1
|
Polacheck WJ, Dixon JB, Aw WY. Understanding the Lymphatic System: Tissue-on-Chip Modeling. Annu Rev Biomed Eng 2025; 27:73-100. [PMID: 39841937 DOI: 10.1146/annurev-bioeng-110222-100246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The lymphatic vasculature plays critical roles in maintaining fluid homeostasis, transporting lipid, and facilitating immune surveillance. A growing body of work has identified lymphatic dysfunction as contributing to the severity of myriad diseases and to systemic inflammation, as well as modulating drug responses. Here, we review efforts to reconstruct lymphatic vessels in vitro toward establishing humanized, functional models to advance understanding of lymphatic biology and pathophysiology. We first review lymphatic endothelial cell biology and the biophysical lymphatic microenvironment, with a focus on features that are unique to the lymphatics and that have been used as design parameters for lymphatic-on-chip devices. We then discuss the state of the art for recapitulating lymphatic function in vitro, and we acknowledge limitations and challenges to current approaches. Finally, we discuss opportunities and the need for further development of microphysiological lymphatic systems to bridge the gap in model systems between lymphatic cell culture and animal physiology.
Collapse
Affiliation(s)
- William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina, USA;
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J Brandon Dixon
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina, USA;
| |
Collapse
|
2
|
Kuonqui KG, Campbell AC, Pollack BL, Shin J, Sarker A, Brown S, Park HJ, Mehrara BJ, Kataru RP. Regulation of VEGFR3 signaling in lymphatic endothelial cells. Front Cell Dev Biol 2025; 13:1527971. [PMID: 40046235 PMCID: PMC11880633 DOI: 10.3389/fcell.2025.1527971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
The receptor tyrosine kinase vascular endothelial growth factor (VEGF) receptor 3 (VEGFR3) is the principal transmembrane receptor responsible for sensing and coordinating cellular responses to environmental lymphangiogenic stimuli in lymphatic endothelial cells (LECs). VEGFC and D (VEGFC/D) function as the cognate ligands to VEGFR3 by stimulating autophosphorylation of intracellular VEGFR3 tyrosine kinase domains that activate signal cascades involved in lymphatic growth and survival. VEGFR3 primarily promotes downstream signaling through the phosphoinositide 3-kinase (PI3K) and Ras signaling cascades that promote functions including cell proliferation and migration. The importance of VEGFR3 cascades in lymphatic physiology is underscored by identification of dysfunctional VEGFR3 signaling across several lymphatic-related diseases. Recently, our group has shown that intracellular modification of VEGFR3 signaling is a potent means of inducing lymphangiogenesis independent of VEGFC. This is important because long-term treatment with recombinant VEGFC may have deleterious consequences due to off-target effects. A more complete understanding of VEGFR3 signaling pathways may lead to novel drug development strategies. The purpose of this review is to 1) characterize molecular mediators of VEGFC/VEGFR3 downstream signaling activation and their functional roles in LEC physiology and 2) explore molecular regulation of overall VEGFR3 expression and activity within LECs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Raghu P. Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
3
|
Jiang M, Chen D, Xu Z, Liu Y, Yang C, Zhang G, Guo Q, Gao F, He Y, Du Y. Tumor cell-derived hyaluronan fragments induce endocytosis of S1PR1 to promote lymphangiogenesis through LYVE-1-Src pathway. J Cancer 2025; 16:1466-1478. [PMID: 39991586 PMCID: PMC11843240 DOI: 10.7150/jca.104309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/26/2024] [Indexed: 02/25/2025] Open
Abstract
Sphingosine-1-phosphate receptor-1 (S1PR1), a G protein-coupled receptor, has been reported to be involved in lymphangiogenesis. Degradations of extracellular matrix (ECM) are recognized as dynamic modulators in regulating the formation of new lymphatic vessels. However, little research has studied the ECM on S1PR1 in the regulation of lymphatic endothelial cells (LECs) in tumor lymphangiogenesis. Here we attempt to investigate hyaluronan fragments abundant in tumor microenvironment (TME) on S1PR1 in new lymphatic vessel formation. First, we verified that low molecular weight hyaluronan (LMW-HA) derived from tumor cells could promote LECs migration and capillary-like tube formation. Then, we demonstrated that S1PR1 on LECs underwent internalization into the endoplasmic reticulum in response to LMW-HA treatments. Notably, the S1PR1 endocytosis could upregulate lymphangiogenesis. Next, we found that the ablation of lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) could attenuate the S1PR1 endocytosis, implying a novel role of LMW-HA/LYVE-1 in the S1PR1 cycling pathway. Furthermore, we identified that LMW-HA/LYVE-1 interaction could activate Src kinase which in turn upregulates S1PR1 tyrosine phosphorylation, resulting in S1PR1 endocytosis. Collectively, our findings suggested that hyaluronan fragments in TME could induce S1PR1 internalization in LECs, leading to lymphangiogenesis promotion.
Collapse
Affiliation(s)
- Mengying Jiang
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Dandan Chen
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Zhangrun Xu
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Qian Guo
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Feng Gao
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Yan Du
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
4
|
Karakousi T, Cristaldi V, Lopes de Oliveira ML, Medeiros Geraldo LH, González-Robles TJ, da Silva G, Breazeale AP, Encarnacion-Rosado J, Pozniak J, Kimmelman AC, Ruggles KV, Chris Marine J, Chandel NS, Lund AW. IFNγ-dependent metabolic reprogramming restrains an immature, pro-metastatic lymphatic state in melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626426. [PMID: 39677662 PMCID: PMC11642899 DOI: 10.1101/2024.12.02.626426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Lymphatic vessels play a crucial role in activating anti-tumor immune surveillance but also contribute to metastasis and systemic tumor progression. Whether distinct lymphatic phenotypes exist that govern the switch between immunity and metastasis remains unclear. Here we reveal that cytotoxic immunity normalizes lymphatic function and uncouples immune and metastatic potential. We find that in mice and humans, intratumoral lymphatic vessel density negatively correlates with productive cytotoxic immune responses and identify IFNγ as an intrinsic inhibitor of lymphangiogenesis. Specific deletion of the Ifngr1 in lymphatic endothelial cells (LECs) greatly expanded the intratumoral lymphatic network and drove the emergence of a tip-like endothelial state, promoting lymph node metastasis but not dendritic cell migration. IFNγ inhibits oxidative phosphorylation, which is required for proliferation and acquisition of the pathologic transcriptional state. Our data indicate that IFNγ induces a phenotypic switch in tumor-associated lymphatic vessels to reinforce canonical immune surveillance and block metastasis.
Collapse
Affiliation(s)
- Triantafyllia Karakousi
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vanessa Cristaldi
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Luiz Henrique Medeiros Geraldo
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY
| | - Tania J. González-Robles
- Institute of Systems Genetics, Department of Precision Medicine, NYU Grossman School of Medicine, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY USA
| | - Gabrielle da Silva
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Alec P. Breazeale
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Joel Encarnacion-Rosado
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, NY, USA
| | - Joanna Pozniak
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alec C. Kimmelman
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Kelly V. Ruggles
- Institute of Systems Genetics, Department of Precision Medicine, NYU Grossman School of Medicine, New York, NY USA
| | - J. Chris Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Amanda W. Lund
- Ronald O Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Cell, Developmental, & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| |
Collapse
|
5
|
Del Gaudio I, Nitzsche A, Boyé K, Bonnin P, Poulet M, Nguyen TQ, Couty L, Ha HTT, Nguyen DT, Cazenave-Gassiot A, Ben Alaya K, Thérond P, Chun J, Wenk MR, Proia RL, Henrion D, Nguyen LN, Eichmann A, Camerer E. Zonation and ligand and dose dependence of sphingosine 1-phosphate receptor-1 signalling in blood and lymphatic vasculature. Cardiovasc Res 2024; 120:1794-1810. [PMID: 39086170 PMCID: PMC11587562 DOI: 10.1093/cvr/cvae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 08/02/2024] Open
Abstract
AIMS Circulating levels of sphingosine 1-phosphate (S1P), an HDL-associated ligand for the endothelial cell (EC) protective S1P receptor-1 (S1PR1), are reduced in disease states associated with endothelial dysfunction. Yet, as S1PR1 has high affinity for S1P and can be activated by ligand-independent mechanisms and EC autonomous S1P production, it is unclear if relative reductions in circulating S1P can cause endothelial dysfunction. It is also unclear how EC S1PR1 insufficiency, whether induced by deficiency in circulating ligand or by S1PR1-directed immunosuppressive therapy, affects different vascular subsets. METHODS AND RESULTS We here fine map the zonation of S1PR1 signalling in the murine blood and lymphatic vasculature, superimpose cell-type-specific and relative deficiencies in S1P production to define ligand source and dose dependence, and correlate receptor engagement to essential functions. In naïve blood vessels, despite broad expression, EC S1PR1 engagement was restricted to resistance-size arteries, lung capillaries, and a subset of high-endothelial venules (HEVs). Similar zonation was observed for albumin extravasation in EC S1PR1-deficient mice, and brain extravasation was reproduced with arterial EC-selective S1pr1 deletion. In lymphatic ECs, S1PR1 engagement was high in collecting vessels and lymph nodes and low in blind-ended capillaries that drain tissue fluids. While EC S1P production sustained S1PR1 signalling in lymphatics and HEV, haematopoietic cells provided ∼90% of plasma S1P and sustained signalling in resistance arteries and lung capillaries. S1PR1 signalling and endothelial function were both surprisingly sensitive to reductions in plasma S1P with apparent saturation around 50% of normal levels. S1PR1 engagement did not depend on sex or age but modestly increased in arteries in hypertension and diabetes. Sphingosine kinase (Sphk)-2 deficiency also increased S1PR1 engagement selectively in arteries, which could be attributed to Sphk1-dependent S1P release from perivascular macrophages. CONCLUSION This study highlights vessel subtype-specific S1PR1 functions and mechanisms of engagement and supports the relevance of S1P as circulating biomarker for endothelial function.
