1
|
Zhang Q, Niu Y, Li Y, Xia C, Chen Z, Chen Y, Feng H. Meningeal lymphatic drainage: novel insights into central nervous system disease. Signal Transduct Target Ther 2025; 10:142. [PMID: 40320416 PMCID: PMC12050339 DOI: 10.1038/s41392-025-02177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 05/08/2025] Open
Abstract
In recent years, increasing evidence has suggested that meningeal lymphatic drainage plays a significant role in central nervous system (CNS) diseases. Studies have indicated that CNS diseases and conditions associated with meningeal lymphatic drainage dysfunction include neurodegenerative diseases, stroke, infections, traumatic brain injury, tumors, functional cranial disorders, and hydrocephalus. However, the understanding of the regulatory and damage mechanisms of meningeal lymphatics under physiological and pathological conditions is currently limited. Given the importance of a profound understanding of the interplay between meningeal lymphatic drainage and CNS diseases, this review covers seven key aspects: the development and structure of meningeal lymphatic vessels, methods for observing meningeal lymphatics, the function of meningeal lymphatics, the molecular mechanisms of meningeal lymphatic injury, the relationships between meningeal lymphatic vessels and CNS diseases, potential regulatory mechanisms of meningeal lymphatics, and conclusions and outstanding questions. We will explore the relationship between the development, structure, and function of meningeal lymphatics, review current methods for observing meningeal lymphatic vessels in both animal models and humans, and identify unresolved key points in meningeal lymphatic research. The aim of this review is to provide new directions for future research and therapeutic strategies targeting meningeal lymphatics by critically analyzing recent advancements in the field, identifying gaps in current knowledge, and proposing innovative approaches to address these gaps.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Neurosurgery, The 961st Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yin Niu
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yingpei Li
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chenyang Xia
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhi Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
2
|
Schoofs H, Daubel N, Schnabellehner S, Grönloh MLB, Palacios Martínez S, Halme A, Marks AM, Jeansson M, Barcos S, Brakebusch C, Benedito R, Engelhardt B, Vestweber D, Gaengel K, Linsenmeier F, Schürmann S, Saharinen P, van Buul JD, Friedrich O, Smith RS, Majda M, Mäkinen T. Dynamic cytoskeletal regulation of cell shape supports resilience of lymphatic endothelium. Nature 2025; 641:465-475. [PMID: 40108458 PMCID: PMC12058511 DOI: 10.1038/s41586-025-08724-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/31/2025] [Indexed: 03/22/2025]
Abstract
Lymphatic capillaries continuously take up interstitial fluid and adapt to resulting changes in vessel calibre1-3. The mechanisms by which the permeable monolayer of loosely connected lymphatic endothelial cells (LECs)4 maintains mechanical stability remain elusive. Here we identify dynamic cytoskeletal regulation of LEC shape, induced by isotropic stretch, as crucial for the integrity and function of dermal lymphatic capillaries. We found that the oak leaf-shaped LECs showed a spectrum of VE-cadherin-based junctional configurations at the lobular intercellular interface and a unique cytoskeletal organization, with microtubules at concave regions and F-actin at convex lobes. Multispectral and longitudinal intravital imaging of capillary LEC shape and actin revealed dynamic remodelling of cellular overlaps in vivo during homeostasis and in response to interstitial fluid volume increase. Akin to puzzle cells of the plant epidermis5,6, LEC shape was controlled by Rho GTPase CDC42-regulated cytoskeletal dynamics, enhancing monolayer stability. Moreover, cyclic isotropic stretch increased cellular overlaps and junction curvature in primary LECs. Our findings indicate that capillary LEC shape results from continuous remodelling of cellular overlaps that maintain vessel integrity while preserving permeable cell-cell contacts compatible with vessel expansion and fluid uptake. We propose a bellows-like fluid propulsion mechanism, in which fluid-induced lumen expansion and shrinkage of LEC overlaps are countered by actin-based lamellipodia-like overlap extension to aid vessel constriction.
Collapse
Affiliation(s)
- Hans Schoofs
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nina Daubel
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sarah Schnabellehner
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Max L B Grönloh
- Department of Medical Biochemistry at the Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Sebastián Palacios Martínez
- Department of Molecular Cytology, Leeuwenhoek Centre for Advanced Microscopy at Swammerdam Institute for Life Sciences at the University of Amsterdam, Amsterdam, The Netherlands
| | - Aleksi Halme
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Amanda M Marks
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marie Jeansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sara Barcos
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Cord Brakebusch
- Biotech Research and Innovation Center, University of Copenhagen, Copenhagen, Denmark
| | - Rui Benedito
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | | | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fabian Linsenmeier
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Schürmann
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Pipsa Saharinen
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Jaap D van Buul
- Department of Medical Biochemistry at the Amsterdam UMC, location AMC, Amsterdam, The Netherlands
- Department of Molecular Cytology, Leeuwenhoek Centre for Advanced Microscopy at Swammerdam Institute for Life Sciences at the University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Mateusz Majda
- Department of Plant Molecular Biology, University of Lausanne, Lausanne, Switzerland
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Helsinki, Finland.
