1
|
Chen WF, Jou C, Pandey SK, Lo SL. Primary Lymphedema: Anatomically Isolated or a Pervasive Systemic Disorder? PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e6328. [PMID: 39712381 PMCID: PMC11661748 DOI: 10.1097/gox.0000000000006328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 12/24/2024]
Abstract
Background Primary lymphedema, a condition characterized by impaired lymphatic function, has long remained underexplored. Current diagnostic approaches rely on clinical history and genetic testing, yet the genetic underpinnings remain elusive in many cases. Traditional thinking suggests that primary lymphedema is confined to specific anatomical regions, but our experience challenges this notion. We hypothesize that primary lymphedema is systemic lymphatic dysfunction. Methods All patients with clinical diagnosis of primary lymphedema from January 2020 to April 2022 were included in our study. Demographic data, medical and surgical history, and indocyanine green (ICG) lymphographic findings were collected. Results A total of 152 patients met our inclusion criteria. We observed a predominance of female patients (75%) and a mean age of 43.9 years. The onset of swelling varied, with most patients (82.3%) experiencing it in their lower extremities. Notably, ICG lymphography revealed abnormal lymphatic findings in all symptomatic limbs, affecting multiple extremities in 97.4% of patients. Importantly, even among patients initially presenting with limited symptoms, asymptomatic extremities exhibited lymphatic defects. In addition, the extent of lymphatic disease, assessed through ICG lymphography, surpassed clinical symptoms in 80% of cases, underscoring the systemic nature of primary lymphedema. Conclusions Our study suggests that primary lymphedema is a systemic lymphatic insufficiency, affecting the entire lymphatic system. This underscores the importance of comprehensive assessments, even with limited symptoms, to facilitate earlier diagnosis and more effective treatment approaches.
Collapse
Affiliation(s)
- Wei F. Chen
- From the Department of Plastic Surgery, Center for Lymphedema Research and Reconstruction, Cleveland Clinic, Cleveland, OH
| | - Christopher Jou
- From the Department of Plastic Surgery, Center for Lymphedema Research and Reconstruction, Cleveland Clinic, Cleveland, OH
| | - Sonia K. Pandey
- From the Department of Plastic Surgery, Center for Lymphedema Research and Reconstruction, Cleveland Clinic, Cleveland, OH
| | - Shih-Lun Lo
- From the Department of Plastic Surgery, Center for Lymphedema Research and Reconstruction, Cleveland Clinic, Cleveland, OH
- Department of Plastic Surgery, Cathay General Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Cowan DB, Wu H, Chen H. Epsin Endocytic Adaptor Proteins in Angiogenic and Lymphangiogenic Signaling. Cold Spring Harb Perspect Med 2024; 14:a041165. [PMID: 37217282 PMCID: PMC10759987 DOI: 10.1101/cshperspect.a041165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Circulating vascular endothelial growth factor (VEGF) ligands and receptors are central regulators of vasculogenesis, angiogenesis, and lymphangiogenesis. In response to VEGF ligand binding, VEGF receptor tyrosine kinases initiate the chain of events that transduce extracellular signals into endothelial cell responses such as survival, proliferation, and migration. These events are controlled by intricate cellular processes that include the regulation of gene expression at multiple levels, interactions of numerous proteins, and intracellular trafficking of receptor-ligand complexes. Endocytic uptake and transport of macromolecular complexes through the endosome-lysosome system helps fine-tune endothelial cell responses to VEGF signals. Clathrin-dependent endocytosis remains the best understood means of macromolecular entry into cells, although the importance of non-clathrin-dependent pathways is increasingly recognized. Many of these endocytic events rely on adaptor proteins that coordinate internalization of activated cell-surface receptors. In the endothelium of both blood and lymphatic vessels, epsins 1 and 2 are functionally redundant adaptors involved in receptor endocytosis and intracellular sorting. These proteins are capable of binding both lipids and proteins and are important for promoting curvature of the plasma membrane as well as binding ubiquitinated cargo. Here, we discuss the role of epsin proteins and other endocytic adaptors in governing VEGF signaling in angiogenesis and lymphangiogenesis and discuss their therapeutic potential as molecular targets.
Collapse
Affiliation(s)
- Douglas B Cowan
- Vascular Biology Program, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
3
|
Debelenko L, Mansukhani MM, Remotti F. Papillary Intralymphatic Angioendothelioma in a Child With PIK3CA-Related Overgrowth Spectrum: Implication of PI3K Pathway in the Vascular Tumorigenesis. Pediatr Dev Pathol 2023; 26:166-171. [PMID: 36775953 DOI: 10.1177/10935266231152370] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Papillary intralymphatic angioendothelioma (PILA) is an extremely rare vascular tumor and its pathogenesis is unknown. Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA)-related overgrowth spectrum (PROS) is a heterogeneous group of disorders caused by mosaicism for activating mutations of PIK3CA and characterized by asymmetric overgrowth, skeletal anomalies, skin lesions, and vascular malformations. An association between PILA and PROS has not been known. We report a case of PILA involving the spleen of a young girl with the clinical and molecular diagnosis of PROS. Sequencing of the patient's germ-line DNA detected a pathogenic PIK3CA variant c.1357G>A in 10.6% of alleles. Splenectomy revealed a 4-cm tumor composed of ectatic lymphatics with intraluminal papillary projections, consistent with PILA. The tumor cells showed immunohistochemical expression of CD31, CD34, ERG, FLI-1, PROX1, and caldesmon, while D2-40 was negative. The latter may suggest that the tumor derived from an endothelial precursor arrested in the final steps of lymphothelial differentiation, in keeping with the known role of the PIK3CA-governed molecular pathway in the progression of vascular progenitors to mature endothelial cells. The data implicates PIK3CA in the pathogenesis of PILA and broadens the spectrum of phenotypic expressions of PROS.
Collapse
Affiliation(s)
- Larisa Debelenko
- Department of Pathology and Cell Biology, Columbia University-Irving Medical Center, New York, NY, USA
| | - Mahesh M Mansukhani
- Department of Pathology and Cell Biology, Columbia University-Irving Medical Center, New York, NY, USA
| | - Fabrizio Remotti
- Department of Pathology and Cell Biology, Columbia University-Irving Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Tabrizi ZB, Ahmed NS, Horder JL, Storr SJ, Benest AV. Transcription Factor Control of Lymphatic Quiescence and Maturation of Lymphatic Neovessels in Development and Physiology. Front Physiol 2021; 12:672987. [PMID: 34795596 PMCID: PMC8593113 DOI: 10.3389/fphys.2021.672987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/30/2021] [Indexed: 11/25/2022] Open
Abstract
The lymphatic system is a vascular system comprising modified lymphatic endothelial cells, lymph nodes and other lymphoid organs. The system has diverse, but critical functions in both physiology and pathology, and forms an interface between the blood vascular and immune system. It is increasingly evident that remodelling of the lymphatic system occurs alongside remodelling of the blood microvascular system, which is now considered a hallmark of most pathological conditions as well as being critical for normal development. Much attention has focussed on how the blood endothelium undergoes phenotypic switching in development and disease, resulting in over two decades of research to probe the mechanisms underlying the resulting heterogeneity. The lymphatic system has received less attention, and consequently there are fewer descriptions of functional and molecular heterogeneity, but differential transcription factor activity is likely an important control mechanism. Here we introduce and discuss significant transcription factors of relevance to coordinating cellular responses during lymphatic remodelling as the lymphatic endothelium dynamically changes from quiescence to actively remodelling.
Collapse
Affiliation(s)
- Zarah B Tabrizi
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Nada S Ahmed
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Joseph L Horder
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, Centre for Cancer Sciences School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Andrew V Benest
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
5
|
Song H, Zhu J, Li P, Han F, Fang L, Yu P. Metabolic flexibility maintains proliferation and migration of FGFR signaling-deficient lymphatic endothelial cells. J Biol Chem 2021; 297:101149. [PMID: 34473994 PMCID: PMC8498002 DOI: 10.1016/j.jbc.2021.101149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
Metabolic flexibility is the capacity of cells to alter fuel metabolism in response to changes in metabolic demand or nutrient availability. It is critical for maintaining cellular bioenergetics and is involved in the pathogenesis of cardiovascular disease and metabolic disorders. However, the regulation and function of metabolic flexibility in lymphatic endothelial cells (LECs) remain unclear. We have previously shown that glycolysis is the predominant metabolic pathway to generate ATP in LECs and that fibroblast growth factor receptor (FGFR) signaling controls lymphatic vessel formation by promoting glycolysis. Here, we found that chemical inhibition of FGFR activity or knockdown of FGFR1 induces substantial upregulation of fatty acid β-oxidation (FAO) while reducing glycolysis and cellular ATP generation in LECs. Interestingly, such compensatory elevation was not observed in glucose oxidation and glutamine oxidation. Mechanistic studies show that FGFR blockade promotes the expression of carnitine palmitoyltransferase 1A (CPT1A), a rate-limiting enzyme of FAO; this is achieved by dampened extracellular signal–regulated protein kinase activation, which in turn upregulates the expression of the peroxisome proliferator–activated receptor alpha. Metabolic analysis further demonstrates that CPT1A depletion decreases total cellular ATP levels in FGFR1-deficient rather than wildtype LECs. This result suggests that FAO, which makes a negligible contribution to cellular energy under normal conditions, can partially compensate for energy deficiency caused by FGFR inhibition. Consequently, CPT1A silencing potentiates the effect of FGFR1 knockdown on impeding LEC proliferation and migration. Collectively, our study identified a key role of metabolic flexibility in modulating the effect of FGFR signaling on LEC growth.