Collapse
Affiliation(s)
- Ilaria Del Gaudio
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Anja Nitzsche
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Kevin Boyé
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Philippe Bonnin
- Physiologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Paris, France
- Université Paris Cité, INSERM U1144, UFR de Pharmacie, Paris, France
| | - Mathilde Poulet
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Toan Q Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Ludovic Couty
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Hoa T T Ha
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Dat T Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Amaury Cazenave-Gassiot
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Khaoula Ben Alaya
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| | - Patrice Thérond
- Service de Biochimie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital de Bicêtre, Le Kremlin Bicêtre, France
- UFR de Pharmacie, EA 4529, Châtenay-Malabry, France
| | - Jerold Chun
- Neuroscience Drug Discovery, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Markus R Wenk
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Richard L Proia
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Institutes of Health, Bethesda, MD, USA
| | - Daniel Henrion
- MitoVasc Department, Angers University, Team 2 (CarMe), Angers University Hospital (CHU of Angers), CNRS, INSERM U1083, Angers, France
| | - Long N Nguyen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Anne Eichmann
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
- Department of Internal Medicine and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, USA
| | - Eric Camerer
- Université Paris Cité, Paris Cardiovascular Research Centre, INSERM U970, 56 Rue Leblanc, F-75015 Paris, France
| |
Collapse
|
6
|
Cao R, Sun R, Ye Y, Tian P, Huang B, Ye H, Dai L, Lan Z, Liu J, Li L. Low shear stress-induced blockage of autophagic flux impairs endothelial barrier and facilitates atherosclerosis in mice. Exp Cell Res 2024; 439:114071. [PMID: 38729336 DOI: 10.1016/j.yexcr.2024.114071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
Atherosclerosis preferentially occurs in areas with low shear stress (LSS) and oscillatory flow. LSS has been demonstrated to correlate with the development of atherosclerosis. The sphingosine 1-phosphate receptor 1 (S1PR1), involving intravascular blood flow sensing, regulates vascular development and vascular barrier function. However, whether LSS affects atherosclerosis via regulating S1PR1 remains incompletely clear. In this study, immunostaining results of F-actin, β-catenin, and VE-cadherin indicated that LSS impaired endothelial barrier function in human umbilical vein endothelial cells (HUVECs). Western blot analysis showed that LSS resulted in blockage of autophagic flux in HUVECs. In addition, autophagy agonist Rapamycin (Rapa) antagonized LSS-induced endothelial barrier dysfunction, whereas autophagic flux inhibitor Bafilomycin A1 (BafA1) exacerbated it, indicating that LSS promoted endothelial barrier dysfunction by triggering autophagic flux blockage. Notably, gene expression analysis revealed that LSS downregulated S1PR1 expression, which was antagonized by Rapa. Selective S1PR1 antagonist W146 impaired endothelial barrier function of HUVECs under high shear stress (HSS) conditions. Moreover, our data showed that expression of GAPARAPL2, a member of autophagy-related gene 8 (Atg8) proteins, was decreased in HUVECs under LSS conditions. Autophagic flux blockage induced by GAPARAPL2 knockdown inhibited S1PR1, aggravated endothelial barrier dysfunction of HUVECs in vitro, and promoted aortic atherosclerosis in ApoE-/- mice in vivo. Our study demonstrates that autophagic flux blockage induced by LSS downregulates S1PR1 expression and impairs endothelial barrier function. GABARAPL2 inhibition is involved in LSS-induced autophagic flux blockage, which impairs endothelial barrier function via downregulation of S1PR1.
Collapse
Affiliation(s)
- Ruhao Cao
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Ruxian Sun
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Pingge Tian
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Bin Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Haowen Ye
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Libing Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Jia Liu
- VIP Medical Service Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China.
| |
Collapse
|
7
|
Carlantoni C, Liekfeld LMH, Hemkemeyer SA, Schreier D, Saygi C, Kurelic R, Cardarelli S, Kalucka J, Schulte C, Beerens M, Mailer RK, Schäffer TE, Naro F, Pellegrini M, Nikolaev VO, Renné T, Frye M. The phosphodiesterase 2A controls lymphatic junctional maturation via cGMP-dependent notch signaling. Dev Cell 2024; 59:308-325.e11. [PMID: 38159569 DOI: 10.1016/j.devcel.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
The molecular mechanisms by which lymphatic vessels induce cell contact inhibition are not understood. Here, we identify the cGMP-dependent phosphodiesterase 2A (PDE2A) as a selective regulator of lymphatic but not of blood endothelial contact inhibition. Conditional deletion of Pde2a in mouse embryos reveals severe lymphatic dysplasia, whereas blood vessel architecture remains unaltered. In the absence of PDE2A, human lymphatic endothelial cells fail to induce mature junctions and cell cycle arrest, whereas cGMP levels, but not cAMP levels, are increased. Loss of PDE2A-mediated cGMP hydrolysis leads to the activation of p38 signaling and downregulation of NOTCH signaling. However, DLL4-induced NOTCH activation restores junctional maturation and contact inhibition in PDE2A-deficient human lymphatic endothelial cells. In postnatal mouse mesenteries, PDE2A is specifically enriched in collecting lymphatic valves, and loss of Pde2a results in the formation of abnormal valves. Our data demonstrate that PDE2A selectively finetunes a crosstalk of cGMP, p38, and NOTCH signaling during lymphatic vessel maturation.
Collapse
Affiliation(s)
- Claudia Carlantoni
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Leon M H Liekfeld
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Sandra A Hemkemeyer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Danny Schreier
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ceren Saygi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Roberta Kurelic
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Silvia Cardarelli
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Christian Schulte
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany; Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tuebingen, 72076 Tuebingen, Germany
| | - Fabio Naro
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Manuela Pellegrini
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, Campus A. Buzzati Traverso, Monterotondo Scalo, Rome 00015, Italy
| | - Viacheslav O Nikolaev
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany; Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany.
| |
Collapse
|
8
|
Chakraborty A, Kim A, AlAbdullatif S, Campbell JD, Alekseyev YO, Kaplan U, Dambal V, Ligresti G, Trojanowska M. Endothelial Erg Regulates Expression of Pulmonary Lymphatic Junctional and Inflammation Genes in Mouse Lungs Impacting Lymphatic Transport. RESEARCH SQUARE 2024:rs.3.rs-3808970. [PMID: 38343832 PMCID: PMC10854286 DOI: 10.21203/rs.3.rs-3808970/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The ETS transcription factor ERG is a master regulator of endothelial gene specificity and highly enriched in the capillary, vein, and arterial endothelial cells. ERG expression is critical for endothelial barrier function, permeability, and vascular inflammation. A dysfunctional vascular endothelial ERG has been shown to impair lung capillary homeostasis, contributing to pulmonary fibrosis as previously observed in IPF lungs. Our preliminary observations indicate that lymphatic endothelial cells (LEC) in the human IPF lung also lack ERG. To understand the role of ERG in pulmonary LECs, we developed LEC-specific inducible Erg-CKO and Erg-GFP-CKO conditional knockout (CKO) mice under Prox1 promoter. Whole lung microarray analysis, flow cytometry, and qPCR confirmed an inflammatory and pro-lymphvasculogenic predisposition in Erg-CKO lung. FITC-Dextran tracing analysis showed an increased pulmonary interstitial lymphatic fluid transport from the lung to the axial lymph node. Single-cell transcriptomics confirmed that genes associated with cell junction integrity were downregulated in Erg-CKO pre-collector and collector LECs. Integrating Single-cell transcriptomics and CellChatDB helped identify LEC specific communication pathways contributing to pulmonary inflammation, trans-endothelial migration, inflammation, and Endo-MT in Erg-CKO lung. Our findings suggest that downregulation of lymphatic Erg crucially affects LEC function, LEC permeability, pulmonary LEC communication pathways and lymphatic transcriptomics.
Collapse
Affiliation(s)
- Adri Chakraborty
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alex Kim
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Salam AlAbdullatif
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joshua D Campbell
- Division of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yuriy O Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ulas Kaplan
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Vrinda Dambal
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Giovanni Ligresti
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maria Trojanowska
- Arthritis & Autoimmune Diseases Research Centre, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Yu B, Shen K, Li T, Li J, Meng M, Liu W, Tang Q, Zhu T, Wang X, Leung SWS, Shi Y. Glycolytic enzyme PFKFB3 regulates sphingosine 1-phosphate receptor 1 in proangiogenic glomerular endothelial cells under diabetic condition. Am J Physiol Cell Physiol 2023; 325:C1354-C1368. [PMID: 37781737 PMCID: PMC10861147 DOI: 10.1152/ajpcell.00261.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Glomerular angiogenesis is a characteristic feature of diabetic nephropathy (DN). Enhanced glycolysis plays a crucial role in angiogenesis. The present study was designed to investigate the role of glycolysis in glomerular endothelial cells (GECs) in a mouse model of DN. Mouse renal cortex and isolated glomerular cells were collected for single-cell and RNA sequencing. Cultured GECs were exposed to high glucose in the presence (proangiogenic) and absence of a vascular sprouting regimen. MicroRNA-590-3p was delivered by lipofectamine in vivo and in vitro. In the present study, a subgroup of GECs with proangiogenic features was identified in diabetic kidneys by using sequencing analyses. In cultured proangiogenic GECs, high glucose increased glycolysis and phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) protein expression, which were inhibited by overexpressing miRNA-590-3p. Mimics of miRNA-590-3p also increased receptor for sphingosine 1-phosphate (S1pR1) expression, an angiogenesis regulator, in proangiogenic GECs challenged with high glucose. Inhibition of PFKFB3 by pharmacological and genetic approaches upregulated S1pR1 protein in vitro. Mimics of miRNA-590-3p significantly reduced migration and angiogenic potential in proangiogenic GECs challenged with high glucose. Ten-week-old type 2 diabetic mice had elevated urinary albumin levels, reduced renal cortex miRNA-590-3p expression, and disarrangement of glomerular endothelial cell fenestration. Overexpressing miRNA-590-3p via perirenal adipose tissue injection restored endothelial cell fenestration and reduced urinary albumin levels in diabetic mice. Therefore, the present study identifies a subgroup of GECs with proangiogenic features in mice with DN. Local administration of miRNA-590-3p mimics reduces glycolytic rate and upregulates S1pR1 protein expression in proangiogenic GECs. The protective effects of miRNA-590-3p provide therapeutic potential in DN treatment.NEW & NOTEWORTHY Proangiogenetic glomerular endothelial cells (GECs) are activated in diabetic nephropathy. High glucose upregulates glycolytic enzyme phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) in proangiogenetic cells. PFKFB3 protects the glomerular filtration barrier by targeting endothelial S1pR1. MiRNA-590-3p restores endothelial cell function and mitigates diabetic nephropathy.