| |
Collapse
|
3
|
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
|
4
|
Jäger J, Thon M, Schimek K, Marx U, Gibbs S, Koning JJ. Differential biomarker expression of blood and lymphatic vasculature in multi-organ-chips. Sci Rep 2025; 15:14492. [PMID: 40281034 PMCID: PMC12032159 DOI: 10.1038/s41598-025-96367-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Since the blood and lymphatic endothelium regulates homeostasis and inflammation during health and disease, establishment of vascularized Organ-on-Chip platforms with blood and lymphatic endothelial cells (BEC/LEC) is a pre-requisite to further advance the field of tissue engineering. Here, we aimed to determine whether characteristics of BECs and LECs cultured under flow in a multi-organ-chip (MOC) are influenced by shear stress or inflammation. Dermis-derived primary BECs and LECs were used to endothelialize a MOC followed by culture for up to 14 days at lymphatic and blood flow rates. Under blood flow, both cell types changed morphology, aligned in flow direction, and showed close cell-cell contacts as in in vivo blood vasculature. Under lymphatic flow, neither BEC nor LEC aligned, and both showed a cobblestone-appearance with limited intercellular contacts similar to lymphatics. Cells retained their cell type-specific phenotype and cytokine secretion profiles. CCL21 expression in LECs was rescued by flow, but diminished again with TNFα exposure, together with the LEC-specific markers PROX1 and TFF3. Homeostatic cytokine secretion was higher in BECs, but the response to TNFα was more pronounced in LECs. Results indicate that BEC and LEC phenotype and cytokine secretion is mostly an intrinsic property with only morphology and CCL21 being influenced by flow.
Collapse
Affiliation(s)
- Jonas Jäger
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Maria Thon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands
| | | | | | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands.
- Amsterdam Institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Deshpande P, Dornbrand-Lo M, Phondge V, Kelly P, Wong AK. Tissue engineering approaches for lymphedema: biomaterial innovations and clinical potential. Front Cell Dev Biol 2025; 13:1537050. [PMID: 40302939 PMCID: PMC12037638 DOI: 10.3389/fcell.2025.1537050] [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: 11/29/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
The lymphatic system plays a critical role in maintaining fluid balance and immune regulation. Lymphedema, and other lymphatic disorders, highlight the need for advanced therapeutic approaches, including tissue engineering. This review examines the latest developments in artificial lymphatic tissue engineering, focusing on scaffold materials, lymphangiogenic factors, and regenerative strategies to replicate the intricacy of lymphatic vessels and nodes. We conducted a thorough literature review of current practices and applications in lymphatic tissue engineering. Findings show that biomaterials such as hydrogels, decellularized matrices, and synthetic polymers provide effective scaffolds for lymphatic endothelial cell proliferation and lymphangiogenesis. Advances in growth factor delivery and stem-cell based therapies have further enhanced the viability of engineered lymphatic tissues. Despite promising progress, challenges in achieving functional replication of lymphatic structures and clinical translation of research remain. Ongoing research must address scaffold biocompatibility, optimized growth factor targeting, and scalable production to advance therapeutic options for lymphatic disorders. This review underscores the potential for transformative patient outcomes through innovative bioengineering solutions.
Collapse
Affiliation(s)
| | | | | | | | - Alex K. Wong
- Rutgers New Jersey Medical School, Division of Plastic and Reconstructive Surgery, Newark, NJ, United States
| |
Collapse
|
6
|
Mastrogiacomo DM, Price A, Fu Y, Banerjee R, Knauer LA, Li K, Yang Y, Davis GE, Dellinger MT, Scallan JP. Lymphatic Malformations with Activating KRAS Mutations Impair Lymphatic Valve Development Through Matrix Metalloproteinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646922. [PMID: 40236167 PMCID: PMC11996454 DOI: 10.1101/2025.04.02.646922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
BACKGROUND Lymphatic malformations (LMs) are lesions due to inherited or somatic mutations that lead to a defective lymphatic vasculature. Activating KRAS mutations have been identified recently in LM patients with lymphedema, chylous ascites, or life-threatening chylothorax. In a LM mouse model, KRAS mutations are associated with a loss of lymphatic valves, which has been proposed to cause chylothorax via retrograde lymph flow into the pleural space. However, the mechanisms underlying the loss of lymphatic valves are unknown. METHODS To investigate the mechanisms leading to valve loss, we combined the lymphatic-specific and tamoxifen-inducible Flt4CreER T2 with Kras-loxP-stop-loxP-G12D ( Kras +/G12D ) mice and Prox1GFP reporter mice to induce the restricted expression of KRAS-G12D and enable valve quantification in postnatal pups. Human dermal lymphatic endothelial cells (hdLECs) expressing KRAS-G12D were probed for changes in mRNA and protein expression with qRT-PCR, western blot, and gel zymography, and mechanistic studies were performed using 3D cell culture in collagen matrices. RESULTS Our data showed that lymphatic-specific expression of KRAS-G12D significantly attenuated valve development in the mesentery, diaphragm, and ear skin. qRT-PCR, western blot, and gel zymography using hdLECs expressing KRAS-G12D revealed the upregulation of the plasminogen activator (PA) pathway and matrix metalloproteinases (MMPs). The MMPs were sufficiently activated by plasmin, the product of the PA pathway, in hdLECs grown in a 3D collagen matrix, indicating a role for MMPs in the degradation of valve ECM core. Furthermore, a broad-spectrum MMP inhibitor given to Flt4CreER T2 ;Kras +/G12D mice rescued lymphatic valve development. CONCLUSIONS We conclude that hyperactive KRAS signaling upregulates MMPs that become excessively activated by the upregulation of the PA pathway. MMPs then degrade the lymphatic valve ECM core preventing valve formation.