Collapse
Affiliation(s)
- Hongyuan Song
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jie Zhu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Ping Li
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Fei Han
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Longhou Fang
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, Texas, USA; Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, Houston, Texas, USA; Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, New York City, New York, USA
| | - Pengchun Yu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
6
|
Ho YC, Srinivasan RS. Lymphatic Vasculature in Energy Homeostasis and Obesity. Front Physiol 2020; 11:3. [PMID: 32038308 PMCID: PMC6987243 DOI: 10.3389/fphys.2020.00003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is a leading cause of cardiovascular diseases and cancer. Body mass is regulated by the balance between energy uptake and energy expenditure. The etiology of obesity is determined by multiple factors including genetics, nutrient absorption, and inflammation. Lymphatic vasculature is starting to be appreciated as a critical modulator of metabolism and obesity. The primary function of lymphatic vasculature is to maintain interstitial fluid homeostasis. Lymphatic vessels absorb fluids that extravasate from blood vessels and return them to blood circulation. In addition, lymphatic vessels absorb digested lipids from the intestine and regulate inflammation. Hence, lymphatic vessels could be an exciting target for treating obesity. In this article, we will review our current understanding regarding the relationship between lymphatic vasculature and obesity, and highlight some open questions.
Collapse
Affiliation(s)
- Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
7
|
Abstract
Lymphoedema is an oedematous condition with a specific and complex tissue biology. In the clinical context of cancer, the pathogenesis of lymphoedema ensues most typically from the modalities employed to stage and treat the cancer (in particular, surgery and radiotherapy). Despite advances in cancer treatment, lifelong lymphoedema (limb swelling and the accompanying chronic inflammatory processes) affects approximately one in seven individuals treated for cancer, although estimates of lymphoedema prevalence following cancer treatment vary widely depending upon the diagnostic criteria used and the duration of follow-up. The natural history of cancer-associated lymphoedema is defined by increasing limb girth, fibrosis, inflammation, abnormal fat deposition and eventual marked cutaneous pathology, which also increases the risk of recurrent skin infections. Lymphoedema can substantially affect the daily quality of life of patients, as, in addition to aesthetic concerns, it can cause discomfort and affect the ability to carry out daily tasks. Clinical diagnosis is dependent on comparison of the affected region with the equivalent region on the unaffected side and, if available, with pre-surgical measurements. Surveillance is indicated in this high-risk population to facilitate disease detection at the early stages, when therapeutic interventions are most effective. Treatment modalities include conservative physical strategies that feature complex decongestive therapy (including compression garments) and intermittent pneumatic compression, as well as an emerging spectrum of surgical interventions, including liposuction for late-stage disease. The future application of pharmacological and microsurgical therapeutics for cancer-associated lymphoedema holds great promise.
Collapse
|
8
|
Izen RM, Yamazaki T, Nishinaka-Arai Y, Hong YK, Mukouyama YS. Postnatal development of lymphatic vasculature in the brain meninges. Dev Dyn 2018; 247:741-753. [PMID: 29493038 DOI: 10.1002/dvdy.24624] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 01/25/2018] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Traditionally, the central nervous system (CNS) has been viewed as an immune-privileged environment with no lymphatic vessels. This view was partially overturned by the discovery of lymphatic vessels in the dural membrane that surrounds the brain, in contact with the interior surface of the skull. We here examine the distribution and developmental timing of these lymphatic vessels. RESULTS Using the Prox1-GFP BAC transgenic reporter and immunostaining with antibodies to lymphatic markers LYVE-1, Prox1, and Podoplanin, we have carried out whole-mount imaging of dural lymphatic vasculature at postnatal stages. We have found that between birth and postnatal day (P) 13, lymphatic vessels extend alongside dural blood vessels from the side of the skull toward the midline. Between P13 and P20, lymphatic vessels along the transverse sinuses reach the superior sagittal sinus (SSS) and extend along the SSS toward the olfactory bulb. CONCLUSIONS Compared with the embryonic developmental timing of lymphatic vessels in other tissues, e.g. skin, dural lymphatic vessel development is dramatically delayed. This study provides useful anatomical data for continuing investigations of the fundamental mechanisms that underlie dural lymphatic vessel development. Developmental Dynamics 247:741-753, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rebecca M Izen
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Tomoko Yamazaki
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Yoko Nishinaka-Arai
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.,Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Young-Kwon Hong
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
9
|
Muley A, Odaka Y, Lewkowich IP, Vemaraju S, Yamaguchi TP, Shawber C, Dickie BH, Lang RA. Myeloid Wnt ligands are required for normal development of dermal lymphatic vasculature. PLoS One 2017; 12:e0181549. [PMID: 28846685 PMCID: PMC5573294 DOI: 10.1371/journal.pone.0181549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/03/2017] [Indexed: 12/20/2022] Open
Abstract
Resident tissue myeloid cells play a role in many aspects of physiology including development of the vascular systems. In the blood vasculature, myeloid cells use VEGFC to promote angiogenesis and can use Wnt ligands to control vascular branching and to promote vascular regression. Here we show that myeloid cells also regulate development of the dermal lymphatic vasculature using Wnt ligands. Using myeloid-specific deletion of the WNT transporter Wntless we show that myeloid Wnt ligands are active at two distinct stages of development of the dermal lymphatics. As lymphatic progenitors are emigrating from the cardinal vein and intersomitic vessels, myeloid Wnt ligands regulate both their numbers and migration distance. Later in lymphatic development, myeloid Wnt ligands regulate proliferation of lymphatic endothelial cells (LEC) and thus control lymphatic vessel caliber. Myeloid-specific deletion of WNT co-receptor Lrp5 or Wnt5a gain-of-function also produce elevated caliber in dermal lymphatic capillaries. These data thus suggest that myeloid cells produce Wnt ligands to regulate lymphatic development and use Wnt pathway co-receptors to regulate the balance of Wnt ligand activity during the macrophage-LEC interaction.
Collapse
Affiliation(s)
- Ajit Muley
- Department of OB-GYN, Columbia University Medical Center, Columbia University, New York City, New York, United States of America
| | - Yoshi Odaka
- Visual Systems Group, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Shruti Vemaraju
- Visual Systems Group, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Terry P. Yamaguchi
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, United States of America
| | - Carrie Shawber
- Department of OB-GYN, Columbia University Medical Center, Columbia University, New York City, New York, United States of America
| | - Belinda H. Dickie
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, United States of America
- * E-mail: (RAL); (BHD)
| | - Richard A. Lang
- Visual Systems Group, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Center for Chronobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail: (RAL); (BHD)
| |
Collapse
|
10
|
Deng J, Dai T, Sun Y, Zhang Q, Jiang Z, Li S, Cao W. Overexpression of Prox1 Induces the Differentiation of Human Adipose-Derived Stem Cells into Lymphatic Endothelial-Like Cells In Vitro. Cell Reprogram 2017; 19:54-63. [PMID: 28055225 DOI: 10.1089/cell.2016.0038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Constant levels of homeobox transcription factor Prox1 expression are required throughout the life of lymphatic endothelial cells (LECs) to maintain their differentiated identity. Recent studies have demonstrated that using human LECs for cell transplantation therapy may improve secondary lymphedema in a nude rat model. However, the application is currently limited by the low yield of LECs. In this study, Prox1 was overexpressed in human adipose tissue-derived stem cells (hADSCs) by using the transfection of lentiviral vectors to induce the differentiation of hADSCs to LECs. After 14 days of Prox1 overexpression, flow cytometry analysis found that the expression of LEC-specific markers such as Podoplanin and VEGFR3, along with the endothelial cell (EC) marker CD31, on Prox1-overexpressed hADSCs was significantly increased; however, the expression of mesenchymal stem cell markers, such as CD29, CD44, and CD90, was substantially reduced. In addition, the mRNA levels of the LEC-specific markers, such as Prox1, Podoplanin, LYVE1, and VEGFR3, in Prox1-overexpressed hADSCs were significantly increased at day 7 and maintained a continuously increased expression level for 28 observation days, according to real-time reverse transcriptase-polymerase chain reaction results. Western blotting and immunofluorescence staining results further confirmed that overexpression of Prox1 in hADSCs significantly increased the protein levels of Podoplanin, LYVE1, and VEGFR3, as well as those of the EC markers such as VWF and CD144, at day 14. Moreover, these differentiated cells were found to form tube-like structures in matrigel, measured by the tube formation assay. These findings suggested that overexpression of Prox1 in hADSCs successfully induced the differentiation of hADSCs into stable lymphatic endothelial-like cells. This study achieved a long-lasting expression of Prox1 in lymphatic endothelial-like cells, and it provided a potentially useful approach for developing novel therapies for limb lymphedema and lymphatic system-related diseases.