Collapse
Affiliation(s)
- Baixue Yu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Kaiyuan Shen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tingting Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiawei Li
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Mei Meng
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Wenjie Liu
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qunye Tang
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tongyu Zhu
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Susan W S Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Aithabathula RV, Pervaiz N, Kathuria I, Swanson M, Singh UP, Kumar S, Park F, Singla B. Hydrogen sulfide donor activates AKT-eNOS signaling and promotes lymphatic vessel formation. PLoS One 2023; 18:e0292663. [PMID: 37883422 PMCID: PMC10602273 DOI: 10.1371/journal.pone.0292663] [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] [Received: 07/10/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
The lymphatic network is pivotal for various physiological functions in the human body. Accumulated evidence supports the role of therapeutic lymphangiogenesis in the treatment of several pathologies. Endogenous gasotransmitter, hydrogen sulfide (H2S) has been extensively studied for its potential as a pro-angiogenic factor and vascular function modulator. However, the role of H2S in governing lymphatic vessel formation, and underlying molecular mechanisms are understudied. The present study was designed to investigate the effects of H2S donor sodium hydrogen sulfide (NaHS) on lymphatic vascularization and pro-angiogenic signaling pathways using both in vitro and in vivo approaches. In vitro dose-response experiments showed increased proliferation and tube formation by NaHS-treated human lymphatic endothelial cells (LECs) compared with control cells. Immunoblotting performed with LEC lysates prepared after time-course NaHS treatment demonstrated increased activation of ERK1/2, AKT and eNOS after 20 min of NaHS stimulation. Further, NaHS treatment induced nitric oxide production, reduced reactive oxygen species generation, and promoted cell cycle in LECs. Additional cell cycle analysis showed that NaHS treatment abrogates oxidized LDL-induced cell cycle arrest in LECs. The results of in vivo Matrigel plug assay revealed increased lymphatic vessel density in Matrigel plugs containing NaHS compared with control plugs, however, no significant differences in angiogenesis and immune cell infiltration were observed. Collectively, these findings suggest that H2S donor NaHS promotes lymphatic vessel formation both in vitro and in vivo and may be utilized to promote reparative lymphangiogenesis to alleviate lymphatic dysfunction-related disorders.
Collapse
Affiliation(s)
- Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Mallory Swanson
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| |
Collapse
|
11
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang JL, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal Lymphatic Sphingosine-1-Phosphate Signaling Aggravates Lymphatic Dysfunction and Tissue Inflammation. Circulation 2023; 148:1231-1249. [PMID: 37609838 PMCID: PMC10592179 DOI: 10.1161/circulationaha.123.064181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T-cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T-cell activation. Characterizing this biology is relevant for developing much needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1-deficient (S1pr1LECKO) mice were generated. Disease progression was quantified by tail-volumetric and -histopathologic measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then cocultured with CD4 T cells, followed by an analysis of CD4 T-cell activation and pathway signaling. Last, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T-cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1P receptor 1 (S1PR1). LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T-cell infiltration in mouse lymphedema. LECs, isolated from S1pr1LECKO mice and cocultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs promoted T-helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. Human dermal LECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro, P-selectin blockade reduced the activation and differentiation of Th cells cocultured with shS1PR1-treated human dermal LECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSIONS This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T-cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Lon Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
12
|
Bertoldi G, Caputo I, Calò L, Rossitto G. Lymphatic vessels and the renin-angiotensin-system. Am J Physiol Heart Circ Physiol 2023; 325:H837-H855. [PMID: 37565265 DOI: 10.1152/ajpheart.00023.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
The lymphatic system is an integral part of the circulatory system and plays an important role in the fluid homeostasis of the human body. Accumulating evidence has recently suggested the involvement of lymphatic dysfunction in the pathogenesis of cardio-reno-vascular (CRV) disease. However, how the sophisticated contractile machinery of lymphatic vessels is modulated and, possibly impaired in CRV disease, remains largely unknown. In particular, little attention has been paid to the effect of the renin-angiotensin-system (RAS) on lymphatics, despite the high concentration of RAS mediators that these tissue-draining vessels are exposed to and the established role of the RAS in the development of classic microvascular dysfunction and overt CRV disease. We herein review recent studies linking RAS to lymphatic function and/or plasticity and further highlight RAS-specific signaling pathways, previously shown to drive adverse arterial remodeling and CRV organ damage that have potential for direct modulation of the lymphatic system.
Collapse
Affiliation(s)
- Giovanni Bertoldi
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
- Nephrology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Ilaria Caputo
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Lorenzo Calò
- Nephrology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Giacomo Rossitto
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
13
|
Fernandes LM, Tresemer J, Zhang J, Rios JJ, Scallan JP, Dellinger MT. Hyperactive KRAS/MAPK signaling disrupts normal lymphatic vessel architecture and function. Front Cell Dev Biol 2023; 11:1276333. [PMID: 37842094 PMCID: PMC10571159 DOI: 10.3389/fcell.2023.1276333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Complex lymphatic anomalies (CLAs) are sporadically occurring diseases caused by the maldevelopment of lymphatic vessels. We and others recently reported that somatic activating mutations in KRAS can cause CLAs. However, the mechanisms by which activating KRAS mutations cause CLAs are poorly understood. Here, we show that KRASG12D expression in lymphatic endothelial cells (LECs) during embryonic development impairs the formation of lymphovenous valves and causes the enlargement of lymphatic vessels. We demonstrate that KRASG12D expression in primary human LECs induces cell spindling, proliferation, and migration. It also increases AKT and ERK1/2 phosphorylation and decreases the expression of genes that regulate the maturation of lymphatic vessels. We show that MEK1/2 inhibition with the FDA-approved drug trametinib suppresses KRASG12D-induced morphological changes, proliferation, and migration. Trametinib also decreases ERK1/2 phosphorylation and increases the expression of genes that regulate the maturation of lymphatic vessels. We also show that trametinib and Cre-mediated expression of a dominant-negative form of MEK1 (Map2k1 K97M) suppresses KRASG12D-induced lymphatic vessel hyperplasia in embryos. Last, we demonstrate that conditional knockout of wild-type Kras in LECs does not affect the formation or function of lymphatic vessels. Together, our data indicate that KRAS/MAPK signaling must be tightly regulated during embryonic development for the proper development of lymphatic vessels and further support the testing of MEK1/2 inhibitors for treating CLAs.
Collapse
Affiliation(s)
- Lorenzo M. Fernandes
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Jeffrey Tresemer
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, United States
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, United States
| | - Jonathan J. Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, United States
- McDermott Center for Human Growth and Development, Dallas, TX, United States
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Michael T. Dellinger
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
14
|
Pal S, Bhowmick S, Sharma A, Sierra-Fonseca JA, Mondal S, Afolabi F, Roy D. Lymphatic vasculature in ovarian cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188950. [PMID: 37419192 PMCID: PMC10754213 DOI: 10.1016/j.bbcan.2023.188950] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Ovarian cancer (OVCA) is the second most common gynecological cancer and one of the leading causes of cancer related mortality among women. Recent studies suggest that among ovarian cancer patients at least 70% of the cases experience the involvement of lymph nodes and metastases through lymphatic vascular network. However, the impact of lymphatic system in the growth, spread and the evolution of ovarian cancer, its contribution towards the landscape of ovarian tissue resident immune cells and their metabolic responses is still a major knowledge gap. In this review first we present the epidemiological aspect of the OVCA, the lymphatic architecture of the ovary, we discuss the role of lymphatic circulation in regulation of ovarian tumor microenvironment, metabolic basis of the upregulation of lymphangiogenesis which is often observed during progression of ovarian metastasis and ascites development. Further we describe the implication of several mediators which influence both lymphatic vasculature as well as ovarian tumor microenvironment and conclude with several therapeutic strategies for targeting lymphatic vasculature in ovarian cancer progression in present day.
Collapse
Affiliation(s)
- Sarit Pal
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77843, United States
| | - Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Anurag Sharma
- Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, United States
| | | | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Favour Afolabi
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, United States
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, United States.
| |
Collapse
|
15
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang J, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal lymphatic S1P signaling aggravates lymphatic dysfunction and tissue inflammation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.08.23291175. [PMID: 37398237 PMCID: PMC10312855 DOI: 10.1101/2023.06.08.23291175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T cell activation. Characterizing this biology is relevant for developing much-needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1 -deficient ( S1pr1 LECKO ) mice were generated. Disease progression was quantified by tail-volumetric and -histopathological measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then co-cultured with CD4 T cells, followed by an analysis of CD4 T cell activation and pathway signaling. Finally, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1PR1. LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T cell infiltration in mouse lymphedema. LECs, isolated from S1pr1 LECKO mice and co-cultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs (HDLECs) promoted T helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. HDLECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro , P-selectin blockade reduced the activation and differentiation of Th cells co-cultured with sh S1PR1 -treated HDLECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSION This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition. Clinical Perspective What is New?: Lymphatic-specific S1pr1 deletion exacerbates lymphatic vessel malfunction and Th1/Th2 immune responses during lymphedema pathogenesis. S1pr1 -deficient LECs directly induce Th1/Th2 cell differentiation and decrease anti-inflammatory Treg populations. Peripheral dermal LECs affect CD4 T cell immune responses through direct cell contact.LEC P-selectin, regulated by S1PR1 signaling, affects CD4 T cell activation and differentiation.P-selectin blockade improves lymphedema tail swelling and decreases Th1/Th2 population in the diseased skin.What Are the Clinical Implications?: S1P/S1PR1 signaling in LECs regulates inflammation in lymphedema tissue.S1PR1 expression levels on LECs may be a useful biomarker for assessing predisposition to lymphatic disease, such as at-risk women undergoing mastectomyP-selectin Inhibitors may be effective for certain forms of lymphedema.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
16
|
Xiong W, Chen S, Xiang H, Zhao S, Xiao J, Li J, Liu Y, Shu Z, Ouyang J, Zhang J, Liu H, Wang X, Zou H, Chen Y, Chen A, Lu H. S1PR1 attenuates pulmonary fibrosis by inhibiting EndMT and improving endothelial barrier function. Pulm Pharmacol Ther 2023:102228. [PMID: 37295666 DOI: 10.1016/j.pupt.2023.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fatal disease of unknown etiology. Its pathological manifestations include excessive proliferation and activation of fibroblasts and deposition of extracellular matrix. Endothelial cell-mesenchymal transformation (EndMT), a novel mechanism that generates fibroblast during IPF, is responsible for fibroblast-like phenotypic changes and activation of fibroblasts into hypersecretory cells. However, the exact mechanism behind EndMT-derived fibroblasts and activation is uncertain. Here, we investigated the role of sphingosine 1-phosphate receptor 1 (S1PR1) in EndMT-driven pulmonary fibrosis. METHODS We treated C57BL/6 mice with bleomycin (BLM) in vivo and pulmonary microvascular endothelial cells with TGF-β1 in vitro. Western blot,flow cytometry, and immunofluorescence were used to detect the expression of S1PR1 in endothelial cells. To evaluate the effect of S1PR1 on EndMT and endothelial barrier and its role in lung fibrosis and related signaling pathways, S1PR1 agonist and antagonist were used in vitro and in vivo. RESULTS Endothelial S1PR1 protein expression was downregulated in both in vitro and in vivo models of pulmonary fibrosis induced by TGF-β1 and BLM, respectively. Downregulation of S1PR1 resulted in EndMT, indicated by decreased expression of endothelial markers CD31 and VE-cadherin, increased expression of mesenchymal markers α-SMA and nuclear transcription factor Snail, and disruption of the endothelial barrier. Further mechanistic studies found that stimulation of S1PR1 inhibited TGF-β1-mediated activation of the Smad2/3 and RhoA/ROCK1 pathways. Moreover, stimulation of S1PR1 attenuated Smad2/3 and RhoA/ROCK1 pathway-mediated damage to endothelial barrier function. CONCLUSIONS Endothelial S1PR1 provides protection against pulmonary fibrosis by inhibiting EndMT and attenuating endothelial barrier damage. Accordingly, S1PR1 may be a potential therapeutic target in progressive IPF.
