , was abolished by LaCl3 and was attenuated ∼70% in LECTs from Trpv4-/- mice. Computational modelling of ion fluxes in LECs indicated that omitting K+ channels supports our experimental results. These findings reveal novel signalling events in LECs, which are devoid of the KCa activity abundant in arterial endothelium. Absence of EDH with effective depolarization of LECs may promote the rapid conduction of contraction waves along lymphatic muscle during lymph propulsion.
Collapse
Affiliation(s)
- Erik J. Behringer
- Basic SciencesLoma Linda UniversityLoma LindaCA92350USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65212USA
| | - Joshua P. Scallan
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65212USA
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFL33612USA
| | | | | | - Scott D. Zawieja
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65212USA
| | - James E. Moore
- Department of BioengineeringImperial College LondonLondonEngland
| | - Michael J. Davis
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65212USA
- Dalton Cardiovascular Research CenterColumbiaMO65211USA
| | - Steven S. Segal
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65212USA
- Dalton Cardiovascular Research CenterColumbiaMO65211USA
| |
Collapse
|
194
|
Urner S, Kelly-Goss M, Peirce SM, Lammert E. Mechanotransduction in Blood and Lymphatic Vascular Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:155-208. [PMID: 29310798 DOI: 10.1016/bs.apha.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood and lymphatic vasculatures are hierarchical networks of vessels, which constantly transport fluids and, therefore, are exposed to a variety of mechanical forces. Considering the role of mechanotransduction is key for fully understanding how these vascular systems develop, function, and how vascular pathologies evolve. During embryonic development, for example, initiation of blood flow is essential for early vascular remodeling, and increased interstitial fluid pressure as well as initiation of lymph flow is needed for proper development and maturation of the lymphatic vasculature. In this review, we introduce specific mechanical forces that affect both the blood and lymphatic vasculatures, including longitudinal and circumferential stretch, as well as shear stress. In addition, we provide an overview of the role of mechanotransduction during atherosclerosis and secondary lymphedema, which both trigger tissue fibrosis.
Collapse
Affiliation(s)
- Sofia Urner
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Molly Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
195
|
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
|
196
|
Surya VN, Michalaki E, Huang EY, Fuller GG, Dunn AR. Sphingosine 1-phosphate receptor 1 regulates the directional migration of lymphatic endothelial cells in response to fluid shear stress. J R Soc Interface 2017; 13:rsif.2016.0823. [PMID: 27974574 DOI: 10.1098/rsif.2016.0823] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/17/2016] [Indexed: 01/03/2023] Open
Abstract
The endothelial cells that line blood and lymphatic vessels undergo complex, collective migration and rearrangement processes during embryonic development, and are known to be exquisitely responsive to fluid flow. At present, the molecular mechanisms by which endothelial cells sense fluid flow remain incompletely understood. Here, we report that both the G-protein-coupled receptor sphingosine 1-phosphate receptor 1 (S1PR1) and its ligand sphingosine 1-phosphate (S1P) are required for collective upstream migration of human lymphatic microvascular endothelial cells in an in vitro setting. These findings are consistent with a model in which signalling via S1P and S1PR1 are integral components in the response of lymphatic endothelial cells to the stimulus provided by fluid flow.
Collapse
Affiliation(s)
- Vinay N Surya
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eleftheria Michalaki
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eva Y Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Gerald G Fuller
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA .,Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
197
|
Janardhan HP, Milstone ZJ, Shin M, Lawson ND, Keaney JF, Trivedi CM. Hdac3 regulates lymphovenous and lymphatic valve formation. J Clin Invest 2017; 127:4193-4206. [PMID: 29035278 PMCID: PMC5663362 DOI: 10.1172/jci92852] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/31/2017] [Indexed: 12/29/2022] Open
Abstract
Lymphedema, the most common lymphatic anomaly, involves defective lymphatic valve development; yet the epigenetic modifiers underlying lymphatic valve morphogenesis remain elusive. Here, we showed that during mouse development, the histone-modifying enzyme histone deacetylase 3 (Hdac3) regulates the formation of both lymphovenous valves, which maintain the separation of the blood and lymphatic vascular systems, and the lymphatic valves. Endothelium-specific ablation of Hdac3 in mice led to blood-filled lymphatic vessels, edema, defective lymphovenous valve morphogenesis, improper lymphatic drainage, defective lymphatic valve maturation, and complete lethality. Hdac3-deficient lymphovenous valves and lymphatic vessels exhibited reduced expression of the transcription factor Gata2 and its target genes. In response to oscillatory shear stress, the transcription factors Tal1, Gata2, and Ets1/2 physically interacted with and recruited Hdac3 to the evolutionarily conserved E-box–GATA–ETS composite element of a Gata2 intragenic enhancer. In turn, Hdac3 recruited histone acetyltransferase Ep300 to form an enhanceosome complex that promoted Gata2 expression. Together, these results identify Hdac3 as a key epigenetic modifier that maintains blood-lymph separation and integrates both extrinsic forces and intrinsic cues to regulate lymphatic valve development.