Collapse
Affiliation(s)
- Jingcheng Deng
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| | - Tingting Dai
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| | - Yiyu Sun
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| | - Qi Zhang
- 2 Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine , Shanghai, P. R. China
| | - Zhaohua Jiang
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| | - Shengli Li
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| | - Weigang Cao
- 1 Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, P. R. China
| |
Collapse
|
11
|
Emerging roles of podoplanin in vascular development and homeostasis. Front Med 2016; 9:421-30. [PMID: 26498027 DOI: 10.1007/s11684-015-0424-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/24/2015] [Indexed: 02/03/2023]
Abstract
Podoplanin (PDPN) is a mucin-type O-glycoprotein expressed in diverse cell types, such as lymphatic endothelial cells (LECs) in the vascular system and fibroblastic reticular cells (FRCs) in lymph nodes. PDPN on LECs or FRCs activates CLEC-2 in platelets, triggering platelet activation and/or aggregation through downstream signaling events, including activation of Syk kinase. This mechanism is required to initiate and maintain separation of blood and lymphatic vessels and to stabilize high endothelial venule integrity within lymphnodes. In the vascular system, normal expression of PDPN at the LEC surface requires transcriptional activation of Pdpn by Prox1 and modification of PDPN with core 1-derived O-glycans. This review provides a comprehensive overview of the roles of PDPN in vascular development and lymphoid organ maintenance and discusses the mechanisms that regulate PDPN expression related to its function.
Collapse
|
12
|
Abstract
The two vascular systems of our body are the blood and the lymphatic vasculature. Our understanding of the genes and molecular mechanisms controlling the development of the lymphatic vasculature network has significantly improved. The availability of novel animal models and better imaging tools led to the identification of lymphatics in tissues and organs previously thought to be devoid of them. Similarly, the classical textbook list of established functional roles of the lymphatic system has been expanded by the addition of novel findings. In this review we provide a historical perspective of some of the important landmarks that opened the doors to researchers working in this field. We also summarize some of the current views about embryonic lymphangiogenesis, particularly about the source(s), commitment, and differentiation of lymphatic endothelial cells.
Collapse
Affiliation(s)
- Noelia Escobedo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Guillermo Oliver
- Center for Vascular & Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611;
| |
Collapse
|
13
|
Wu SP, Yu CT, Tsai SY, Tsai MJ. Choose your destiny: Make a cell fate decision with COUP-TFII. J Steroid Biochem Mol Biol 2016; 157:7-12. [PMID: 26658017 PMCID: PMC4724268 DOI: 10.1016/j.jsbmb.2015.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 06/04/2015] [Accepted: 11/15/2015] [Indexed: 02/06/2023]
Abstract
Cell fate specification is a critical process to generate cells with a wide range of characteristics from stem and progenitor cells. Emerging evidence demonstrates that the orphan nuclear receptor COUP-TFII serves as a key regulator in determining the cell identity during embryonic development. The present review summarizes our current knowledge on molecular mechanisms by which COUP-TFII employs to define the cell fates, with special emphasis on cardiovascular and renal systems. These novel insights pave the road for future studies of regenerative medicine.
Collapse
Affiliation(s)
- San-Pin Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Cheng-Tai Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Nassiri N, Rootman J, Rootman DB, Goldberg RA. Orbital lymphaticovenous malformations: Current and future treatments. Surv Ophthalmol 2015; 60:383-405. [DOI: 10.1016/j.survophthal.2015.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 03/02/2015] [Accepted: 03/06/2015] [Indexed: 12/23/2022]
|
15
|
Defining the lymphatic system of the anterior abdominal wall: an anatomical study. Plast Reconstr Surg 2015; 135:1027-1032. [PMID: 25811569 DOI: 10.1097/prs.0000000000001136] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Studies describing recent abdominoplasty modifications have reported a decreased incidence of seroma, attributed to preservation of abdominal lymphatics. However, there are limited anatomical data to support this hypothesis. The authors sought to characterize the lymphatic architecture of the abdominal wall and provide a conceptual basis for further refinement of abdominoplasty techniques. METHODS Fifteen tissue samples from five patients undergoing abdominoplasty were sectioned and analyzed. Slides were stained with hematoxylin and eosin, CD31, and D2-40 and assessed by a pathologist and a plastic surgeon for the presence and number of lymphatics. Results were reported as mean percentage of lymphatic-specific antibody per analyzed area. RESULTS Lymphatic vessels were observed in the dermis, superficial fascia, and loose areolar tissue but not in deep or superficial fat. The highest concentration was in the dermis (mean, 82.6 percent; range, 69 to 89.2 percent). The Scarpa fascia contained 9.4 percent of lymphatics (range, 7.0 to 11.4 percent), whereas the loose areolar tissue at the specimen base contained an average of 7.9 percent (range, 2.6 to 19.5 percent). These lymphatics were consistently located in the deepest third, with the Scarpa fascia lying an average of 34 percent of the total tissue thickness above muscle fascia. Lymphatic prevalence did not increase in specimens near the superficial epigastric vessels. CONCLUSIONS Abdominoplasty flap lymphatics are most common in the dermis, with a significant proportion (approximately 17 percent) also appearing near fascial layers. This confirms the presence of deep lymphatic channels that could potentially be preserved during abdominoplasty or lipoabdominoplasty.
Collapse
|
16
|
Qu X, Zhou B, Scott Baldwin H. Tie1 is required for lymphatic valve and collecting vessel development. Dev Biol 2015; 399:117-128. [PMID: 25576926 DOI: 10.1016/j.ydbio.2014.12.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 12/10/2014] [Accepted: 12/17/2014] [Indexed: 12/29/2022]
Abstract
Tie1 is a receptor tyrosine kinase with broad expression in embryonic endothelium. Reduction of Tie1 levels in mouse embryos with a hypomorphic Tie1 allele resulted in abnormal lymphatic patterning and architecture, decreased lymphatic draining efficiency, and ultimately, embryonic demise. Here we report that Tie1 is present uniformly throughout the lymphatics and from late embryonic/early postnatal stages, becomes more restricted to lymphatic valve regions. To investigate later events of lymphatic development, we employed Cre-loxP recombination utilizing a floxed Tie1 allele and an Nfatc1Cre line, to provide loxP excision predominantly in lymphatic endothelium and developing valves. Interestingly, unlike the early prenatal defects previously described by ubiquitous endothelial deletion, excision of Tie1 with Nfatc1Cre resulted in abnormal lymphatic defects in postnatal mice and was characterized by agenesis of lymphatic valves and a deficiency of collecting lymphatic vessels. Attenuation of Tie1 signaling in lymphatic endothelium prevented initiation of lymphatic valve specification by Prox1 high expression lymphatic endothelial cells that is associated with the onset of turbulent flow in the lymphatic circulation. Our findings reveal a fundamental role for Tie1 signaling during lymphatic vessel remodeling and valve morphogenesis and implicate it as a candidate gene involved in primary lymphedema.
Collapse
Affiliation(s)
- Xianghu Qu
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, NY 10461, USA
| | - H Scott Baldwin
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Development Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
17
|
Yu P, Tung JK, Simons M. Lymphatic fate specification: an ERK-controlled transcriptional program. Microvasc Res 2014; 96:10-5. [PMID: 25132472 DOI: 10.1016/j.mvr.2014.07.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 10/24/2022]
Abstract
Lymphatic vessels are intimately involved in the regulation of water and solute homeostasis by returning interstitial fluid back to the venous circulation and play an equally important role in immune responses by providing avenues for immune cell transport. Defects in the lymphatic vasculature result in a number of pathological conditions, including lymphedema and lymphangiectasia. Knowledge of molecular mechanisms underlying lymphatic development and maintenance is therefore critical for understanding, prevention and treatment of lymphatic circulation-related diseases. Research in the past two decades has uncovered several key transcriptional factors (Prox1, Sox18 and Coup-TFII) controlling lymphatic fate specification. Most recently, ERK signaling has emerged as a critical regulator of this transcriptional program. This review summarizes our current understanding of lymphatic fate determination and its transcriptional controls.