Collapse
Affiliation(s)
- Wenfang Xiong
- Health Management Center, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China; Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Hong Xiang
- Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Shaoli Zhao
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jie Xiao
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jialing Li
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Yulan Liu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Zhihao Shu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jie Ouyang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jing Zhang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Huiqin Liu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Xuewen Wang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Hang Zou
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Ying Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Alex Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Hongwei Lu
- Health Management Center, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China; Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China.
| |
Collapse
|
17
|
Weigel C, Bellaci J, Spiegel S. Sphingosine-1-phosphate and its receptors in vascular endothelial and lymphatic barrier function. J Biol Chem 2023; 299:104775. [PMID: 37142226 PMCID: PMC10220486 DOI: 10.1016/j.jbc.2023.104775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jacqueline Bellaci
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA.
| |
Collapse
|
18
|
Peluzzo AM, Bkhache M, Do LNH, Autieri MV, Liu X. Differential regulation of lymphatic junctional morphology and the potential effects on cardiovascular diseases. Front Physiol 2023; 14:1198052. [PMID: 37187962 PMCID: PMC10175597 DOI: 10.3389/fphys.2023.1198052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
The lymphatic vasculature provides an essential route to drain fluid, macromolecules, and immune cells from the interstitium as lymph, returning it to the bloodstream where the thoracic duct meets the subclavian vein. To ensure functional lymphatic drainage, the lymphatic system contains a complex network of vessels which has differential regulation of unique cell-cell junctions. The lymphatic endothelial cells lining initial lymphatic vessels form permeable "button-like" junctions which allow substances to enter the vessel. Collecting lymphatic vessels form less permeable "zipper-like" junctions which retain lymph within the vessel and prevent leakage. Therefore, sections of the lymphatic bed are differentially permeable, regulated in part by its junctional morphology. In this review, we will discuss our current understanding of regulating lymphatic junctional morphology, highlighting how it relates to lymphatic permeability during development and disease. We will also discuss the effect of alterations in lymphatic permeability on efficient lymphatic flux in health and how it may affect cardiovascular diseases, with a focus on atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Xiaolei Liu
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
19
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
20
|
Jablon KL, Akerstrom VL, Li M, Braun SE, Norton CE, Castorena-Gonzalez JA. Isolation and short-term culturing of primary lymphatic endothelial cells from collecting lymphatics: A techniques study. Microcirculation 2023; 30:e12778. [PMID: 35879879 PMCID: PMC9873843 DOI: 10.1111/micc.12778] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To develop an experimental method for routine isolation and short-term culture of primary lymphatic endothelial cells from specific collecting vessels. METHODS Lymphatic endothelial cell tubes (LECTs) were isolated from micro-dissected collecting vessels. LECTs were allowed to attach and grow for ~3 weeks before being passaged. Non-purified cultures were partially characterized by immunofluorescence and RT-PCR at passages 1-2. RESULTS The method was validated in cultures of primary lymphatic endothelial cells (LECs) from male and female mice. After 1 or 2 passages, >60% of the LECs maintained expression of Prox1. Expression of 22 different genes was assessed using RT-PCR. Prox1, Vegfr3, eNos, Cdh5, Pecam1, Cx43, Cx37, and Cx47, among others, were expressed in these short-term cultured LECs, while Myh11, Cnn1, Desmin, and Cd11b were not detected. Prox1 expression, as determined by western blotting, was similar in cultured LECs from age-matched male and female mice. Confocal imaging of intracellular calcium in cultures of primary LECs from Cdh5-GCaMP8 mice demonstrated that a functional phenotype was maintained, similar to lymphatic endothelial cells in freshly isolated vessels. CONCLUSIONS This method provides an innovative tool for routine isolation and study of primary LECs from specific collecting lymphatic vessels from any mouse, and in fact, from other species.
Collapse
Affiliation(s)
- Kelli L. Jablon
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Victoria L. Akerstrom
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Min Li
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Stephen E. Braun
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Division of Immunology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Charles E. Norton
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| | | |
Collapse
|
21
|
Patnam M, Dommaraju SR, Masood F, Herbst P, Chang JH, Hu WY, Rosenblatt MI, Azar DT. Lymphangiogenesis Guidance Mechanisms and Therapeutic Implications in Pathological States of the Cornea. Cells 2023; 12:319. [PMID: 36672254 PMCID: PMC9856498 DOI: 10.3390/cells12020319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/22/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Corneal lymphangiogenesis is one component of the neovascularization observed in several inflammatory pathologies of the cornea including dry eye disease and corneal graft rejection. Following injury, corneal (lymph)angiogenic privilege is impaired, allowing ingrowth of blood and lymphatic vessels into the previously avascular cornea. While the mechanisms underlying pathological corneal hemangiogenesis have been well described, knowledge of the lymphangiogenesis guidance mechanisms in the cornea is relatively scarce. Various signaling pathways are involved in lymphangiogenesis guidance in general, each influencing one or multiple stages of lymphatic vessel development. Most endogenous factors that guide corneal lymphatic vessel growth or regression act via the vascular endothelial growth factor C signaling pathway, a central regulator of lymphangiogenesis. Several exogenous factors have recently been repurposed and shown to regulate corneal lymphangiogenesis, uncovering unique signaling pathways not previously known to influence lymphatic vessel guidance. A strong understanding of the relevant lymphangiogenesis guidance mechanisms can facilitate the development of targeted anti-lymphangiogenic therapeutics for corneal pathologies. In this review, we examine the current knowledge of lymphatic guidance cues, their regulation of inflammatory states in the cornea, and recently discovered anti-lymphangiogenic therapeutic modalities.
Collapse
Affiliation(s)
- Mehul Patnam
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil R. Dommaraju
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Faisal Masood
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Paula Herbst
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Dimitri T. Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
22
|
Levesque MV, Hla T. Signal Transduction and Gene Regulation in the Endothelium. Cold Spring Harb Perspect Med 2023; 13:a041153. [PMID: 35667710 PMCID: PMC9722983 DOI: 10.1101/cshperspect.a041153] [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: 01/07/2023]
Abstract
Extracellular signals act on G-protein-coupled receptors (GPCRs) to regulate homeostasis and adapt to stress. This involves rapid intracellular post-translational responses and long-lasting gene-expression changes that ultimately determine cellular phenotype and fate changes. The lipid mediator sphingosine 1-phosphate (S1P) and its receptors (S1PRs) are examples of well-studied GPCR signaling axis essential for vascular development, homeostasis, and diseases. The biochemical cascades involved in rapid S1P signaling are well understood. However, gene-expression regulation by S1PRs are less understood. In this review, we focus our attention to how S1PRs regulate nuclear chromatin changes and gene transcription to modulate vascular and lymphatic endothelial phenotypic changes during embryonic development and adult homeostasis. Because S1PR-targeted drugs approved for use in the treatment of autoimmune diseases cause substantial vascular-related adverse events, these findings are critical not only for general understanding of stimulus-evoked gene regulation in the vascular endothelium, but also for therapeutic development of drugs for autoimmune and perhaps vascular diseases.
Collapse
Affiliation(s)
- Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
23
|
Abstract
Button-like junctions are discontinuous contacts at the border of oak-leaf-shaped endothelial cells of initial lymphatic vessels. These junctions are distinctively different from continuous zipper-like junctions that create the endothelial barrier in collecting lymphatics and blood vessels. Button junctions are point contacts, spaced about 3 µm apart, that border valve-like openings where fluid and immune cells enter lymphatics. In intestinal villi, openings between button junctions in lacteals also serve as entry routes for chylomicrons. Like zipper junctions that join endothelial cells, buttons consist of adherens junction proteins (VE-cadherin) and tight junction proteins (claudin-5, occludin, and others). Buttons in lymphatics form from zipper junctions during embryonic development, can convert into zippers in disease or after experimental genetic or pharmacological manipulation, and can revert back to buttons with treatment. Multiple signaling pathways and local microenvironmental factors have been found to contribute to button junction plasticity and could serve as therapeutic targets in pathological conditions ranging from pulmonary edema to obesity.