Collapse
|
7
|
Andrey T, Alexander S, Inna B, Daria Z, Viktoria A, Anastasiia SG, Kumar A, Ivan F, Arina E, Oxana SG. Age as a limiting factor for effectiveness of photostimulation of brain drainage and cognitive functions. FRONTIERS OF OPTOELECTRONICS 2025; 18:6. [PMID: 40163163 PMCID: PMC11958890 DOI: 10.1007/s12200-025-00149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/21/2025] [Indexed: 04/02/2025]
Abstract
The progressive number of old adults with cognitive impairment worldwide and the lack of effective pharmacologic therapies require the development of non-pharmacologic strategies. The photobiomodulation (PBM) is a promising method in prevention of early or mild age-related cognitive impairments. However, it remains unclear the efficacy of PBM for old patients with significant age-related cognitive dysfunction. In our study on male mice, we show a gradual increase in the brain amyloid beta (Aβ) levels and a decrease in brain drainage with age, which, however, is associated with a decline in cognitive function only in old (24 months of age) mice but not in middle-aged (12 months of age) and young (3 month of age) animals. These age-related features are accompanied by the development of hyperplasia of the meningeal lymphatic vessels (MLVs) in old mice underlying the decrease in brain drainage. PBM improves cognitive training exercises and Aβ clearance only in young and middle-aged mice, while old animals are not sensitive to PBM. These results clearly demonstrate that the PBM effects on cognitive function are correlated with age-mediated changes in the MLV network and may be effective if the MLV function is preserved. These findings expand fundamental knowledge about age differences in the effectiveness of PBM for improvement of cognitive functions and Aβ clearance as well as about the lymphatic mechanisms responsible for age decline in sensitivity to the therapeutic PBM effects.
Collapse
Affiliation(s)
- Terskov Andrey
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | - Shirokov Alexander
- Department of Biology, Saratov State University, Saratov, 410012, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, 410049, Russia
| | - Blokhina Inna
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Adushkina Viktoria
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Atul Kumar
- The Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, Varanasi, 221005, India
| | - Fedosov Ivan
- Institute of Physics, Saratov State University, Saratov, 410012, Russia
| | - Evsukova Arina
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | |
Collapse
|
8
|
Razavi MS, Munn LL, Padera TP. Mechanics of Lymphatic Pumping and Lymphatic Function. Cold Spring Harb Perspect Med 2025; 15:a041171. [PMID: 38692743 PMCID: PMC11875091 DOI: 10.1101/cshperspect.a041171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
The lymphatic system plays a crucial role in maintaining tissue fluid balance, immune surveillance, and the transport of lipids and macromolecules. Lymph is absorbed by initial lymphatics and then driven through lymph nodes and to the blood circulation by the contraction of collecting lymphatic vessels. Intraluminal valves in collecting lymphatic vessels ensure the unidirectional flow of lymph centrally. The lymphatic muscle cells that invest in collecting lymphatic vessels impart energy to propel lymph against hydrostatic pressure gradients and gravity. A variety of mechanical and biochemical stimuli modulate the contractile activity of lymphatic vessels. This review focuses on the recent advances in our understanding of the mechanisms involved in regulating and collecting lymphatic vessel pumping in normal tissues and the association between lymphatic pumping, infection, inflammatory disease states, and lymphedema.
Collapse
Affiliation(s)
- Mohammad S Razavi
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Timothy P Padera
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
9
|
Tso P, Bernier-Latmani J, Petrova TV, Liu M. Transport functions of intestinal lymphatic vessels. Nat Rev Gastroenterol Hepatol 2025; 22:127-145. [PMID: 39496888 DOI: 10.1038/s41575-024-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/06/2024]
Abstract
Lymphatic vessels are crucial for fluid absorption and the transport of peripheral immune cells to lymph nodes. However, in the small intestine, the lymphatic fluid is rich in diet-derived lipids incorporated into chylomicrons and gut-specific immune cells. Thus, intestinal lymphatic vessels have evolved to handle these unique cargoes and are critical for systemic dietary lipid delivery and metabolism. This Review covers mechanisms of lipid absorption from epithelial cells to the lymphatics as well as unique features of the gut microenvironment that affect these functions. Moreover, we discuss details of the intestinal lymphatics in gut immune cell trafficking and insights into the role of inter-organ communication. Lastly, we highlight the particularities of fat absorption that can be harnessed for efficient lipid-soluble drug distribution for novel therapies, including the ability of chylomicron-associated drugs to bypass first-pass liver metabolism for systemic delivery. In all, this Review will help to promote an understanding of intestinal lymphatic-systemic interactions to guide future research directions.
Collapse
Affiliation(s)
- Patrick Tso
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Min Liu
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
10
|
Fukui H, Chow RWY, Yap CH, Vermot J. Rhythmic forces shaping the zebrafish cardiac system. Trends Cell Biol 2025; 35:166-176. [PMID: 39665884 DOI: 10.1016/j.tcb.2024.10.012] [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: 03/06/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024]
Abstract
The structural development of the heart depends heavily on mechanical forces, and rhythmic contractions generate essential physical stimuli during morphogenesis. Cardiac cells play a critical role in coordinating this process by sensing and responding to these mechanical forces. In vivo, cells experience rhythmic spatial and temporal variations in deformation-related stresses throughout heart development. What impact do these mechanical forces have on heart morphogenesis? Recent work in zebrafish (Danio rerio) offers important insights into this question. This review focuses on endocardial (EdCs) and myocardial cells (cardiomyocytes, CMs), key cell types in the heart, and provides a comprehensive overview of forces and tissue mechanics in zebrafish and their direct influence on cardiac cell identity.