Collapse
Affiliation(s)
- Pengchun Yu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT 06520, United States
| | - Joe K Tung
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT 06520, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT 06520, United States; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
18
|
Chen H, Griffin C, Xia L, Srinivasan RS. Molecular and cellular mechanisms of lymphatic vascular maturation. Microvasc Res 2014; 96:16-22. [PMID: 24928499 DOI: 10.1016/j.mvr.2014.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/03/2014] [Indexed: 01/24/2023]
Abstract
Lymphatic vasculature is necessary for maintaining fluid homeostasis in vertebrates. During embryogenesis lymphatic endothelial cells originate from the veins as a homogeneous population. These cells undergo a series of changes at the morphological and molecular levels to become mature lymphatic vasculature that consists of lymphatic capillaries, collecting lymphatic vessels and valves. In this article we summarize our current knowledge about these steps and highlight some black boxes that require further clarification.
Collapse
Affiliation(s)
- Hong Chen
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Courtney Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA.
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA.
| |
Collapse
|
19
|
Karpinich NO, Caron KM. Apelin signaling: new G protein-coupled receptor pathway in lymphatic vascular development. Arterioscler Thromb Vasc Biol 2014; 34:239-41. [PMID: 24431421 DOI: 10.1161/atvbaha.113.302905] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Natalie O Karpinich
- From the Departments of Cell Biology and Physiology (N.O.K., K.M.C.) and Genetics (K.M.C.), University of North Carolina at Chapel Hill, NC
| | | |
Collapse
|
20
|
Walsh D, Collins S, Winterford C, Pollitt C. The equine foot lamellar lymphatic system. J Equine Vet Sci 2013. [DOI: 10.1016/j.jevs.2013.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
James JM, Nalbandian A, Mukouyama YS. TGFβ signaling is required for sprouting lymphangiogenesis during lymphatic network development in the skin. Development 2013; 140:3903-14. [PMID: 23946447 DOI: 10.1242/dev.095026] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dermal lymphatic endothelial cells (LECs) emerge from the dorsolateral region of the cardinal veins within the anterior trunk to form an intricate, branched network of lymphatic vessels during embryogenesis. Multiple growth factors and receptors are required for specification and maintenance of LECs, but the mechanisms coordinating LEC movements and morphogenesis to develop three-dimensional lymphatic network architecture are not well understood. Here, we demonstrate in mice that precise LEC sprouting is a key process leading to stereotypical lymphatic network coverage throughout the developing skin, and that transforming growth factor β (TGFβ) signaling is required for LEC sprouting and proper lymphatic network patterning in vivo. We utilized a series of conditional mutants to ablate the TGFβ receptors Tgfbr1 (Alk5) and Tgfbr2 in LECs. To analyze lymphatic defects, we developed a novel, whole-mount embryonic skin imaging technique to visualize sprouting lymphangiogenesis and patterning at the lymphatic network level. Loss of TGFβ signaling in LECs leads to a severe reduction in local lymphangiogenic sprouting, resulting in a significant decrease in global lymphatic network branching complexity within the skin. Our results also demonstrate that TGFβ signaling negatively regulates LEC proliferation during lymphatic network formation. These data suggest a dual role for TGFβ signaling during lymphatic network morphogenesis in the skin, such that it enhances LEC sprouting and branching complexity while attenuating LEC proliferation.
Collapse
Affiliation(s)
- Jennifer M James
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
22
|
Cermenati S, Moleri S, Neyt C, Bresciani E, Carra S, Grassini DR, Omini A, Goi M, Cotelli F, François M, Hogan BM, Beltrame M. Sox18 Genetically Interacts With VegfC to Regulate Lymphangiogenesis in Zebrafish. Arterioscler Thromb Vasc Biol 2013; 33:1238-47. [DOI: 10.1161/atvbaha.112.300254] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Solei Cermenati
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Silvia Moleri
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Christine Neyt
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Erica Bresciani
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Silvia Carra
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Daniela R. Grassini
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Alice Omini
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Michela Goi
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Franco Cotelli
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Mathias François
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Benjamin M. Hogan
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| | - Monica Beltrame
- From the Dipartimento di Scienze Biomolecolari e Biotecnologie (S. Cermenati, S.M., D.R.G., M.G., M.B.), Dipartimento di Bioscienze (S. Cermenati, S.M., S. Carra, A.O., F.C., M.B.), and Dipartimento di Biologia (E.B., S. Carra, F.C.), Universita’ degli Studi di Milano, Milan, Italy; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia (C.N., M.F., B.M.H.)
| |
Collapse
|
23
|
Onder L, Danuser R, Scandella E, Firner S, Chai Q, Hehlgans T, Stein JV, Ludewig B. Endothelial cell-specific lymphotoxin-β receptor signaling is critical for lymph node and high endothelial venule formation. ACTA ACUST UNITED AC 2013; 210:465-73. [PMID: 23420877 PMCID: PMC3600902 DOI: 10.1084/jem.20121462] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of lymph nodes (LNs) and formation of LN stromal cell microenvironments is dependent on lymphotoxin-β receptor (LTβR) signaling. In particular, the LTβR-dependent crosstalk between mesenchymal lymphoid tissue organizer and hematopoietic lymphoid tissue inducer cells has been regarded as critical for these processes. Here, we assessed whether endothelial cell (EC)-restricted LTβR signaling impacts on LN development and the vascular LN microenvironment. Using EC-specific ablation of LTβR in mice, we found that conditionally LTβR-deficient animals failed to develop a significant proportion of their peripheral LNs. However, remnant LNs showed impaired formation of high endothelial venules (HEVs). Venules had lost their cuboidal shape, showed reduced segment length and branching points, and reduced adhesion molecule and constitutive chemokine expression. Due to the altered EC-lymphocyte interaction, homing of lymphocytes to peripheral LNs was significantly impaired. Thus, this study identifies ECs as an important LTβR-dependent lymphoid tissue organizer cell population and indicates that continuous triggering of the LTβR on LN ECs is critical for lymphocyte homeostasis.
Collapse
Affiliation(s)
- Lucas Onder
- Institute of Immunobiology, Kantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Jia L, Xie Z, Zheng J, Liu L, He Y, Liu F, He Y. Morphological studies of lymphatic labyrinths in the rat mesenteric lymph node. Anat Rec (Hoboken) 2012; 295:1291-301. [PMID: 22678756 DOI: 10.1002/ar.22509] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 02/25/2012] [Indexed: 01/02/2023]
Abstract
To supplement and correct the morphological features of lymphatic labyrinths (LLs) in rat mesenteric lymph node, the distribution, morphology and origin of LLs, and cellular elements in LLs, particularly the organization and integrity of the wall of LLs were examined by silver impregnation, transmission electron microscopy (TEM), scanning electron microscopy (SEM), and immunohistochemistry. LLs consisted of labyrinthine tubules and ran through not only the periphery of the deep cortical unit (DCU) but interfollicular cortex. LLs originated at the edge of the center of the DCU and of the follicle. At the site of their origin, the fibers in the wall of LL were continuous with the fibers located in the follicle and the center of DCU. The wall of LLs was a trilaminar membrane: a layer of flattened lymphatic endothelium; a layer of fibroblastic reticular cells; and amorphous substance and collagen fibers sandwiched between the above two layers. Under SEM and TEM, the whole amoeboid lymphocytes were moving through the pores in the wall of LL, which showed that lymphocytes end their journey through the paracortical cord by migrating into LLs. Immunohistochemical lymphatic vessel endothelial hyaluronan receptor-1 expression was present in cells lining the LLs and intraluminal stellate cells, which may belong to the "sinus endothelial/virgultar cells." LLs are specific channels that are different from lymphatic sinuses. LL may be regarded as a special part of lymphatic vascular system in lymph nodes. We confirm that LLs are important transport pathway of lymphocytes in lymph nodes. The structural framework of LLs facilitates the migration of lymphocytes.