Collapse
Affiliation(s)
- Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143-0452, USA
| | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143-0452, USA
| |
Collapse
|
24
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
25
|
Li Q, Zhou C, Zhao K, Duan Y, Yue J, Liu X, Wu J, Deng S. Lymphatic endothelial sphingosine 1-phosphate receptor 1 enhances macrophage clearance via lymphatic system following myocardial infarction. Front Cardiovasc Med 2022; 9:872102. [PMID: 36003911 PMCID: PMC9393290 DOI: 10.3389/fcvm.2022.872102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Lymphatic endothelial cell homeostasis plays important roles in normal physiological cardiac functions, and its dysfunction significantly influences pathological cardiac remodeling after myocardial infarction (MI). Our results revealed that sphingosine 1-phosphate receptor 1 (S1pr1) expression in cardiac lymphatic endothelial cells (LECs) was sharply changed after MI. It has been shown that S1pr1 tightly controlled LEC functions and homeostasis. We thus hypothesized that lymphatic endothelial S1pr1 might be involved in post-MI cardiac remodeling. We generated LEC-conditional S1pr1 transgenic mice, in which S1pr1 expression was reduced in cardiac LECs. We performed the left anterior descending coronary artery (LAD) ligation operation to induce MI in these mice. Cardiac functions and remodeling were examined by echocardiography analysis and serial histological analysis. Meanwhile, we performed adoptive cell transfer experiments to monitor macrophage trafficking in post-MI myocardium and their draining lymphatic system. Furthermore, in vitro cell culture experiments and mechanism studies were undertaken to uncover the molecular mechanism by which LEC-S1pr1 regulated cardiac inflammation and remodeling after MI. Our results showed that S1pr1 expression significantly decreased in cardiac LECs after MI. Our in vivo experiments showed that the reduced expression of LEC-S1pr1 deteriorated cardiac function and worsened pathological cardiac remodeling after MI. Our further results demonstrated that the reduced expression of LEC-S1pr1 did not influence macrophage infiltration in an early inflammatory phase of MI, but significantly affected macrophages clearance in the later phase of MI via afferent cardiac lymphatics, and thus influenced inflammatory responses and cardiac outcome after MI. Further study showed that S1P/S1pr1 activated ERK signaling pathway and enhanced CCL2 expression, which promoted macrophage trafficking in a paracrine manner. This study reveals that cardiac lymphatic endothelial cells tightly control macrophage trafficking via lymphatic vessels in injured hearts via S1P/S1pr1/ERK/CCL2 pathway and thus regulate post-MI immune modulation and heart repair. This study highlights the importance of cardiac lymphatic vessel system in orchestrating post-MI immune responses and cardiac remodeling by regulating macrophage transit in injured hearts. Our finding implies that a feasible modulation of S1pr1 signaling in LECs might provide a promising target to resolve excessive inflammation and to ameliorate adverse cardiac remodeling after MI.
Collapse
Affiliation(s)
- Qinyu Li
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Ningxia, China
| | - Caixia Zhou
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kang Zhao
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Ningxia, China
| | - Yunhao Duan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinnan Yue
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiuxiang Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjin Wu
- Cardiovascular Department, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jinjin Wu,
| | - Shengqiong Deng
- Department of Clinical Laboratory, School of Medicine, Gongli Hospital, Shanghai University, Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Shanghai, China
- Shengqiong Deng,
| |
Collapse
|
26
|
Geng X, Srinivasan RS. Molecular Mechanisms Driving Lymphedema and Other Lymphatic Anomalies. Cold Spring Harb Perspect Med 2022; 12:a041272. [PMID: 35817543 PMCID: PMC9341459 DOI: 10.1101/cshperspect.a041272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lymphatic vasculature regulates fluid homeostasis by absorbing interstitial fluid and returning it to blood. Lymphatic vasculature is also critical for lipid absorption and inflammatory response. Lymphatic vasculature is composed of lymphatic capillaries, collecting lymphatic vessels, lymphatic valves, and lymphovenous valves. Defects in any of these structures could lead to lymphatic anomalies such as lymphedema, cystic lymphatic malformation, and Gorham-Stout disease. Basic research has led to a deeper understanding of the stepwise development of the lymphatic vasculature. VEGF-C and shear stress signaling pathways have evolved as critical regulators of lymphatic vascular development. Loss-of-function and gain-of-function mutations in genes that are involved in these signaling pathways are associated with lymphatic anomalies. Importantly, drugs that target these molecules are showing outstanding efficacy in treating certain lymphatic anomalies. In this article, we summarize these exciting developments and highlight the future challenges.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73013, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73013, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, USA
| |
Collapse
|
27
|
Bonetti G, Paolacci S, Samaja M, Maltese PE, Michelini S, Michelini S, Michelini S, Ricci M, Cestari M, Dautaj A, Medori MC, Bertelli M. Low Efficacy of Genetic Tests for the Diagnosis of Primary Lymphedema Prompts Novel Insights into the Underlying Molecular Pathways. Int J Mol Sci 2022; 23:ijms23137414. [PMID: 35806420 PMCID: PMC9267137 DOI: 10.3390/ijms23137414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Lymphedema is a chronic inflammatory disorder caused by ineffective fluid uptake by the lymphatic system, with effects mainly on the lower limbs. Lymphedema is either primary, when caused by genetic mutations, or secondary, when it follows injury, infection, or surgery. In this study, we aim to assess to what extent the current genetic tests detect genetic variants of lymphedema, and to identify the major molecular pathways that underlie this rather unknown disease. We recruited 147 individuals with a clinical diagnosis of primary lymphedema and used established genetic tests on their blood or saliva specimens. Only 11 of these were positive, while other probands were either negative (63) or inconclusive (73). The low efficacy of such tests calls for greater insight into the underlying mechanisms to increase accuracy. For this purpose, we built a molecular pathways diagram based on a literature analysis (OMIM, Kegg, PubMed, Scopus) of candidate and diagnostic genes. The PI3K/AKT and the RAS/MAPK pathways emerged as primary candidates responsible for lymphedema diagnosis, while the Rho/ROCK pathway appeared less critical. The results of this study suggest the most important pathways involved in the pathogenesis of lymphedema, and outline the most promising diagnostic and candidate genes to diagnose this disease.
Collapse
Affiliation(s)
- Gabriele Bonetti
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- Correspondence: ; Tel.: +39-0365-62-061
| | - Stefano Paolacci
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | | | | | - Sandro Michelini
- Vascular Diagnostics and Rehabilitation Service, Marino Hospital, ASL Roma 6, 00047 Marino, Italy;
| | - Serena Michelini
- Unit of Physical Medicine, “Sapienza” University of Rome, 00185 Rome, Italy;
| | | | - Maurizio Ricci
- Division of Rehabilitation Medicine, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy;
| | - Marina Cestari
- Study Centre Pianeta Linfedema, 05100 Terni, Italy;
- Lymphology Sector of the Rehabilitation Service, USLUmbria2, 05100 Terni, Italy
| | - Astrit Dautaj
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Maria Chiara Medori
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
| | - Matteo Bertelli
- MAGI’s LAB, 38068 Rovereto, Italy; (S.P.); (P.E.M.); (A.D.); (M.C.M.); (M.B.)
- MAGI Group, 25010 San Felice del Benaco, Italy;
- MAGI Euregio, 39100 Bolzano, Italy
| |
Collapse
|
28
|
Liu J, Yu C. Lymphangiogenesis and Lymphatic Barrier Dysfunction in Renal Fibrosis. Int J Mol Sci 2022; 23:ijms23136970. [PMID: 35805972 PMCID: PMC9267103 DOI: 10.3390/ijms23136970] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
As an integral part of the vascular system, the lymphatic vasculature is essential for tissue fluid homeostasis, nutritional lipid assimilation and immune regulation. The composition of the lymphatic vasculature includes fluid-absorbing initial lymphatic vessels (LVs), transporting collecting vessels and anti-regurgitation valves. Although, in recent decades, research has drastically enlightened our view of LVs, investigations of initial LVs, also known as lymphatic capillaries, have been stagnant due to technical limitations. In the kidney, the lymphatic vasculature mainly presents in the cortex, keeping the local balance of fluid, solutes and immune cells. The contribution of renal LVs to various forms of pathology, especially chronic kidney diseases, has been addressed in previous studies, however with diverging and inconclusive results. In this review, we discuss the most recent advances in the proliferation and permeability of lymphatic capillaries as well as their influencing factors. Novel technologies to visualize and measure LVs function are described. Then, we highlight the role of the lymphatic network in renal fibrosis and the crosstalk between kidney and other organs, such as gut and heart.
Collapse
|
29
|
Xu W, Nelson-Maney NP, Bálint L, Kwon HB, Davis RB, Dy DCM, Dunleavey JM, St. Croix B, Caron KM. Orphan G-Protein Coupled Receptor GPRC5B Is Critical for Lymphatic Development. Int J Mol Sci 2022; 23:ijms23105712. [PMID: 35628521 PMCID: PMC9146384 DOI: 10.3390/ijms23105712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
Numerous studies have focused on the molecular signaling pathways that govern the development and growth of lymphatics in the hopes of elucidating promising druggable targets. G protein-coupled receptors (GPCRs) are currently the largest family of membrane receptors targeted by FDA-approved drugs, but there remain many unexplored receptors, including orphan GPCRs with no known biological ligand or physiological function. Thus, we sought to illuminate the cadre of GPCRs expressed at high levels in lymphatic endothelial cells and identified four orphan receptors: GPRC5B, AGDRF5/GPR116, FZD8 and GPR61. Compared to blood endothelial cells, GPRC5B is the most abundant GPCR expressed in cultured human lymphatic endothelial cells (LECs), and in situ RNAscope shows high mRNA levels in lymphatics of mice. Using genetic engineering approaches in both zebrafish and mice, we characterized the function of GPRC5B in lymphatic development. Morphant gprc5b zebrafish exhibited failure of thoracic duct formation, and Gprc5b-/- mice suffered from embryonic hydrops fetalis and hemorrhage associated with subcutaneous edema and blood-filled lymphatic vessels. Compared to Gprc5+/+ littermate controls, Gprc5b-/- embryos exhibited attenuated developmental lymphangiogenesis. During the postnatal period, ~30% of Gprc5b-/- mice were growth-restricted or died prior to weaning, with associated attenuation of postnatal cardiac lymphatic growth. In cultured human primary LECs, expression of GPRC5B is required to maintain cell proliferation and viability. Collectively, we identify a novel role for the lymphatic-enriched orphan GPRC5B receptor in lymphangiogenesis of fish, mice and human cells. Elucidating the roles of orphan GPCRs in lymphatics provides new avenues for discovery of druggable targets to treat lymphatic-related conditions such as lymphedema and cancer.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - Nathan P. Nelson-Maney
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - László Bálint
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - Hyouk-Bum Kwon
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - Reema B. Davis
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - Danielle C. M. Dy
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
| | - James M. Dunleavey
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute–Frederick, NIH, Frederick, MD 21702, USA; (J.M.D.); (B.S.C.)
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute–Frederick, NIH, Frederick, MD 21702, USA; (J.M.D.); (B.S.C.)