Collapse
Affiliation(s)
- Hajime Fukui
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan; Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Renee Wei-Yan Chow
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Choon Hwai Yap
- Department of Bioengineering, Imperial College London, London, UK
| | - Julien Vermot
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
11
|
Chen D, Tang Y, Lapinski PE, Wiggins D, Sevick EM, Davis MJ, King PD. EPHB4-RASA1 Inhibition of PIEZO1 Ras Activation Drives Lymphatic Valvulogenesis. Circ Res 2024; 135:1048-1066. [PMID: 39421925 PMCID: PMC11560524 DOI: 10.1161/circresaha.124.325383] [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: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND EPHB4 (ephrin receptor B4) and the RASA1 (p120 Ras GTPase-activating protein) are necessary for the development of lymphatic vessel (LV) valves. However, precisely how EPHB4 and RASA1 regulate LV valve development is unknown. In this study, we examine the mechanisms by which EPHB4 and RASA1 regulate the development of LV valves. METHODS We used LV-specific inducible EPHB4-deficient mice and EPHB4 knockin mice that express a form of EPHB4 that is unable to bind RASA1 yet retains protein tyrosine kinase activity (EPHB4 2YP) to study the role of EPHB4 and RASA1 in LV valve development in the embryo and LV valve maintenance in adults. We also used human dermal lymphatic endothelial cells in vitro to study the role of EPHB4 and RASA1 as regulators of LV valve specification induced by oscillatory shear stress, considered the trigger for LV valve specification in vivo. RESULTS LV valve specification, continued valve development postspecification, and LV valve maintenance were blocked upon induced loss of EPHB4 in LV. LV valve specification and maintenance were also impaired in EPHB4 2YP mice. Defects in LV valve development were reversed by inhibition of the Ras-MAPK (mitogen-activated protein kinase) signaling pathway. In human dermal lymphatic endothelial cells, loss of expression of EPHB4 or its ephrin b2 ligand, loss of expression of RASA1, and inhibition of physical interaction between EPHB4 and RASA1 resulted in dysregulated oscillatory shear stress-induced Ras-MAPK activation and impaired expression of LV specification markers that could be rescued by Ras-MAPK pathway inhibition. The same results were observed when human dermal lymphatic endothelial cells were stimulated with the Yoda1 agonist of the PIEZO1 oscillatory shear stress sensor. Although Yoda1 increased the number of LV valves when administered to wild-type embryos, it did not increase LV valve number when administered to EPHB4 2YP embryos. CONCLUSIONS EPHB4 is necessary for LV valve specification, continued valve development postspecification, and valve maintenance. LV valve specification requires physical interaction between EPHB4 and RASA1 to limit activation of the Ras-MAPK pathway in lymphatic endothelial cells. Specifically, EPHB4-RASA1 physical interaction is necessary to dampen Ras-MAPK activation induced through the PIEZO1 oscillatory shear stress sensor. These findings reveal the mechanism by which EPHB4 and RASA1 regulate the development of LV valves.
Collapse
Affiliation(s)
- Di Chen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - Yipei Tang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - Philip E. Lapinski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| | - David Wiggins
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Eva M. Sevick
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Philip D. King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48103, USA
| |
Collapse
|
12
|
Janardhan HP, Wachter BT, Trivedi CM. Lymphatic System Development and Function. Curr Cardiol Rep 2024; 26:1209-1219. [PMID: 39172295 DOI: 10.1007/s11886-024-02120-8] [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] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE OF REVIEW This review delves into recent advancements in understanding generalized and organ-specific lymphatic development. It emphasizes the distinct characteristics and critical anomalies that can impair lymphatic function. By exploring developmental mechanisms, the review seeks to illuminate the profound impact of lymphatic malformations on overall health and disease progression. RECENT FINDINGS The introduction of genome sequencing, single-cell transcriptomic analysis, and advanced imaging technologies has significantly enhanced our ability to identify and characterize developmental defects within the lymphatic system. As a result, a wide range of lymphatic anomalies have been uncovered, spanning from congenital abnormalities present at birth to conditions that can become life-threatening in adulthood. Additionally, recent research highlights the heterogeneity of lymphatics, revealing organ-specific developmental pathways, unique molecular markers, and specialized physiological functions specific to each organ. A deeper understanding of the unique characteristics of lymphatic cell populations in an organ-specific context is essential for guiding future research into lymphatic disease processes. An integrated approach to translational research could revolutionize personalized medicine, where treatments are precisely tailored to individual lymphatic profiles, enhancing effectiveness and minimizing side effects.
Collapse
Affiliation(s)
- Harish P Janardhan
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Brianna T Wachter
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
- MD-PhD Program, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
- MD-PhD Program, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Department of Molecular, Cell, and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
13
|
Arroyo-Ataz G, Jones D. Overview of Lymphatic Muscle Cells in Development, Physiology, and Disease. Microcirculation 2024; 31:e12887. [PMID: 39329178 PMCID: PMC11560633 DOI: 10.1111/micc.12887] [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: 06/17/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Lymphatic muscle cells (LMCs) are indispensable for proper functioning of the lymphatic system, as they provide the driving force for lymph transport. Recent studies have advanced our understanding of the molecular mechanisms that regulate LMCs, which control rhythmic contraction and vessel tone of lymphatic vessels-traits also found in cardiac and vascular smooth muscle. In this review, we discuss the molecular pathways that orchestrate LMC-mediated contractility and summarize current knowledge about their developmental origin, which may shed light on the distinct contractile characteristics of LMCs. Further, we highlight the growing evidence implicating LMC dysregulation in the pathogenesis of lymphedema and other diseases related to lymphatic vessel dysfunction. Given the limited number and efficacy of existing therapies to treat lymphedema, LMCs present a promising focus for identifying novel therapeutic targets aimed at improving lymphatic vessel contractility. Here, we discuss LMCs in health and disease, as well as therapeutic strategies aimed at targeting them to improve lymphatic vessel function.