Collapse
Affiliation(s)
- Limin Jia
- Department of Anatomy, Harbin Medical University, Harbin, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Tanabe M, Shimizu A, Masuda Y, Kataoka M, Ishikawa A, Wakamatsu K, Mii A, Fujita E, Higo S, Kaneko T, Kawachi H, Fukuda Y. Development of lymphatic vasculature and morphological characterization in rat kidney. Clin Exp Nephrol 2012; 16:833-42. [PMID: 22581062 DOI: 10.1007/s10157-012-0637-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 04/12/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND The mechanisms and morphological characteristics of lymphatic vascular development in embryonic kidneys remain uncertain. METHODS We examined the distribution and characteristics of lymphatic vessels in developing rat kidneys using immunostaining for podoplanin, prox-1, Ki-67, type IV collagen (basement membrane: BM), and α-smooth muscle actin (αSMA: pericytes or mural cells). We also examined the expression of VEGF-C. RESULTS At embryonic day 17 (E17), podoplanin-positive lymphatic vessels were observed mainly in the kidney hilus. At E20, lymphatic vessels extended further into the developing kidneys along the interlobar vasculature. In 1-day-old pups (P1) to P20, lymphatic vessels appeared around the arcuate arteries and veins of the kidneys, with some reaching the developing cortex via interlobular vessels. In 8-week-old adult rats, lymphatic vessels were extensively distributed around the blood vasculature from the renal hilus to cortex. Only lymphatic capillaries lacking continuous BM and αSMA-positive cells were present within adult kidneys, with none observed in renal medulla. VEGF-C was upregulated in the developing kidneys and expressed mainly in tubules. Importantly, the developing lymphatic vessels were characterized by endothelial cells immunopositive for podoplanin, prox-1, and Ki-67, with no surrounding BM or αSMA-positive cells. CONCLUSION During nephrogenesis, lymphatic vessels extend from the renal hilus into the renal cortex along the renal blood vasculature. Podoplanin, prox-1, Ki-67, type IV collagen, and αSMA immunostaining can detect lymphatic vessels during lymphangiogenesis.
Collapse
Affiliation(s)
- Maki Tanabe
- Department of Analytic Human Pathology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Dynamics of airway blood vessels and lymphatics: lessons from development and inflammation. Ann Am Thorac Soc 2012; 8:504-7. [PMID: 22052927 DOI: 10.1513/pats.201102-022mw] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Blood vessels and lymphatic vessels in the respiratory tract play key roles in inflammation. By undergoing adaptive remodeling and growth, blood vessels undergo changes that enable the extravasation of plasma and leukocytes into inflamed tissues, and lymphatic vessels adjust to the increased fluid clearance and cell traffic involved in immune responses. Blood vessels and lymphatics in adult airways are strikingly different from those of late-stage embryos. Before birth, blood vessels in mouse airways make up a primitive plexus similar to that of the yolk sac. This plexus undergoes rapid and extensive remodeling at birth. In the early neonatal period, parts of the plexus regress. Capillaries then rapidly regrow, and with arterioles and venules form the characteristic adult vascular pattern. Lymphatic vessels of the airways also undergo rapid changes around birth, when lymphatic endothelial cells develop button-like intercellular junctions specialized for efficient fluid uptake. Among the mechanisms that underlie the onset of rapid vascular remodeling at birth, changes in tissue oxygen tension and mechanical forces associated with breathing are likely to be involved, along with growth factors that promote the growth and maturation of blood vessels and lymphatics. Whatever the mechanisms, the dynamic nature of airway blood vessels and lymphatics during perinatal development foretells the extraordinary vascular plasticity found in many diseases.
Collapse
|
28
|
Intravital two-photon microscopy of lymphatic vessel development and function using a transgenic Prox1 promoter-directed mOrange2 reporter mouse. Biochem Soc Trans 2012; 39:1674-81. [PMID: 22103506 DOI: 10.1042/bst20110722] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Lymphatic vessels, the second vascular system of higher vertebrates, are indispensable for fluid tissue homoeostasis, dietary fat resorption and immune surveillance. Not only are lymphatic vessels formed during fetal development, when the lymphatic endothelium differentiates and separates from blood endothelial cells, but also lymphangiogenesis occurs during adult life under conditions of inflammation, wound healing and tumour formation. Under all of these conditions, haemopoietic cells can exert instructive influences on lymph vessel growth and are essential for the vital separation of blood and lymphatic vessels. LECs (lymphatic endothelial cells) are characterized by expression of a number of unique genes that distinguish them from blood endothelium and can be utilized to drive reporter genes in a lymph endothelial-specific fashion. In the present paper, we describe the Prox1 (prospero homeobox protein 1) promoter-driven expression of the fluorescent protein mOrange2, which allows the specific intravital visualization of lymph vessel growth and behaviour during mouse fetal development and in adult mice.
Collapse
|
29
|
Datar SA, Johnson EG, Oishi PE, Johengen M, Tang E, Aramburo A, Barton J, Kuo HC, Bennett S, Xoinis K, Reel B, Kalkan G, Sajti E, Osorio O, Raff GW, Matthay MA, Fineman JR. Altered lymphatics in an ovine model of congenital heart disease with increased pulmonary blood flow. Am J Physiol Lung Cell Mol Physiol 2011; 302:L530-40. [PMID: 22207591 DOI: 10.1152/ajplung.00324.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abnormalities of the lymphatic circulation are well recognized in patients with congenital heart defects. However, it is not known how the associated abnormal blood flow patterns, such as increased pulmonary blood flow (PBF), might affect pulmonary lymphatic function and structure. Using well-established ovine models of acute and chronic increases in PBF, we cannulated the efferent lymphatic duct of the caudal mediastinal node and collected and analyzed lymph effluent from the lungs of lambs with acutely increased PBF (n = 6), chronically increased PBF (n = 6), and age-matched normal lambs (n = 8). When normalized to PBF, we found that lymph flow was unchanged following acute increases in PBF but decreased following chronic increases in PBF. The lymph:plasma protein ratio decreased with both acute and chronic increases in PBF. Lymph bioavailable nitric oxide increased following acute increases in PBF but decreased following chronic increases in PBF. In addition, we found perturbations in the transit kinetics of contrast material through the pleural lymphatics of lambs with chronic increases in PBF. Finally, there were structural changes in the pulmonary lymphatic system in lambs with chronic increases in PBF: lymphatics from these lambs were larger and more dilated, and there were alterations in the expression of vascular endothelial growth factor-C, lymphatic vessel endothelial hyaluronan receptor-1, and Angiopoietin-2, proteins known to be important for lymphatic growth, development, and remodeling. Taken together these data suggest that chronic increases in PBF lead to both functional and structural aberrations of lung lymphatics. These findings have important therapeutic implications that warrant further study.
Collapse
Affiliation(s)
- Sanjeev A Datar
- Department of Pediatrics, University of California, San Francisco, 94143-0106, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Alitalo A, Detmar M. Interaction of tumor cells and lymphatic vessels in cancer progression. Oncogene 2011; 31:4499-508. [PMID: 22179834 DOI: 10.1038/onc.2011.602] [Citation(s) in RCA: 288] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastatic spread of cancer through the lymphatic system affects hundreds of thousands of patients yearly. Growth of new lymphatic vessels, lymphangiogenesis, is activated in cancer and inflammation, but is largely inactive in normal physiology, and therefore offers therapeutic potential. Key mediators of lymphangiogenesis have been identified in developmental studies. During embryonic development, lymphatic endothelial cells derive from the blood vascular endothelium and differentiate under the guidance of lymphatic-specific regulators, such as the prospero homeobox 1 transcription factor. Vascular endothelial growth factor-C (VEGF-C) and VEGF receptor 3 signaling are essential for the further development of lymphatic vessels and therefore they provide a promising target for inhibition of tumor lymphangiogenesis. Lymphangiogenesis is important for the progression of solid tumors as shown for melanoma and breast cancer. Tumor cells may use chemokine gradients as guidance cues and enter lymphatic vessels through intercellular openings between endothelial cell junctions or, possibly, by inducing larger discontinuities in the endothelial cell layer. Tumor-draining sentinel lymph nodes show enhanced lymphangiogenesis even before cancer metastasis and they may function as a permissive 'lymphovascular niche' for the survival of metastatic cells. Although our current knowledge indicates that the development of anti-lymphangiogenic therapies may be beneficial for the treatment of cancer patients, several open questions remain with regard to the frequency, mechanisms and biological importance of lymphatic metastases.
Collapse
Affiliation(s)
- A Alitalo
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
31
|
Srinivasan RS, Oliver G. Prox1 dosage controls the number of lymphatic endothelial cell progenitors and the formation of the lymphovenous valves. Genes Dev 2011; 25:2187-97. [PMID: 22012621 DOI: 10.1101/gad.16974811] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Arteries, veins, and lymphatic vessels are functionally linked, and their physical interaction is tightly regulated. The lymphatic vessels communicate with the blood vessels only at the junction of the jugular and subclavian veins. Here, we characterize the embryonic lymphovenous valves controlling this vital communication and show that they are formed by the intercalation of lymphatic endothelial cells (LECs) with a subpopulation of venous endothelial cells (ECs) at the junction of the jugular and subclavian veins. We found that unlike LEC progenitors, which move out from the veins and differentiate into mature LECs, these Prox1-expressing ECs remain in the veins and do not acquire LEC features. We demonstrate that the development of this Prox1-expressing venous EC population, and therefore of lymphovenous valves, requires two functional copies of Prox1, as the valves are absent in Prox1 heterozygous mice. We show that this is due to a defect in the maintenance of Prox1 expression in venous ECs and LEC progenitors promoted by a reduction in Coup-TFII/Prox1 complex formation. This is the first report describing the molecular mechanism controlling lymphovenous communication.