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA; (W.X.); (N.P.N.-M.); (L.B.); (H.-B.K.); (R.B.D.); (D.C.M.D.)
- Correspondence:
| |
Collapse
|
30
|
Meng Q, Pu L, Qi M, Li S, Sun B, Wang Y, Liu B, Li F. Laminar shear stress inhibits inflammation by activating autophagy in human aortic endothelial cells through HMGB1 nuclear translocation. Commun Biol 2022; 5:425. [PMID: 35523945 PMCID: PMC9076621 DOI: 10.1038/s42003-022-03392-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 04/21/2022] [Indexed: 11/09/2022] Open
Abstract
Prevention and treatment of atherosclerosis (AS) by targeting the inflammatory response in vascular endothelial cells has attracted much attention in recent years. Laminar shear stress (LSS) has well-recognized anti-AS properties, however, the exact molecular mechanism remains unclear. In this study, we found that LSS could inhibit the increased expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), cyclooxygenase-2 (COX-2), and matrix metallopeptidase-9 (MMP-9) caused by TNF-α in an autophagy-dependent pathway in human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs). Whole-transcriptome sequencing analysis revealed that erythropoietin-producing hepatocyte receptor B2 (EPHB2) was a key gene in response to LSS. Moreover, co-immunoprecipitation assay indicated that LSS could enhance the EPHB2-mediated nuclear translocation of high mobility group box-1 (HMGB1), which interacts with Beclin-1 (BECN1) and finally leads to autophagy. Simultaneously, we identified an LSS-sensitive long non-coding RNA (lncRNA), LOC10798635, and constructed an LSS-related LOC107986345/miR-128-3p/EPHB2 regulatory axis. Further research revealed the anti-inflammatory effect of LSS depends on autophagy activation resulting from the nuclear translocation of HMGB1 via the LOC107986345/miR-128-3p/EPHB2 axis. Our study demonstrates that LSS could regulate the expression of EPHB2 in HAECs, and the LOC107986345/miR-128-3p/EPHB2 axis plays a vital role in AS development.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Luya Pu
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Mingran Qi
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Shuai Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Banghao Sun
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Yaru Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, China.
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China. .,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China. .,Key Laboratory for Health Biomedical Materials of Jilin Province, Jilin University, Changchun, China. .,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China. .,The Key Laboratory for Bionics Engineering, Ministry of Education, Jilin University, Changchun, China.
| |
Collapse
|
31
|
Kamel NM, El-Tanbouly DM, Abdallah DM, Sayed HM. PAR1, a therapeutic target for remote lung injury associated with hind limb ischemia/reperfusion: ERK5/KLF2-dependent lung capillary barrier preservation. Chem Biol Interact 2022; 354:109809. [PMID: 35031271 DOI: 10.1016/j.cbi.2022.109809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/28/2021] [Accepted: 01/05/2022] [Indexed: 11/03/2022]
Abstract
Protease-activated receptor 1 (PAR1) is expressed in pneumocytes and endothelial cells of the alveolar barrier. Its activation by thrombin disrupts the barrier integrity dynamics and induces lung injury in in vitro and in vivo paradigms. Nonetheless, the role of PAR1, as a therapeutic target, in hind limb ischemia/reperfusion (I/R)-mediated remote lung injury has been unclear. Therefore, this study aimed to determine the potential benefit of PAR1 blockade using the selective antagonist SCH79797 in distant lung dysfunction following hind limb I/R injury with special emphasis on the extracellular signal-regulated kinase 5 (ERK5)/Krüppel-like factor 2 (KLF2) axis. Rats were subdivided into control, bilateral hind limb I/R, SCH79797, and SCH79797+BIX02189 (ERK5 inhibitor) groups. PAR1 blockade, ERK5-dependently, alleviated alveolar barrier disruption as evidenced by reductions in both pulmonary systemic leakage of surfactant protein-D and lung fluid accumulation with increase in pulmonary claudin 5, vascular endothelial cadherin, and connexin 37 levels. Such improvements are downstream targets of the ERK5/KLF2-mediated sphingosine-1-phosphate receptor 1 (S1PR1) upregulated expression and pS536-nuclear factor-κB (NF-κB) p65 inhibition. SCH79797 effectively impedes the evoked inflammatory response and oxidative burst by suppressing vascular endothelial growth factor, tumor necrosis factor-α, lipid peroxidation, and neutrophil infiltration while boosting the glutathione antioxidant defense. Accordingly, PAR1 could be a therapeutic target, where its blockade mitigated pulmonary-endothelial barrier disruption via mutual S1PR1 enhancement and NF-κB p65 inhibition following ERK5/KLF2 activation.
Collapse
Affiliation(s)
- Nada M Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Faculty of Pharmacy, Kasr El Aini St., Cairo, 11562, Egypt.
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Faculty of Pharmacy, Kasr El Aini St., Cairo, 11562, Egypt.
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Faculty of Pharmacy, Kasr El Aini St., Cairo, 11562, Egypt.
| | - Helmy M Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Faculty of Pharmacy, Kasr El Aini St., Cairo, 11562, Egypt.
| |
Collapse
|
32
|
Angiopoietin-2-induced lymphatic endothelial cell migration drives lymphangiogenesis via the β1 integrin-RhoA-formin axis. Angiogenesis 2022; 25:373-396. [PMID: 35103877 DOI: 10.1007/s10456-022-09831-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 11/08/2021] [Indexed: 11/01/2022]
Abstract
Lymphangiogenesis is an essential physiological process but also a determining factor in vascular-related pathological conditions. Angiopoietin-2 (Ang2) plays an important role in lymphatic vascular development and function and its upregulation has been reported in several vascular-related diseases, including cancer. Given the established role of the small GTPase RhoA on cytoskeleton-dependent endothelial functions, we investigated the relationship between RhoA and Ang2-induced cellular activities. This study shows that Ang2-driven human dermal lymphatic endothelial cell migration depends on RhoA. We demonstrate that Ang2-induced migration is independent of the Tie receptors, but dependent on β1 integrin-mediated RhoA activation with knockdown, pharmacological approaches, and protein sequencing experiments. Although the key proteins downstream of RhoA, Rho kinase (ROCK) and myosin light chain, were activated, blockade of ROCK did not abrogate the Ang2-driven migratory effect. However, formins, an alternative target of RhoA, were identified as key players, and especially FHOD1. The Ang2-RhoA relationship was explored in vivo, where lymphatic endothelial RhoA deficiency blocked Ang2-induced lymphangiogenesis, highlighting RhoA as an important target for anti-lymphangiogenic treatments.
Collapse
|
33
|
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
|
34
|
Geng X, Srinivasan SR. Whole-mount Immunohistochemistry to Visualize Mouse Embryonic Dermal Lymphatic Vasculature. Bio Protoc 2021; 11:e4186. [PMID: 34761060 PMCID: PMC8554805 DOI: 10.21769/bioprotoc.4186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/02/2022] Open
Abstract
Lymphatic vessels are abundant in the skin where they regulate interstitial fluid uptake and immune surveillance. Defects in dermal lymphatic vessels, such as fewer vessels and abnormal lymphatic vessel coverage with mural cells, are frequently associated with lymphedema and other lymphatic disorders. Whole-mount immunohistochemistry allows the visualization of dermal lymphatic vessels and identifies morphogenetic defects. Most dermal lymphatic vessels start growing during embryogenesis from lymph sacs that are located close to the axilla towards the dorsal and ventral midlines. Here, we present an approach that we have developed to permeabilize, immunolabel, clear, and visualize the lymphatic vessels. These simple and inexpensive techniques reproducibly generate images of dermal lymphatic vessels with great clarity.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73013, USA
| | - Sathish R. Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73013, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| |
Collapse
|
35
|
Muley A, Kim Uh M, Salazar-De Simone G, Swaminathan B, James JM, Murtomaki A, Youn SW, McCarron JD, Kitajewski C, Gnarra Buethe M, Riitano G, Mukouyama YS, Kitajewski J, Shawber CJ. Unique functions for Notch4 in murine embryonic lymphangiogenesis. Angiogenesis 2021; 25:205-224. [PMID: 34665379 PMCID: PMC9054879 DOI: 10.1007/s10456-021-09822-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 10/08/2021] [Indexed: 11/08/2022]
Abstract
In mice, embryonic dermal lymphatic development is well understood and used to study gene functions in lymphangiogenesis. Notch signaling is an evolutionarily conserved pathway that modulates cell fate decisions, which has been shown to both inhibit and promote dermal lymphangiogenesis. Here, we demonstrate distinct roles for Notch4 signaling versus canonical Notch signaling in embryonic dermal lymphangiogenesis. Actively growing embryonic dermal lymphatics expressed NOTCH1, NOTCH4, and DLL4 which correlated with Notch activity. In lymphatic endothelial cells (LECs), DLL4 activation of Notch induced a subset of Notch effectors and lymphatic genes, which were distinctly regulated by Notch1 and Notch4 activation. Treatment of LECs with VEGF-A or VEGF-C upregulated Dll4 transcripts and differentially and temporally regulated the expression of Notch1 and Hes/Hey genes. Mice nullizygous for Notch4 had an increase in the closure of the lymphangiogenic fronts which correlated with reduced vessel caliber in the maturing lymphatic plexus at E14.5 and reduced branching at E16.5. Activation of Notch4 suppressed LEC migration in a wounding assay significantly more than Notch1, suggesting a dominant role for Notch4 in regulating LEC migration. Unlike Notch4 nulls, inhibition of canonical Notch signaling by expressing a dominant negative form of MAML1 (DNMAML) in Prox1+ LECs led to increased lymphatic density consistent with an increase in LEC proliferation, described for the loss of LEC Notch1. Moreover, loss of Notch4 did not affect LEC canonical Notch signaling. Thus, we propose that Notch4 signaling and canonical Notch signaling have distinct functions in the coordination of embryonic dermal lymphangiogenesis.