Collapse
Affiliation(s)
- Guillermo Arroyo-Ataz
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| | - Dennis Jones
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| |
Collapse
|
14
|
Chen Y, Liu X, Yuan J, Dong S, Nie M, Jiang W, Wu D, Liu M, Liu T, Wu C, Gao C, Zhang J, Jiang R. Vitamin D accelerates the subdural hematoma clearance through improving the meningeal lymphatic vessel function. Mol Cell Biochem 2024; 479:3129-3140. [PMID: 38294731 DOI: 10.1007/s11010-023-04918-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024]
Abstract
Subdural hematoma (SDH) drains into the extracranial lymphatic system through the meningeal lymphatic vessels (mLVs) but the formation of SDH impairs mLVs. Because vitamin D (Vit D) can protect the endothelial cells, we hypothesized that Vit D may enhance the SDH clearance. SDH was induced in Sprague-Dawley rats and treated with Vit D or vehicle. Hematoma volume in each group was measured by H&E staining and hemoglobin quantification. Evans blue (EB) quantification and red blood cells injection were used to evaluated the drainage of mLVs. Western blot analysis and immunofluorescence were conducted to assess the expression of lymphatic protein markers. We also examined the inflammatory factors levels in subdural space by ELISA. Vit D treatment significantly reduced SDH volume and improved the drainage of SDH to cervical lymph nodes. The structure of mLVs in SDH rats were protected by Vit D, and the expressions of LYVE1, PROX1, FOXC2, and VE-cadherin were increased after Vit D treatment. The TNF-α, IL-6, and IL-8 levels were reduced in Vit D group. In vitro, Vit D also increased the VE-cadherin expression levels under inflammation. Vit D protects the structure of mLVs and enhances the absorption of SDH, partly by the anti-inflammatory effect of Vit D.
Collapse
Affiliation(s)
- Yupeng Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Xuanhui Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Shiying Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Meng Nie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Weiwei Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Di Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Mingqi Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Tao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China
| | - Chuang Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China.
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education, Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, 154 Anshan Road, Helping District, Tianjin, 300052, China.
| |
Collapse
|
15
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Shalaby H, Dehghan R, Scallan JP, Yang Y. Rictor, an mTORC2 Protein, Regulates Murine Lymphatic Valve Formation Through the AKT-FOXO1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:2004-2023. [PMID: 39087350 PMCID: PMC11335088 DOI: 10.1161/atvbaha.124.321164] [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: 04/29/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Lymphatic valves are specialized structures in collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves. Conventionally, in many cell types, AKT is phosphorylated at Ser473 by the mTORC2 (mammalian target of rapamycin complex 2). However, mTORC2 has not yet been implicated in lymphatic valve formation. METHODS In vivo and in vitro techniques were used to investigate the role of Rictor, a critical component of mTORC2, in lymphatic endothelium. RESULTS Here, we showed that embryonic and postnatal lymphatic deletion of Rictor, a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human dermal lymphatic endothelial cells not only reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions but also abolished the upregulation of AKT activity and valve-forming genes in response to oscillatory shear stress. We further showed that the AKT target, FOXO1 (forkhead box protein O1), a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric lymphatic endothelial cells in vivo. Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in lymphatic vessels of the ear and mesentery. CONCLUSIONS Our work identifies a novel role for RICTOR in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately enables the formation and maintenance of lymphatic valves.
Collapse
Affiliation(s)
- Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Luz A. Knauer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Sara E. Barlow
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Hanan Shalaby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Razieh Dehghan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
16
|
Bertram CD, Macaskill C. Fluid-Dynamic Modeling of Flow in Embryonic Tissue Indicates That Lymphatic Valve Location Is Not Consistently Determined by the Local Fluid Shear or Its Gradient. Microcirculation 2024; 31:e12873. [PMID: 38953384 PMCID: PMC11303113 DOI: 10.1111/micc.12873] [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: 03/04/2024] [Revised: 05/02/2024] [Accepted: 06/08/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVE Intravascular lymphatic valves often occur in proximity to vessel junctions. It is commonly held that disturbed flow at junctions is responsible for accumulation of valve-forming cells (VFCs) at these locations as the initial step in valve creation, and the one which explains the association with these sites. However, evidence in favor is largely limited to cell culture experiments. METHODS We acquired images of embryonic lymphatic vascular networks from day E16.5, when VFC accumulation has started but the developing valve has not yet altered the local vessel geometry, stained for Prox1, which co-localizes with Foxc2. Using finite-element computational fluid mechanics, we simulated the flow through the networks, under conditions appropriate to this early development stage. Then we correlated the Prox1 distributions with the distributions of simulated fluid shear and shear stress gradient. RESULTS Across a total of 16 image sets, no consistent correlation was found between Prox1 distribution and the local magnitude of fluid shear, or its positive or negative gradient. CONCLUSIONS This, the first direct semi-empirical test of the localization hypothesis to interrogate the tissue from in vivo at the critical moment of development, does not support the idea that a feature of the local flow determines valve localization.