Collapse
Affiliation(s)
- R Sathish Srinivasan
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | |
Collapse
|
32
|
Guo W, Zhu Y, Yu PK, Yu X, Sun X, Cringle SJ, Su EN, Yu DY. Quantitative study of the topographic distribution of conjunctival lymphatic vessels in the monkey. Exp Eye Res 2011; 94:90-7. [PMID: 22138558 DOI: 10.1016/j.exer.2011.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 11/15/2011] [Accepted: 11/15/2011] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to quantify the topographic distribution of bulbar conjunctival microlymphatic vessels in the monkey. Sixteen eyes from 8 rhesus monkeys were used. Full thickness pieces of globe wall were excised from each quadrant. Cryosections were stained for 5'-nucleotidase, an enzyme histochemical staining for lymphatic vessels, or vascular endothelial growth factor receptor-3, an immunohistochemical marker for the identification of lymphatic endothelial cells, and then counterstained by hematoxylin. The remaining bulbar conjunctiva was dissected and flat mounted. The tissue was then processed with 5'-nucleotidase and alkaline phosphatase, an enzyme histochemical stain with higher activity in blood vessels. Microscope images were further analysed by image processing. The density of lymphatics, diameter of lymphatic vessels, and the size of the drainage zone of each blind end of the initial lymphatics were studied. Conjunctival lymphatics consisted of initial lymphatics and pre-collectors. The initial lymphatics with blind ends were predominately distributed just under the epithelium. The density of these lymphatics (∼50%) and the drainage zone area (∼0.81 mm(2)) was similar in each quadrant, with no difference in the limbus and fornix regions. The average diameter of lymphatic vessels in each quadrant ranged from 82 to 111 μm, and was greater in the superior and nasal regions. Larger calibre pre-collectors with valve-like structures were mostly located sub Tenon's membrane and predominantly located in the region mid-way between the limbus and fornix. There was a marked depth difference in initial lymphatic distribution, with the initial lymphatics mostly confined to the region between Tenon's membrane and the conjunctival epithelium. Detailed knowledge of the topographic distribution of conjunctival lymphatics have significant relevance to a better understanding of immunology, drug delivery, glaucoma filtration surgery, and tumour metastasis in the conjunctiva.
Collapse
Affiliation(s)
- Wenyi Guo
- Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ebina M, Shibata N, Ohta H, Hisata S, Tamada T, Ono M, Okaya K, Kondo T, Nukiwa T. The disappearance of subpleural and interlobular lymphatics in idiopathic pulmonary fibrosis. Lymphat Res Biol 2011; 8:199-207. [PMID: 21190492 DOI: 10.1089/lrb.2010.0008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The lymphatics in the interlobular and subpleural parenchyma contribute to alveolar clearance in the lung, but the information on the remodeling of these lymphatics is quite limited in idiopathic pulmonary fibrosis lungs that contain severe fibrosis in these regions. We compared the alteration of these lymphatics and lymphangiogenesis among idiopathic pulmonary fibrosis and nonfibrotic interstitial pneumonias with a better prognosis. METHODS AND RESULTS The lung tissue specimens of eighteen patients with idiopathic pulmonary fibrosis (ten surgical biopsies and eight autopsies), six with organizing pneumonia, six with cellular nonspecific interstitial pneumonia, and five normal controls were examined by morphometric analysis of the lymphatics identified by immunohistochemistry. In addition, three-dimensional reconstruction of lymphatics, apoptosis of lymphatic endothelial cells and the cells producing growth factors for lymphangiogenesis were also evaluated. Both the subpleural and the interlobular lymphatics in idiopathic pulmonary fibrosis lungs were significantly decreased in the severe fibroconnective lesions, with rare lymphangiogenesis. The three-dimensional images of the subpleural lymphatics in idiopathic pulmonary fibrosis clearly revealed destruction by fibrosis; apoptosis was observed in these lymphatic endothelial cells. In contrast, organizing pneumonia and cellular nonspecific interstitial pneumonia preserved these lymphatics, and active lymphangiogenesis occurred in the alveolar lesions. CONCLUSIONS These results reveal severe damage of the subpleural and interlobular lymphatics in idiopathic pulmonary fibrosis lungs, and suggest impaired alveolar clearance as another pathogenesis of its refractoriness.
Collapse
Affiliation(s)
- Masahito Ebina
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kanady JD, Dellinger MT, Munger SJ, Witte MH, Simon AM. Connexin37 and Connexin43 deficiencies in mice disrupt lymphatic valve development and result in lymphatic disorders including lymphedema and chylothorax. Dev Biol 2011; 354:253-66. [PMID: 21515254 PMCID: PMC3134316 DOI: 10.1016/j.ydbio.2011.04.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 04/06/2011] [Accepted: 04/08/2011] [Indexed: 12/31/2022]
Abstract
Intraluminal valves are required for the proper function of lymphatic collecting vessels and large lymphatic trunks like the thoracic duct. Despite recent progress in the study of lymphvasculogenesis and lymphangiogenesis, the molecular mechanisms controlling the morphogenesis of lymphatic valves remain poorly understood. Here, we report that gap junction proteins, or connexins (Cxs), are required for lymphatic valvulogenesis. Cx37 and Cx43 are expressed early in mouse lymphatic development in the jugular lymph sacs, and later in development these Cxs become enriched and differentially expressed by lymphatic endothelial cells on the upstream and downstream sides of the valves. Specific deficiencies of Cx37 and Cx43 alone or in combination result in defective valve formation in lymphatic collecting vessels, lymphedema, and chylothorax. We also show that Cx37 regulates jugular lymph sac size and that both Cx37 and Cx43 are required for normal thoracic duct development, including valve formation. Another Cx family member, Cx47, whose human analog is mutated in some families with lymphedema, is also highly enriched in a subset of endothelial cells in lymphatic valves. Mechanistically, we present data from Foxc2-/- embryos suggesting that Cx37 may be a target of regulation by Foxc2, a transcription factor that is mutated in human lymphedema-distichiasis syndrome. These results show that at least three Cxs are expressed in the developing lymphatic vasculature and, when defective, are associated with clinically manifest lymphatic disorders in mice and man.
Collapse
Affiliation(s)
- John D. Kanady
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| | - Michael T. Dellinger
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85724, USA
| | | | - Marlys H. Witte
- Department of Surgery, University of Arizona, Tucson, AZ 85724, USA
| | | |
Collapse
|
35
|
Rouzaut A, Garasa S, Teijeira A, González I, Martinez-Forero I, Suarez N, Larrea E, Alfaro C, Palazón A, Dubrot J, Hervás-Stubbs S, Melero I. Dendritic cells adhere to and transmigrate across lymphatic endothelium in response to IFN-α. Eur J Immunol 2010; 40:3054-63. [PMID: 21061437 DOI: 10.1002/eji.201040523] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 08/06/2010] [Accepted: 08/26/2010] [Indexed: 02/01/2023]
Abstract
Migration of DC into lymphatic vessels ferries antigenic cargo and pro-inflammatory stimuli into the draining LN. Given that tissues under the influence of viral infections produce type I IFN, it is conceivable that these cytokines enhance DC migration in order to facilitate an antiviral immune response. Cultured lymphatic endothelium monolayers pretreated with TNF-α were used to model this phenomenon under inflammatory conditions. DC differentiated in the presence of either IFN-α2b or IFN-α5 showed enhanced adhesion to cultured lymphatic endothelial cells. These pro-adhesive effects were mediated by DC, not the lymphatic endothelium, and correlated with increased DC transmigration across lymphatic endothelial cell monolayers. Transmigration was guided by chemokines acting on DC, and blocking experiments with mAb indicated a role for LFA-1. Furthermore, incubation of DC with IFN-α led to the appearance of active conformation epitopes on the CD11a integrin chains expressed by DC. Differentiation of mouse DC in the presence of IFN-α also increased DC migration from inflammed footpads toward popliteal LN. Collectively, these results indicate a role for type I IFN in directing DC toward LN under inflammatory conditions.
Collapse
Affiliation(s)
- Ana Rouzaut
- Center for Applied Medical Research, School of Medicine, University of Navarra, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wang Y, Oliver G. Current views on the function of the lymphatic vasculature in health and disease. Genes Dev 2010; 24:2115-26. [PMID: 20889712 DOI: 10.1101/gad.1955910] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lymphatic vascular system is essential for lipid absorption, fluid homeostasis, and immune surveillance. Until recently, lymphatic vessel dysfunction had been associated with symptomatic pathologic conditions such as lymphedema. Work in the last few years had led to a better understanding of the functional roles of this vascular system in health and disease. Furthermore, recent work has also unraveled additional functional roles of the lymphatic vasculature in fat metabolism, obesity, inflammation, and the regulation of salt storage in hypertension. In this review, we summarize the functional roles of the lymphatic vasculature in health and disease.