Collapse
Affiliation(s)
- Ajit Muley
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Minji Kim Uh
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Department of Pharmacology, Columbia University Medical Center, New York, NY, 10032, USA
| | | | - Bhairavi Swaminathan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Jennifer M James
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Aino Murtomaki
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu, 8, 00290, Helsinki, Finland.,Translational Cancer Medicine Program, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Seock Won Youn
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Joseph D McCarron
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chris Kitajewski
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Maria Gnarra Buethe
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gloria Riitano
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA.,Departments of Molecular Medicine and Experimental Medicine, Sapienza University, 00185, Rome, Italy
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Carrie J Shawber
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA. .,Department of Surgery, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
36
|
Cui M, Göbel V, Zhang H. Uncovering the 'sphinx' of sphingosine 1-phosphate signalling: from cellular events to organ morphogenesis. Biol Rev Camb Philos Soc 2021; 97:251-272. [PMID: 34585505 PMCID: PMC9292677 DOI: 10.1111/brv.12798] [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: 12/10/2020] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 11/02/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite, functioning as a signalling molecule in diverse cellular processes. Over the past few decades, studies of S1P signalling have revealed that the physiological activity of S1P largely depends on S1P metabolizing enzymes, transporters and receptors on the plasma membrane, as well as on the intracellular proteins that S1P binds directly to. In addition to its roles in cancer signalling, immunity and inflammation, a large body of evidence has identified a close link of S1P signalling with organ morphogenesis. Here we discuss the vital role of S1P signalling in orchestrating various cellular events during organ morphogenesis through analysing each component along the extracellular and intracellular S1P signalling axes. For each component, we review advances in our understanding of S1P signalling and function from the upstream regulators to the downstream effectors and from cellular behaviours to tissue organization, primarily in the context of morphogenetic mechanisms. S1P-mediated vesicular trafficking is also discussed as a function independent of its signalling function. A picture emerges that reveals a multifaceted role of S1P-dependent pathways in the development and maintenance of organ structure and function.
Collapse
Affiliation(s)
- Mengqiao Cui
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Verena Göbel
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, U.S.A
| | - Hongjie Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| |
Collapse
|
37
|
Geng X, Ho YC, Srinivasan RS. Biochemical and mechanical signals in the lymphatic vasculature. Cell Mol Life Sci 2021; 78:5903-5923. [PMID: 34240226 PMCID: PMC11072415 DOI: 10.1007/s00018-021-03886-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
Lymphatic vasculature is an integral part of the cardiovascular system where it maintains interstitial fluid balance. Additionally, lymphatic vasculature regulates lipid assimilation and inflammatory response. Lymphatic vasculature is composed of lymphatic capillaries, collecting lymphatic vessels and valves that function in synergy to absorb and transport fluid against gravitational and pressure gradients. Defects in lymphatic vessels or valves leads to fluid accumulation in tissues (lymphedema), chylous ascites, chylothorax, metabolic disorders and inflammation. The past three decades of research has identified numerous molecules that are necessary for the stepwise development of lymphatic vasculature. However, approaches to treat lymphatic disorders are still limited to massages and compression bandages. Hence, better understanding of the mechanisms that regulate lymphatic vascular development and function is urgently needed to develop efficient therapies. Recent research has linked mechanical signals such as shear stress and matrix stiffness with biochemical pathways that regulate lymphatic vessel growth, patterning and maturation and valve formation. The goal of this review article is to highlight these innovative developments and speculate on unanswered questions.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
38
|
Stritt S, Koltowska K, Mäkinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med 2021; 27:955-970. [PMID: 34332911 DOI: 10.1016/j.molmed.2021.07.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.
Collapse
Affiliation(s)
- Simon Stritt
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Katarzyna Koltowska
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden.
| |
Collapse
|
39
|
Regulation of VEGFR Signalling in Lymphatic Vascular Development and Disease: An Update. Int J Mol Sci 2021; 22:ijms22147760. [PMID: 34299378 PMCID: PMC8306507 DOI: 10.3390/ijms22147760] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/02/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
The importance of lymphatic vessels in a myriad of human diseases is rapidly gaining recognition; lymphatic vessel dysfunction is a feature of disorders including congenital lymphatic anomalies, primary lymphoedema and obesity, while improved lymphatic vessel function increases the efficacy of immunotherapy for cancer and neurological disease and promotes cardiac repair following myocardial infarction. Understanding how the growth and function of lymphatic vessels is precisely regulated therefore stands to inform the development of novel therapeutics applicable to a wide range of human diseases. Lymphatic vascular development is initiated during embryogenesis following establishment of the major blood vessels and the onset of blood flow. Lymphatic endothelial progenitor cells arise from a combination of venous and non-venous sources to generate the initial lymphatic vascular structures in the vertebrate embryo, which are then further ramified and remodelled to elaborate an extensive lymphatic vascular network. Signalling mediated via vascular endothelial growth factor (VEGF) family members and vascular endothelial growth factor receptor (VEGFR) tyrosine kinases is crucial for development of both the blood and lymphatic vascular networks, though distinct components are utilised to different degrees in each vascular compartment. Although much is known about the regulation of VEGFA/VEGFR2 signalling in the blood vasculature, less is understood regarding the mechanisms by which VEGFC/VEGFD/VEGFR3 signalling is regulated during lymphatic vascular development. This review will focus on recent advances in our understanding of the cellular and molecular mechanisms regulating VEGFA-, VEGFC- and VEGFD-mediated signalling via VEGFRs which are important for driving the construction of lymphatic vessels during development and disease.
Collapse
|
40
|
Francois M, Oszmiana A, Harvey NL. When form meets function: the cells and signals that shape the lymphatic vasculature during development. Development 2021; 148:268989. [PMID: 34080610 DOI: 10.1242/dev.167098] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The lymphatic vasculature is an integral component of the cardiovascular system. It is essential to maintain tissue fluid homeostasis, direct immune cell trafficking and absorb dietary lipids from the digestive tract. Major advances in our understanding of the genetic and cellular events important for constructing the lymphatic vasculature during development have recently been made. These include the identification of novel sources of lymphatic endothelial progenitor cells, the recognition of lymphatic endothelial cell specialisation and heterogeneity, and discovery of novel genes and signalling pathways underpinning developmental lymphangiogenesis. Here, we review these advances and discuss how they inform our understanding of lymphatic network formation, function and dysfunction.
Collapse
Affiliation(s)
- Mathias Francois
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing Program, The Centenary Institute, The University of Sydney, SOLES, 2006 Camperdown, Australia
| | - Anna Oszmiana
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5001, Australia
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5001, Australia
| |
Collapse
|
41
|
Hernández Vásquez MN, Ulvmar MH, González-Loyola A, Kritikos I, Sun Y, He L, Halin C, Petrova TV, Mäkinen T. Transcription factor FOXP2 is a flow-induced regulator of collecting lymphatic vessels. EMBO J 2021; 40:e107192. [PMID: 33934370 PMCID: PMC8204859 DOI: 10.15252/embj.2020107192] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
The lymphatic system is composed of a hierarchical network of fluid absorbing lymphatic capillaries and transporting collecting vessels. Despite distinct functions and morphologies, molecular mechanisms that regulate the identity of the different vessel types are poorly understood. Through transcriptional analysis of murine dermal lymphatic endothelial cells (LECs), we identified Foxp2, a member of the FOXP family of transcription factors implicated in speech development, as a collecting vessel signature gene. FOXP2 expression was induced after initiation of lymph flow in vivo and upon shear stress on primary LECs in vitro. Loss of FOXC2, the major flow-responsive transcriptional regulator of lymphatic valve formation, abolished FOXP2 induction in vitro and in vivo. Genetic deletion of Foxp2 in mice using the endothelial-specific Tie2-Cre or the tamoxifen-inducible LEC-specific Prox1-CreERT2 line resulted in enlarged collecting vessels and defective valves characterized by loss of NFATc1 activity. Our results identify FOXP2 as a new flow-induced transcriptional regulator of collecting lymphatic vessel morphogenesis and highlight the existence of unique transcription factor codes in the establishment of vessel-type-specific endothelial cell identities.
Collapse
Affiliation(s)
| | - Maria H Ulvmar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Alejandra González-Loyola
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Ioannis Kritikos
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Tatiana V Petrova
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
42
|
Mechanosensation and Mechanotransduction by Lymphatic Endothelial Cells Act as Important Regulators of Lymphatic Development and Function. Int J Mol Sci 2021; 22:ijms22083955. [PMID: 33921229 PMCID: PMC8070425 DOI: 10.3390/ijms22083955] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Our understanding of the function and development of the lymphatic system is expanding rapidly due to the identification of specific molecular markers and the availability of novel genetic approaches. In connection, it has been demonstrated that mechanical forces contribute to the endothelial cell fate commitment and play a critical role in influencing lymphatic endothelial cell shape and alignment by promoting sprouting, development, maturation of the lymphatic network, and coordinating lymphatic valve morphogenesis and the stabilization of lymphatic valves. However, the mechanosignaling and mechanotransduction pathways involved in these processes are poorly understood. Here, we provide an overview of the impact of mechanical forces on lymphatics and summarize the current understanding of the molecular mechanisms involved in the mechanosensation and mechanotransduction by lymphatic endothelial cells. We also discuss how these mechanosensitive pathways affect endothelial cell fate and regulate lymphatic development and function. A better understanding of these mechanisms may provide a deeper insight into the pathophysiology of various diseases associated with impaired lymphatic function, such as lymphedema and may eventually lead to the discovery of novel therapeutic targets for these conditions.
Collapse
|
43
|
Oxidatively Modified LDL Suppresses Lymphangiogenesis via CD36 Signaling. Antioxidants (Basel) 2021; 10:antiox10020331. [PMID: 33672291 PMCID: PMC7926875 DOI: 10.3390/antiox10020331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Arterial accumulation of plasma-derived LDL and its subsequent oxidation contributes to atherosclerosis. Lymphatic vessel (LV)-mediated removal of arterial cholesterol has been shown to reduce atherosclerotic lesion formation. However, the precise mechanisms that regulate LV density and function in atherosclerotic vessels remain to be identified. The aim of this study was to investigate the role of native LDL (nLDL) and oxidized LDL (oxLDL) in modulating lymphangiogenesis and underlying molecular mechanisms. Western blotting and immunostaining experiments demonstrated increased oxLDL expression in human atherosclerotic arteries. Furthermore, elevated oxLDL levels were detected in the adventitial layer, where LV are primarily present. Treatment of human lymphatic endothelial cells (LEC) with oxLDL inhibited in vitro tube formation, while nLDL stimulated it. Similar results were observed with Matrigel plug assay in vivo. CD36 deletion in mice and its siRNA-mediated knockdown in LEC prevented oxLDL-induced inhibition of lymphangiogenesis. In addition, oxLDL via CD36 receptor suppressed cell cycle, downregulated AKT and eNOS expression, and increased levels of p27 in LEC. Collectively, these results indicate that oxLDL inhibits lymphangiogenesis via CD36-mediated regulation of AKT/eNOS pathway and cell cycle. These findings suggest that therapeutic blockade of LEC CD36 may promote arterial lymphangiogenesis, leading to increased cholesterol removal from the arterial wall and reduced atherosclerosis.