Collapse
Affiliation(s)
- Christopher D Bertram
- School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| | - Charlie Macaskill
- School of Mathematics and Statistics, University of Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Du J, Liu P, Zhou Y, Misener S, Sharma I, Leeaw P, Thomson BR, Jin J, Quaggin SE. The mechanosensory channel PIEZO1 functions upstream of angiopoietin/TIE/FOXO1 signaling in lymphatic development. J Clin Invest 2024; 134:e176577. [PMID: 38747287 PMCID: PMC11093609 DOI: 10.1172/jci176577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/12/2024] [Indexed: 05/19/2024] Open
Abstract
Lymphedema is a debilitating disease with no effective cure and affects an estimated 250 million individuals worldwide. Prior studies have identified mutations in piezo-type mechanosensitive ion channel component 1 (PIEZO1), angiopoietin 2 (ANGPT2), and tyrosine kinase with Ig-like and EGF-like domains 1 (TIE1) in patients with primary lymphedema. Here, we identified crosstalk between these molecules and showed that activation of the mechanosensory channel PIEZO1 in lymphatic endothelial cells (LECs) caused rapid exocytosis of the TIE ligand ANGPT2, ectodomain shedding of TIE1 by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), and increased TIE/PI3K/AKT signaling, followed by nuclear export of the transcription factor FOXO1. These data establish a functional network between lymphedema-associated genes and provide what we believe to be the first molecular mechanism bridging channel function with vascular signaling and intracellular events culminating in transcriptional regulation of genes expressed in LECs. Our study provides insights into the regulation of lymphatic function and molecular pathways involved in human disease.
Collapse
Affiliation(s)
- Jing Du
- Feinberg Cardiovascular and Renal Research Institute
| | - Pan Liu
- Feinberg Cardiovascular and Renal Research Institute
| | - Yalu Zhou
- Feinberg Cardiovascular and Renal Research Institute
| | - Sol Misener
- Feinberg Cardiovascular and Renal Research Institute
| | - Isha Sharma
- Feinberg Cardiovascular and Renal Research Institute
| | - Phoebe Leeaw
- Feinberg Cardiovascular and Renal Research Institute
| | - Benjamin R. Thomson
- Feinberg Cardiovascular and Renal Research Institute
- Department of Ophthalmology, and
| | - Jing Jin
- Feinberg Cardiovascular and Renal Research Institute
- Division of Nephrology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Susan E. Quaggin
- Feinberg Cardiovascular and Renal Research Institute
- Division of Nephrology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
18
|
K. C. R, Patel NR, Shenoy A, Scallan JP, Chiang MY, Galazo MJ, Meadows SM. Zmiz1 is a novel regulator of lymphatic endothelial cell gene expression and function. PLoS One 2024; 19:e0302926. [PMID: 38718095 PMCID: PMC11078365 DOI: 10.1371/journal.pone.0302926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of ZMIZ1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.
Collapse
Affiliation(s)
- Rajan K. C.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Nehal R. Patel
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Anoushka Shenoy
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Joshua P. Scallan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States of America
| | - Mark Y. Chiang
- Department of Internal Medicine, Division of Hematology-Oncology, Medical School, University of Michigan, Ann Arbor, MI, United States of America
| | - Maria J. Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Stryder M. Meadows
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| |
Collapse
|
19
|
Bowman C, Rockson SG. Genetic causes of lymphatic disorders: recent updates on the clinical and molecular aspects of lymphatic disease. Curr Opin Cardiol 2024; 39:170-177. [PMID: 38483006 DOI: 10.1097/hco.0000000000001116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW The lymphatic system facilitates several key functions that limit significant morbidity and mortality. Despite the impact and burden of lymphatic disorders, there are many remaining disorders whose genetic substrate remains unknown. The purpose of this review is to provide an update on the genetic causes of lymphatic disorders, while reporting on newly proposed clinical classifications of lymphatic disease. RECENT FINDINGS We reviewed several new mutations in genes that have been identified as potential causes of lymphatic disorders including: MDFIC, EPHB 4 , and ANGPT2. Furthermore, the traditional St. George's Classification system for primary lymphatic anomalies has been updated to reflect the use of genetic testing, both as a tool for the clinical identification of lymphatic disease and as a method through which new sub-classifications of lymphatic disorders have been established within this framework. Finally, we highlighted recent clinical studies that have explored the impact of therapies such as sirolimus, ketoprofen, and acebilustat on lymphatic disorders. SUMMARY Despite a growing body of evidence, current literature demonstrates a persistent gap in the number of known genes responsible for lymphatic disease entities. Recent clinical classification tools have been introduced in order to integrate traditional symptom- and time-based diagnostic approaches with modern genetic classifications, as highlighted in the updated St. George's classification system. With the introduction of this novel approach, clinicians may be better equipped to recognize established disease and, potentially, to identify novel causal mutations. Further research is needed to identify additional genetic causes of disease and to optimize current clinical tools for diagnosis and treatment.
Collapse
Affiliation(s)
- Catharine Bowman
- Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
20
|
Panara V, Yu H, Peng D, Staxäng K, Hodik M, Filipek-Gorniok B, Kazenwadel J, Skoczylas R, Mason E, Allalou A, Harvey NL, Haitina T, Hogan BM, Koltowska K. Multiple cis-regulatory elements control prox1a expression in distinct lymphatic vascular beds. Development 2024; 151:dev202525. [PMID: 38722096 PMCID: PMC11128278 DOI: 10.1242/dev.202525] [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: 11/07/2023] [Accepted: 03/21/2024] [Indexed: 05/15/2024]
Abstract
During embryonic development, lymphatic endothelial cell (LEC) precursors are distinguished from blood endothelial cells by the expression of Prospero-related homeobox 1 (Prox1), which is essential for lymphatic vasculature formation in mouse and zebrafish. Prox1 expression initiation precedes LEC sprouting and migration, serving as the marker of specified LECs. Despite its crucial role in lymphatic development, Prox1 upstream regulation in LECs remains to be uncovered. SOX18 and COUP-TFII are thought to regulate Prox1 in mice by binding its promoter region. However, the specific regulation of Prox1 expression in LECs remains to be studied in detail. Here, we used evolutionary conservation and chromatin accessibility to identify enhancers located in the proximity of zebrafish prox1a active in developing LECs. We confirmed the functional role of the identified sequences through CRISPR/Cas9 mutagenesis of a lymphatic valve enhancer. The deletion of this region results in impaired valve morphology and function. Overall, our results reveal an intricate control of prox1a expression through a collection of enhancers. Ray-finned fish-specific distal enhancers drive pan-lymphatic expression, whereas vertebrate-conserved proximal enhancers refine expression in functionally distinct subsets of lymphatic endothelium.