Collapse
Affiliation(s)
- Yingdi Wang
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | |
Collapse
|
37
|
Nguyen VPKH, Hanna G, Rodrigues N, Pizzuto K, Yang E, Van Slyke P, Kim H, Chen SH, Dumont DJ. Differential proteomic analysis of lymphatic, venous, and arterial endothelial cells extracted from bovine mesenteric vessels. Proteomics 2010; 10:1658-72. [PMID: 20186751 DOI: 10.1002/pmic.200900594] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Differential protein profiling by 2-D PAGE is generally useful in biomarker discovery, proteome analysis and routine sample preparation prior to analysis by MS. The goal of this study was to compare 2-D PAGE-resolved protein profile of lymphatic endothelial cells to those of venous, and arterial endothelial cells isolated from lymphatic and blood vessels of bovine mesentery (bm). Three 2-D PAGE electrophoretograms were produced for each of the three cell types and quantitatively analyzed. Protein identification by LC-MS/MS was performed to identify 39 proteins found to be present at statistically significantly different levels in the three cell types (p<0.05). Most of the 39 proteins have not been previously reported in EC proteomic studies of 2-D PAGE electrophoretograms. Three proteins, HSPA1B (HSP70 family member), HSPB1 (HSP27 family member), and UBE2D3 (a member of E2 ubiquitin-conjugating enzymes) found to be at highest levels in bm arterial endothelial cells, bm venous endothelial cells, and bm lymphatic endothelial cells, respectively, were validated by immunoblotting with appropriate antibodies. The lack of substantial overlap between our results and those of other groups' comparative studies are discussed. Functional implications of differences in levels of various proteins identified in the three cell types are also discussed.
Collapse
Affiliation(s)
- Vicky P K H Nguyen
- Molecular and Cellular Biology Research, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mkonyi LE, Bletsa A, Fristad I, Wiig H, Berggreen E. Importance of lymph vessels in the transcapillary fluid balance in the gingiva studied in a transgenic mouse model. Am J Physiol Heart Circ Physiol 2010; 299:H275-83. [DOI: 10.1152/ajpheart.01199.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The gingiva is frequently challenged by oral bacterial products leading to inflammatory responses such as increased fluid filtration and edema formation. The role of initial lymphatics for transcapillary fluid balance in the gingiva is unknown and was therefore investigated in genetically engineered K14-VEGF receptor 3-Ig (K14) lymphedema mice. The mutant mice demonstrated a total lack of lymphatics in the gingiva, whereas lymphatics were found in the submucosal parts of the alveolar mucosa, although they were almost completely absent in the mucosa. In wild-type (WT) mice, lymphatic vessels were detected in mucosal and submucosal parts of the alveolar mucosa. Interstitial fluid pressure (Pif) measured with micropipettes was increased in the gingiva of K14 mice in the normal situation ( P < 0.001) and after inflammation ( P < 0.01) induced by lipopolysaccharide from the oral bacteria Porphyromonas gingivalis compared with WT littermates. Fluid volume expansion caused a >75% increase in interstitial fluid volume followed by a drop in Pif after recovery in both strains. Continuous measurements during the expansion showed an increase in Pif followed by a decline, suggesting that compliance is increased after the disruption of the extracellular matrix during edema formation. In the alveolar mucosa, no strain differences were observed in Pif in the normal situation or after fluid volume expansion, suggesting that lymph vessels in the mucosa are not critical for tissue fluid regulation in any situation. Our study demonstrates an important role of gingival lymphatics in transcapillary fluid balance in the steady-state condition and during acute perturbations.
Collapse
Affiliation(s)
| | - Athanasia Bletsa
- Departments of 1Biomedicine and
- Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Inge Fristad
- Clinical Dentistry, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|
39
|
Rockson SG. Current concepts and future directions in the diagnosis and management of lymphatic vascular disease. Vasc Med 2010; 15:223-31. [DOI: 10.1177/1358863x10364553] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Despite the central, complex role for the lymphatic system in the maintenance of human health, the biology of this important and complex vasculature has been relatively under-investigated. However, the last decade has witnessed a substantial growth in the elucidation of lymphatic structural biology and the function of this system in health and in disease. These newly gained insights can be used to formulate our evolving concepts about the diagnostic and therapeutic approaches to patients with lymphatic vascular disorders. In lymphedema, there is a spectrum of disease that extends from primary (heritable) to secondary (acquired) causes. Once detected, the presence of lymphatic edema mandates very specific modalities of intervention, predominated by physiotherapeutic techniques. In addition, a physiological basis for adjunctive, intermittent pneumatic compression has been established, and these modalities may be indicated in selected patient populations. The acknowledgement of a unique biology in lymphatic edemas is, increasingly, guiding research efforts within this field. Increasing investigative attention is being directed toward animal models of lymphatic vascular disease. As insight into the complex biology of the lymphatic vasculature continues to expand through focused biomedical investigation, the translation of these mechanistic insights into targeted, rationally conceived therapeutics will become increasingly feasible.
Collapse
Affiliation(s)
- Stanley G Rockson
- Stanford Center for Lymphatic and Venous Disorders, Stanford University School of Medicine, Stanford, California, USA,
| |
Collapse
|
40
|
Srinivasan RS, Geng X, Yang Y, Wang Y, Mukatira S, Studer M, Porto MPR, Lagutin O, Oliver G. The nuclear hormone receptor Coup-TFII is required for the initiation and early maintenance of Prox1 expression in lymphatic endothelial cells. Genes Dev 2010; 24:696-707. [PMID: 20360386 DOI: 10.1101/gad.1859310] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The homeobox gene Prox1 is crucial for mammalian lymphatic vascular development. In the absence of Prox1, lymphatic endothelial cells (LECs) are not specified. The maintenance of LEC identity also requires the constant expression of Prox1. However, the mechanisms controlling the expression of this gene in LECs remain poorly understood. The SRY-related gene Sox18 is required to induce Prox1 expression in venous LEC progenitors. Although Sox18 is also expressed in embryonic arteries, these vessels do not express Prox1, nor do they give rise to LECs. This finding suggests that some venous endothelial cell-specific factor is required for the activation of Prox1. Here we demonstrate that the nuclear hormone receptor Coup-TFII is necessary for the activation of Prox1 in embryonic veins by directly binding a conserved DNA domain in the regulatory region of Prox1. In addition, we show that the direct interaction between nuclear hormone receptors and Prox1 is also necessary for the maintenance of Prox1 expression during early stages of LEC specification and differentiation.
Collapse
Affiliation(s)
- R Sathish Srinivasan
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Oliver G, Srinivasan RS. Endothelial cell plasticity: how to become and remain a lymphatic endothelial cell. Development 2010; 137:363-72. [PMID: 20081185 DOI: 10.1242/dev.035360] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Lineage commitment and differentiation into mature cell types are mostly considered to be unidirectional and irreversible processes. However, recent results have challenged this by showing that terminally differentiated cell types can be reprogrammed into other cell types, an important step towards devising strategies for gene therapy and tissue regeneration. In this Review, we summarize recent data on the earliest steps in the development of the mammalian lymphatic vasculature: the specification of lymphatic endothelial cells (LECs). We elaborate on a developmental model that integrates the different steps leading to LEC differentiation and lymphatic network formation, discuss evidence that suggests that LEC fate is plastic, and consider the potentially far-reaching implications of the ability to convert one cell type into another.
Collapse
Affiliation(s)
- Guillermo Oliver
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| | | |
Collapse
|
42
|
Qu X, Tompkins K, Batts LE, Puri M, Baldwin HS, Baldwin S. Abnormal embryonic lymphatic vessel development in Tie1 hypomorphic mice. Development 2010; 137:1285-95. [PMID: 20223757 DOI: 10.1242/dev.043380] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tie1 is an endothelial receptor tyrosine kinase that is essential for development and maintenance of the vascular system; however, the role of Tie1 in development of the lymphatic vasculature is unknown. To address this question, we first documented that Tie1 is expressed at the earliest stages of lymphangiogenesis in Prox1-positive venous lymphatic endothelial cell (LEC) progenitors. LEC Tie1 expression is maintained throughout embryonic development and persists in postnatal mice. We then generated two lines of Tie1 mutant mice: a hypomorphic allele, which has reduced expression of Tie1, and a conditional allele. Reduction of Tie1 levels resulted in abnormal lymphatic patterning and in dilated and disorganized lymphatic vessels in all tissues examined and in impaired lymphatic drainage in embryonic skin. Homozygous hypomorphic mice also exhibited abnormally dilated jugular lymphatic vessels due to increased production of Prox1-positive LECs during initial lymphangiogenesis, indicating that Tie1 is required for the early stages of normal lymphangiogenesis. During later stages of lymphatic development, we observed an increase in LEC apoptosis in the hypomorphic embryos after mid-gestation that was associated with abnormal regression of the lymphatic vasculature. Therefore, Tie1 is required for early LEC proliferation and subsequent survival of developing LECs. The severity of the phenotypes observed correlated with the expression levels of Tie1, confirming a dosage dependence for Tie1 in LEC integrity and survival. No defects were observed in the arterial or venous vasculature. These results suggest that the developing lymphatic vasculature is particularly sensitive to alterations in Tie1 expression.