Collapse
Affiliation(s)
| | | | - Masahiro Shin
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - John F Keaney
- Division of Cardiovascular Medicine.,Department of Medicine, and
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine.,Department of Medicine, and.,Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
198
|
Abstract
The lymphatic system is essential for the maintenance of tissue fluid homeostasis, gastrointestinal lipid absorption, and immune trafficking. Whereas lymphatic regeneration occurs physiologically in wound healing and tissue repair, pathological lymphangiogenesis has been implicated in a number of chronic diseases such as lymphedema, atherosclerosis, and cancer. Insight into the regulatory mechanisms of lymphangiogenesis and the manner in which uncontrolled inflammation promotes lymphatic dysfunction is urgently needed to guide the development of novel therapeutics: These would be designed to reverse lymphatic dysfunction, either primary or acquired. Recent investigation has demonstrated the mechanistic role of leukotriene B4 (LTB4) in the molecular pathogenesis of lymphedema. LTB4, a product of the innate immune response, is a constituent of the eicosanoid inflammatory mediator family of molecules that promote both physiological and pathological inflammation. Here we provide an overview of lymphatic development, the pathophysiology of lymphedema, and the role of leukotrienes in lymphedema pathogenesis.
Collapse
Affiliation(s)
- Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California 94304, USA.,Stanford University School of Medicine, Stanford, California 94305, USA;
| | - Mark R Nicolls
- VA Palo Alto Health Care System, Palo Alto, California 94304, USA.,Stanford University School of Medicine, Stanford, California 94305, USA;
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California 94304, USA.,Stanford University School of Medicine, Stanford, California 94305, USA;
| | - Stanley G Rockson
- Stanford University School of Medicine, Stanford, California 94305, USA;
| |
Collapse
|
199
|
Kim J, Park DY, Bae H, Park DY, Kim D, Lee CK, Song S, Chung TY, Lim DH, Kubota Y, Hong YK, He Y, Augustin HG, Oliver G, Koh GY. Impaired angiopoietin/Tie2 signaling compromises Schlemm's canal integrity and induces glaucoma. J Clin Invest 2017; 127:3877-3896. [PMID: 28920924 PMCID: PMC5617682 DOI: 10.1172/jci94668] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022] Open
Abstract
Primary open-angle glaucoma (POAG) is often caused by elevated intraocular pressure (IOP), which arises due to increased resistance to aqueous humor outflow (AHO). Aqueous humor flows through Schlemm's canal (SC), a lymphatic-like vessel encircling the cornea, and via intercellular spaces of ciliary muscle cells. However, the mechanisms underlying increased AHO resistance are poorly understood. Here, we demonstrate that signaling between angiopoietin (Angpt) and the Angpt receptor Tie2, which is critical for SC formation, is also indispensable for maintaining SC integrity during adulthood. Deletion of Angpt1/Angpt2 or Tie2 in adult mice severely impaired SC integrity and transcytosis, leading to elevated IOP, retinal neuron damage, and impairment of retinal ganglion cell function, all hallmarks of POAG in humans. We found that SC integrity is maintained by interconnected and coordinated functions of Angpt-Tie2 signaling, AHO, and Prox1 activity. These functions diminish in the SC during aging, leading to impaired integrity and transcytosis. Intriguingly, Tie2 reactivation using a Tie2 agonistic antibody rescued the POAG phenotype in Angpt1/Angpt2-deficient mice and rejuvenated the SC in aged mice. These results indicate that the Angpt-Tie2 system is essential for SC integrity. The impairment of this system underlies POAG-associated pathogenesis, supporting the possibility that Tie2 agonists could be a therapeutic option for glaucoma.
Collapse
Affiliation(s)
- Jaeryung Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Dae-Young Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hosung Bae
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Do Young Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Dongkyu Kim
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Choong-kun Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sukhyun Song
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Preventive Medicine, Catholic University School of Medicine, Seoul, Republic of Korea
| | - Yoshiaki Kubota
- The Laboratory of Vascular Biology, School of Medicine, Keio University, Tokyo, Japan
| | - Young-Kwon Hong
- Department of Surgery, Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yulong He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| |
Collapse
|
200
|
Bernier-Latmani J, Petrova TV. Intestinal lymphatic vasculature: structure, mechanisms and functions. Nat Rev Gastroenterol Hepatol 2017; 14:510-526. [PMID: 28655884 DOI: 10.1038/nrgastro.2017.79] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mammalian intestine is richly supplied with lymphatic vasculature, which has functions ranging from maintenance of interstitial fluid balance to transport of antigens, antigen-presenting cells, dietary lipids and fat-soluble vitamins. In this Review, we provide in-depth information concerning the organization and structure of intestinal lymphatics, the current view of their developmental origins, as well as molecular mechanisms of intestinal lymphatic patterning and maintenance. We will also discuss physiological aspects of intestinal lymph flow regulation and the known and emerging roles of intestinal lymphatic vessels in human diseases, such as IBD, infection and cancer.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale 1015, Lausanne, Switzerland
| |
Collapse
|