Collapse
Affiliation(s)
- Virginia Panara
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden
- Beijer Gene and Neuro Laboratory, Uppsala University, Uppsala 75185, Sweden
| | - Hujun Yu
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology and Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Karin Staxäng
- BioVis Core Facility, Platform EM, Uppsala University, Uppsala 75185, Sweden
| | - Monika Hodik
- BioVis Core Facility, Platform EM, Uppsala University, Uppsala 75185, Sweden
| | - Beata Filipek-Gorniok
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Jan Kazenwadel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5001, Australia
| | - Renae Skoczylas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Elizabeth Mason
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Amin Allalou
- Uppsala University, Department of Information Technology, Division of Visual Information and Interaction, and SciLifeLab BioImage Informatics Facility, Uppsala University, Uppsala 75185, Sweden
| | - Natasha L. Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5001, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, Uppsala 75236, Sweden
| | - Benjamin M. Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology and Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden
- Beijer Gene and Neuro Laboratory, Uppsala University, Uppsala 75185, Sweden
| |
Collapse
|
21
|
Garlisi Torales LD, Sempowski BA, Krikorian GL, Woodis KM, Paulissen SM, Smith CL, Sheppard SE. Central conducting lymphatic anomaly: from bench to bedside. J Clin Invest 2024; 134:e172839. [PMID: 38618951 PMCID: PMC11014661 DOI: 10.1172/jci172839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Central conducting lymphatic anomaly (CCLA) is a complex lymphatic anomaly characterized by abnormalities of the central lymphatics and may present with nonimmune fetal hydrops, chylothorax, chylous ascites, or lymphedema. CCLA has historically been difficult to diagnose and treat; however, recent advances in imaging, such as dynamic contrast magnetic resonance lymphangiography, and in genomics, such as deep sequencing and utilization of cell-free DNA, have improved diagnosis and refined both genotype and phenotype. Furthermore, in vitro and in vivo models have confirmed genetic causes of CCLA, defined the underlying pathogenesis, and facilitated personalized medicine to improve outcomes. Basic, translational, and clinical science are essential for a bedside-to-bench and back approach for CCLA.
Collapse
Affiliation(s)
- Luciana Daniela Garlisi Torales
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Benjamin A. Sempowski
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Georgia L. Krikorian
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Kristina M. Woodis
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Scott M. Paulissen
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Christopher L. Smith
- Division of Cardiology, Jill and Mark Fishman Center for Lymphatic Disorders, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sarah E. Sheppard
- Unit on Vascular Malformations, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Feng J, Ren Y, Wang X, Li X, Zhu X, Zhang B, Zhao Q, Sun X, Tian X, Liu H, Dong F, Li XL, Qi L, Wei B. Impaired meningeal lymphatic drainage in Listeria monocytogenes infection. Front Immunol 2024; 15:1382971. [PMID: 38638427 PMCID: PMC11024298 DOI: 10.3389/fimmu.2024.1382971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
Previous studies have demonstrated an association between lymphatic vessels and diseases caused by bacterial infections. Listeria monocytogenes (LM) bacterial infection can affect multiple organs, including the intestine, brain, liver and spleen, which can be fatal. However, the impacts of LM infection on morphological and functional changes of lymphatic vessels remain unexplored. In this study, we found that LM infection not only induces meningeal and mesenteric lymphangiogenesis in mice, but also impairs meningeal lymphatic vessels (MLVs)-mediated macromolecules drainage. Interestingly, we found that the genes associated with lymphatic vessel development and function, such as Gata2 and Foxc2, were downregulated, suggesting that LM infection may affect cellular polarization and valve development. On the other hand, photodynamic ablation of MLVs exacerbated inflammation and bacterial load in the brain of mice with LM infection. Overall, our findings indicate that LM infection induces lymphangiogenesis and may affect cell polarization, cavity formation, and valve development during lymphangiogenesis, ultimately impairing MLVs drainage.
Collapse
Affiliation(s)
- Jian Feng
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yuanzhen Ren
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Xilin Wang
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Xiaojing Li
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Xingguo Zhu
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Baokai Zhang
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Qi Zhao
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaochen Sun
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Xinxin Tian
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Hongyang Liu
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fan Dong
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiu-Li Li
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Linlin Qi
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Bin Wei
- Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| |
Collapse
|
23
|
Osinski V, Yellamilli A, Firulyova MM, Zhang MJ, Peck A, Auger JL, Faragher JL, Marath A, Voeller RK, O’Connell TD, Zaitsev K, Binstadt BA. Profibrotic VEGFR3-Dependent Lymphatic Vessel Growth in Autoimmune Valvular Carditis. Arterioscler Thromb Vasc Biol 2024; 44:807-821. [PMID: 38269589 PMCID: PMC10978259 DOI: 10.1161/atvbaha.123.320326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.