Collapse
Affiliation(s)
- Xianghu Qu
- Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
43
|
Xu Y, Yuan L, Mak J, Pardanaud L, Caunt M, Kasman I, Larrivée B, Del Toro R, Suchting S, Medvinsky A, Silva J, Yang J, Thomas JL, Koch AW, Alitalo K, Eichmann A, Bagri A. Neuropilin-2 mediates VEGF-C-induced lymphatic sprouting together with VEGFR3. ACTA ACUST UNITED AC 2010; 188:115-30. [PMID: 20065093 PMCID: PMC2812843 DOI: 10.1083/jcb.200903137] [Citation(s) in RCA: 266] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
If neuropilin-2 and the growth factor VEGF-C don’t come together, lymphatic vessels don’t branch apart. Vascular sprouting is a key process-driving development of the vascular system. In this study, we show that neuropilin-2 (Nrp2), a transmembrane receptor for the lymphangiogenic vascular endothelial growth factor C (VEGF-C), plays an important role in lymphatic vessel sprouting. Blocking VEGF-C binding to Nrp2 using antibodies specifically inhibits sprouting of developing lymphatic endothelial tip cells in vivo. In vitro analyses show that Nrp2 modulates lymphatic endothelial tip cell extension and prevents tip cell stalling and retraction during vascular sprout formation. Genetic deletion of Nrp2 reproduces the sprouting defects seen after antibody treatment. To investigate whether this defect depends on Nrp2 interaction with VEGF receptor 2 (VEGFR2) and/or 3, we intercrossed heterozygous mice lacking one allele of these receptors. Double-heterozygous nrp2vegfr2 mice develop normally without detectable lymphatic sprouting defects. In contrast, double-heterozygote nrp2vegfr3 mice show a reduction of lymphatic vessel sprouting and decreased lymph vessel branching in adult organs. Thus, interaction between Nrp2 and VEGFR3 mediates proper lymphatic vessel sprouting in response to VEGF-C.
Collapse
Affiliation(s)
- Yunling Xu
- Institut National de la Santé et de la Recherche Médicale, Unité 833, 75005 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Clinically and on a molecular level, ovarian cancer is a unique and complex disease. The explosion in potential molecular targets over the last decade has led to the arrival of many novel therapies into oncology. In the present article, we review the most promising of these agents in ovarian cancer. RECENT FINDINGS Targeted therapies, such as epidermal growth factor receptor inhibitors, that have worked well in other cancers have shown only moderate success in ovarian cancer, whereas other treatment approaches have yielded surprisingly positive outcomes. An example is anti-vascular endothelial growth factor and proapoptotic strategies, which are effective in both primary and relapsed ovarian cancer. Use of poly (ADP-ribose)-polymerase inhibitors has shown that targeting one form of DNA repair profoundly affects cell survival in those with a hereditary failure to mend DNA damage using another mechanism. This can be extrapolated to patients with sporadic ovarian cancers, with or without the 'BRCAness' phenotype. SUMMARY Using targeted agents in ovarian cancer, we are discovering not only how these novel therapies work but are also unveiling the complex 'wiring' of the disease itself, and the interconnections between what were previously believed to be distinct molecular pathways. The addition of targeted agents to our therapeutic armoury is likely to significantly and positively impact on patient survival.
Collapse
|
45
|
Pham THM, Baluk P, Xu Y, Grigorova I, Bankovich AJ, Pappu R, Coughlin SR, McDonald DM, Schwab SR, Cyster JG. Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning. ACTA ACUST UNITED AC 2009; 207:17-27. [PMID: 20026661 PMCID: PMC2812554 DOI: 10.1084/jem.20091619] [Citation(s) in RCA: 372] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Lymphocyte egress from lymph nodes (LNs) is dependent on sphingosine-1-phosphate (S1P), but the cellular source of this S1P is not defined. We generated mice that expressed Cre from the lymphatic vessel endothelial hyaluronan receptor 1 (Lyve-1) locus and that showed efficient recombination of loxP-flanked genes in lymphatic endothelium. We report that mice with Lyve-1 CRE-mediated ablation of sphingosine kinase (Sphk) 1 and lacking Sphk2 have a loss of S1P in lymph while maintaining normal plasma S1P. In Lyve-1 Cre+ Sphk-deficient mice, lymphocyte egress from LNs and Peyer's patches is blocked. Treatment with pertussis toxin to overcome Gαi-mediated retention signals restores lymphocyte egress. Furthermore, in the absence of lymphatic Sphks, the initial lymphatic vessels in nonlymphoid tissues show an irregular morphology and a less organized vascular endothelial cadherin distribution at cell–cell junctions. Our data provide evidence that lymphatic endothelial cells are an in vivo source of S1P required for lymphocyte egress from LNs and Peyer's patches, and suggest a role for S1P in lymphatic vessel maturation.
Collapse
Affiliation(s)
- Trung H M Pham
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Johnson NC, Dillard ME, Baluk P, McDonald DM, Harvey NL, Frase SL, Oliver G. Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity. Genes Dev 2009; 22:3282-91. [PMID: 19056883 DOI: 10.1101/gad.1727208] [Citation(s) in RCA: 266] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The activity of the homeobox gene Prox1 is necessary and sufficient for venous blood endothelial cells (BECs) to acquire a lymphatic endothelial cell (LEC) fate. We determined that the differentiated LEC phenotype is a plastic, reprogrammable condition that depends on constant Prox1 activity for its maintenance. We show that conditional down-regulation of Prox1 during embryonic, postnatal, or adult stages is sufficient to reprogram LECs into BECs. Consequently, the identity of the mutant lymphatic vessels is also partially reprogrammed as they acquire some features typical of the blood vasculature. siRNA-mediated down-regulation of Prox1 in LECs in culture demonstrates that reprogramming of LECs into BECs is a Prox1-dependent, cell-autonomous process. We propose that Prox1 acts as a binary switch that suppresses BEC identity and promotes and maintains LEC identity; switching off Prox1 activity is sufficient to initiate a reprogramming cascade leading to the dedifferentiation of LECs into BECs. Therefore, LECs are one of the few differentiated cell types that require constant expression of a certain gene to maintain their phenotypic identity.
Collapse
Affiliation(s)
- Nicole C Johnson
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
De Val S, Black BL. Transcriptional control of endothelial cell development. Dev Cell 2009; 16:180-95. [PMID: 19217421 DOI: 10.1016/j.devcel.2009.01.014] [Citation(s) in RCA: 270] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Revised: 01/26/2009] [Accepted: 01/26/2009] [Indexed: 12/14/2022]
Abstract
The transcription factors that regulate endothelial cell development have been a focus of active research for several years, and many players in the endothelial transcriptional program have been identified. This review discusses the function of several major regulators of endothelial transcription, including members of the Sox, Ets, Forkhead, GATA, and Kruppel-like families. This review also highlights recent developments aimed at unraveling the combinatorial mechanisms and transcription factor interactions that regulate endothelial cell specification and differentiation during vasculogenesis and angiogenesis.
Collapse
Affiliation(s)
- Sarah De Val
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, 94158, USA
| | | |
Collapse
|
48
|
Dunworth WP, Caron KM. G protein-coupled receptors as potential drug targets for lymphangiogenesis and lymphatic vascular diseases. Arterioscler Thromb Vasc Biol 2009; 29:650-6. [PMID: 19265032 DOI: 10.1161/atvbaha.109.185066] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are widely expressed cell surface receptors that have been successfully exploited for the treatment of a variety of human diseases. Recent studies in genetically engineered mouse models have led to the identification of several GPCRs important for lymphatic vascular development and function. The adrenomedullin receptor, which consists of an oligomer between calcitonin receptor-like receptor and receptor activity modifying protein 2, is required for normal lymphatic vascular development and regulates lymphatic capillary permeability in mice. Numerous studies also suggest that lysophospholipid receptors are involved in the development of lymphatic vessels and lymphatic endothelial cell permeability. Given our current lack of pharmacological targets for the treatment of lymphatic vascular diseases like lymphedema, the continued identification and study of GPCRs in lymphatic endothelial cells may eventually lead to major breakthroughs and new pharmacological strategies for the treatment of lymphedema.
Collapse
Affiliation(s)
- William P Dunworth
- Department of Cell and Molecular Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|