Collapse
|
44
|
Norden PR, Kume T. Molecular Mechanisms Controlling Lymphatic Endothelial Junction Integrity. Front Cell Dev Biol 2021; 8:627647. [PMID: 33521001 PMCID: PMC7841202 DOI: 10.3389/fcell.2020.627647] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The lymphatic system is essential for lipid absorption/transport from the digestive system, maintenance of tissue fluid and protein homeostasis, and immune surveillance. Despite recent progress toward understanding the cellular and molecular mechanisms underlying the formation of the lymphatic vascular system, the nature of lymphatic vessel abnormalities and disease in humans is complex and poorly understood. The mature lymphatic vasculature forms a hierarchical network in which lymphatic endothelial cells (LECs) are joined by functionally specialized cell-cell junctions to maintain the integrity of lymphatic vessels. Blind-ended and highly permeable lymphatic capillaries drain interstitial fluid via discontinuous, button-like LEC junctions, whereas collecting lymphatic vessels, surrounded by intact basement membranes and lymphatic smooth muscle cells, have continuous, zipper-like LEC junctions to transport lymph to the blood circulatory system without leakage. In this review, we discuss the recent advances in our understanding of the mechanisms by which lymphatic button- and zipper-like junctions play critical roles in lymphatic permeability and function in a tissue- and organ-specific manner, including lacteals of the small intestine. We also provide current knowledge related to key pathways and factors such as VEGF and RhoA/ROCK signaling that control lymphatic endothelial cell junctional integrity.
Collapse
Affiliation(s)
- Pieter R Norden
- Department of Medicine, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, United States
| |
Collapse
|
45
|
Paulson D, Harms R, Ward C, Latterell M, Pazour GJ, Fink DM. Loss of Primary Cilia Protein IFT20 Dysregulates Lymphatic Vessel Patterning in Development and Inflammation. Front Cell Dev Biol 2021; 9:672625. [PMID: 34055805 PMCID: PMC8160126 DOI: 10.3389/fcell.2021.672625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Microenvironmental signals produced during development or inflammation stimulate lymphatic endothelial cells to undergo lymphangiogenesis, in which they sprout, proliferate, and migrate to expand the vascular network. Many cell types detect changes in extracellular conditions via primary cilia, microtubule-based cellular protrusions that house specialized membrane receptors and signaling complexes. Primary cilia are critical for receipt of extracellular cues from both ligand-receptor pathways and physical forces such as fluid shear stress. Here, we report the presence of primary cilia on immortalized mouse and primary adult human dermal lymphatic endothelial cells in vitro and on both luminal and abluminal domains of mouse corneal, skin, and mesenteric lymphatic vessels in vivo. The purpose of this study was to determine the effects of disrupting primary cilia on lymphatic vessel patterning during development and inflammation. Intraflagellar transport protein 20 (IFT20) is part of the transport machinery required for ciliary assembly and function. To disrupt primary ciliary signaling, we generated global and lymphatic endothelium-specific IFT20 knockout mouse models and used immunofluorescence microscopy to quantify changes in lymphatic vessel patterning at E16.5 and in adult suture-mediated corneal lymphangiogenesis. Loss of IFT20 during development resulted in edema, increased and more variable lymphatic vessel caliber and branching, as well as red blood cell-filled lymphatics. We used a corneal suture model to determine ciliation status of lymphatic vessels during acute, recurrent, and tumor-associated inflammatory reactions and wound healing. Primary cilia were present on corneal lymphatics during all of the mechanistically distinct lymphatic patterning events of the model and assembled on lymphatic endothelial cells residing at the limbus, stalk, and vessel tip. Lymphatic-specific deletion of IFT20 cell-autonomously exacerbated acute corneal lymphangiogenesis resulting in increased lymphatic vessel density and branching. These data are the first functional studies of primary cilia on lymphatic endothelial cells and reveal a new dimension in regulation of lymphatic vascular biology.
Collapse
Affiliation(s)
- Delayna Paulson
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Rebecca Harms
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Cody Ward
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Mackenzie Latterell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Darci M. Fink
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
- *Correspondence: Darci M. Fink,
| |
Collapse
|
46
|
Engelbrecht E, MacRae CA, Hla T. Lysolipids in Vascular Development, Biology, and Disease. Arterioscler Thromb Vasc Biol 2020; 41:564-584. [PMID: 33327749 DOI: 10.1161/atvbaha.120.305565] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Membrane phospholipid metabolism forms lysophospholipids, which possess unique biochemical and biophysical properties that influence membrane structure and dynamics. However, lysophospholipids also function as ligands for G-protein-coupled receptors that influence embryonic development, postnatal physiology, and disease. The 2 most well-studied species-lysophosphatidic acid and S1P (sphingosine 1-phosphate)-are particularly relevant to vascular development, physiology, and cardiovascular diseases. This review summarizes the role of lysophosphatidic acid and S1P in vascular developmental processes, endothelial cell biology, and their roles in cardiovascular disease processes. In addition, we also point out the apparent connections between lysophospholipid biology and the Wnt (int/wingless family) pathway, an evolutionarily conserved fundamental developmental signaling system. The discovery that components of the lysophospholipid signaling system are key genetic determinants of cardiovascular disease has warranted current and future research in this field. As pharmacological approaches to modulate lysophospholipid signaling have entered the clinical sphere, new findings in this field promise to influence novel therapeutic strategies in cardiovascular diseases.
Collapse
Affiliation(s)
- Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery (E.E., T.H.), Harvard Medical School, Boston, MA
| | - Calum A MacRae
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Department of Medicine (C.A.M.), Harvard Medical School, Boston, MA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery (E.E., T.H.), Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Cha B, Ho YC, Geng X, Mahamud MR, Chen L, Kim Y, Choi D, Kim TH, Randolph GJ, Cao X, Chen H, Srinivasan RS. YAP and TAZ maintain PROX1 expression in the developing lymphatic and lymphovenous valves in response to VEGF-C signaling. Development 2020; 147:dev195453. [PMID: 33060128 PMCID: PMC7758626 DOI: 10.1242/dev.195453] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/22/2020] [Indexed: 01/07/2023]
Abstract
Lymphatic vasculature is an integral part of digestive, immune and circulatory systems. The homeobox transcription factor PROX1 is necessary for the development of lymphatic vessels, lymphatic valves (LVs) and lymphovenous valves (LVVs). We and others previously reported a feedback loop between PROX1 and vascular endothelial growth factor-C (VEGF-C) signaling. PROX1 promotes the expression of the VEGF-C receptor VEGFR3 in lymphatic endothelial cells (LECs). In turn, VEGF-C signaling maintains PROX1 expression in LECs. However, the mechanisms of PROX1/VEGF-C feedback loop remain poorly understood. Whether VEGF-C signaling is necessary for LV and LVV development is also unknown. Here, we report for the first time that VEGF-C signaling is necessary for valve morphogenesis. We have also discovered that the transcriptional co-activators YAP and TAZ are required to maintain PROX1 expression in LVs and LVVs in response to VEGF-C signaling. Deletion of Yap and Taz in the lymphatic vasculature of mouse embryos did not affect the formation of LVs or LVVs, but resulted in the degeneration of these structures. Our results have identified VEGF-C, YAP and TAZ as a crucial molecular pathway in valve development.
Collapse
Affiliation(s)
- Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Md Riaj Mahamud
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Lijuan Chen
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Yeunhee Kim
- Department of Biological Sciences and Center for Systems Biology, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Dongwon Choi
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tae Hoon Kim
- Department of Biological Sciences and Center for Systems Biology, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Xinwei Cao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| |
Collapse
|
48
|
Baluk P, Naikawadi RP, Kim S, Rodriguez F, Choi D, Hong YK, Wolters PJ, McDonald DM. Lymphatic Proliferation Ameliorates Pulmonary Fibrosis after Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2355-2375. [PMID: 33039355 DOI: 10.1016/j.ajpath.2020.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/09/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Despite many reports about pulmonary blood vessels in lung fibrosis, the contribution of lymphatics to fibrosis is unknown. We examined the mechanism and consequences of lymphatic remodeling in mice with lung fibrosis after bleomycin injury or telomere dysfunction. Widespread lymphangiogenesis was observed after bleomycin treatment and in fibrotic lungs of prospero homeobox 1-enhanced green fluorescent protein (Prox1-EGFP) transgenic mice with telomere dysfunction. In loss-of-function studies, blocking antibodies revealed that lymphangiogenesis 14 days after bleomycin treatment was dependent on vascular endothelial growth factor (Vegf) receptor 3 signaling, but not on Vegf receptor 2. Vegfc gene and protein expression increased specifically. Extensive extravasated plasma, platelets, and macrophages at sites of lymphatic growth were potential sources of Vegfc. Lymphangiogenesis peaked at 14 to 28 days after bleomycin challenge, was accompanied by doubling of chemokine (C-C motif) ligand 21 in lung lymphatics and tertiary lymphoid organ formation, and then decreased as lung injury resolved by 56 days. In gain-of-function studies, expansion of the lung lymphatic network by transgenic overexpression of Vegfc in club cell secretory protein (CCSP)/VEGF-C mice reduced macrophage accumulation and fibrosis and accelerated recovery after bleomycin treatment. These findings suggest that lymphatics have an overall protective effect in lung injury and fibrosis and fit with a mechanism whereby lung lymphatic network expansion reduces lymph stasis and increases clearance of fluid and cells, including profibrotic macrophages.
Collapse
Affiliation(s)
- Peter Baluk
- Department of Anatomy, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
| | - Ram P Naikawadi
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, California
| | - Shineui Kim
- Department of Anatomy, University of California, San Francisco, San Francisco, California
| | - Felipe Rodriguez
- Department of Anatomy, University of California, San Francisco, San Francisco, California
| | - Dongwon Choi
- Department of Surgery, University of Southern California, Los Angeles, California
| | - Young-Kwon Hong
- Department of Surgery, University of Southern California, Los Angeles, California
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, California
| | - Donald M McDonald
- Department of Anatomy, University of California, San Francisco, San Francisco, California; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
| |
Collapse
|