Collapse
Affiliation(s)
- Victoria Osinski
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Amritha Yellamilli
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alyssa Peck
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jennifer L. Auger
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jessica L. Faragher
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | | | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Bryce A. Binstadt
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
24
|
Chen Y, He X, Cai J, Li Q. Functional aspects of the brain lymphatic drainage system in aging and neurodegenerative diseases. J Biomed Res 2024; 38:206-221. [PMID: 38430054 PMCID: PMC11144931 DOI: 10.7555/jbr.37.20230264] [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: 10/31/2023] [Revised: 12/30/2023] [Accepted: 02/29/2024] [Indexed: 03/03/2024] Open
Abstract
The phenomenon of an aging population is advancing at a precipitous rate. Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common age-associated neurodegenerative diseases, both of which are primarily characterized by the accumulation of toxic proteins and the progressive demise of neuronal structures. Recent discoveries about the brain lymphatic drainage system have precipitated a growing body of investigations substantiating its novel roles, including the clearance of macromolecular waste and the trafficking of immune cells. Notably, aquaporin 4-mediated glymphatic transport, crucial for maintaining neural homeostasis, becomes disrupted during the aging process and is further compromised in the pathogenesis of AD and PD. Functional meningeal lymphatic vessels, which facilitate the drainage of cerebrospinal fluid into the deep cervical lymph nodes, are integral in bridging the central nervous system with peripheral immune responses. Dysfunction in these meningeal lymphatic vessels exacerbates pathological trajectory of the age-related neurodegenerative disease. This review explores modulatory influence of the glymphatic system and meningeal lymphatic vessels on the aging brain and its associated neurodegenerative disorders. It also encapsulates the insights of potential mechanisms and prospects of the targeted non-pharmacological interventions.
Collapse
Affiliation(s)
- Yan Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Shandong Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiaoxin He
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiachen Cai
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qian Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
25
|
Gu X, Chen X, Zhang X, Liu K, Li JJ, Lv W, Zeng L, Wu M, Zhou W, Wang W, Shi S, Deng Y, Li Y, Gao X, Ju R, Dubrac A, Liu X, Zhang F. Macrophage-induced integrin signaling promotes Schlemm's canal formation to prevent intraocular hypertension and glaucomatous optic neuropathy. Cell Rep 2024; 43:113799. [PMID: 38367239 DOI: 10.1016/j.celrep.2024.113799] [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: 05/15/2023] [Revised: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/19/2024] Open
Abstract
Schlemm's canal (SC) functions to maintain proper intraocular pressure (IOP) by draining aqueous humor and has emerged as a promising therapeutic target for glaucoma, the second-leading cause of irreversible blindness worldwide. However, our current understanding of the mechanisms governing SC development and functionality remains limited. Here, we show that vitronectin (VTN) produced by limbal macrophages promotes SC formation and prevents intraocular hypertension by activating integrin αvβ3 signaling. Genetic inactivation of this signaling system inhibited the phosphorylation of AKT and FOXO1 and reduced β-catenin activity and FOXC2 expression, thereby causing impaired Prox1 expression and deteriorated SC morphogenesis. This ultimately led to increased IOP and glaucomatous optic neuropathy. Intriguingly, we found that aged SC displayed downregulated integrin β3 in association with dampened Prox1 expression. Conversely, FOXO1 inhibition rejuvenated the aged SC by inducing Prox1 expression and SC regrowth, highlighting a possible strategy by targeting VTN/integrin αvβ3 signaling to improve SC functionality.
Collapse
Affiliation(s)
- Xinyu Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Keli Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jing-Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Wenyu Lv
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Lei Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Mingjuan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Weibin Zhou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Weifa Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Shunhua Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yicheng Deng
- School of Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yunhua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xinbo Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada; Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| |
Collapse
|
26
|
Davis MJ, Zawieja SD, Yang Y. Developmental progression of lymphatic valve morphology and function. Front Cell Dev Biol 2024; 12:1331291. [PMID: 38450249 PMCID: PMC10915029 DOI: 10.3389/fcell.2024.1331291] [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: 10/31/2023] [Accepted: 01/12/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction: The bileaflet valves found in collecting lymphatic vessels and some veins are essential for maintaining a unidirectional flow, which is important for lymphatic and venous function. Under an adverse pressure gradient, the two leaflets tightly overlap to prevent backflow. Valves are proposed to share four main stages of development, based on images obtained from randomly oriented valves in fixed mouse embryos, with the best structural views obtained from larger venous valves. It is not known at what stage lymphatic valves (LVs) become functional (e.g., able to oppose backflow), although a requirement for stage 4 is presumed. Methods: To gain an insight into this sequence of events for LVs, we used Prox1CreER T2 :Foxo1 fl/fl mice and Foxc2CreER T2 :Foxo1 fl/fl mouse models, in which deletion of the valve repressor factor Foxo1 promotes the development of new LVs in adult lymphatic vessels. Both strains also contained a Prox1eGFP reporter to image the lymphatic endothelium. Mesenteric collecting lymphatic vessels were dissected, cannulated, and pressurized for ex vivo tests of valve function. LVs at various stages (1-4 and intermediate) were identified in multi-valve segments, which were subsequently shortened to perform the backleak test on single valves. The GFP signal was then imaged at high magnification using a confocal microscope. Z-stack reconstructions enabled 1:1 comparisons of LV morphology with a quantitative measurement of back leak. Results: As expected, LVs of stages 1-3 were completely leaky in response to outflow pressure elevation. Stage 4 valves were generally not leaky, but valve integrity depended on the Cre line used to induce new valve formation. A high percentage of valves at leaflet an intermediate stage (3.5), in which there was an insertion of a second commissure, but without proper luminal alignment, effectively resisted back leak when the outflow pressure was increased. Discussion: Our findings represent the first 3D images of developing lymphatic valves and indicate that valves become competent between stages 3 and 4 of development.
Collapse
Affiliation(s)
- Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States
| | - Scott D. Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
27
|
Dong H, Dai X, Zhou Y, Shi C, Bhuiyan P, Sun Z, Li N, Jin W. Enhanced meningeal lymphatic drainage ameliorates lipopolysaccharide-induced brain injury in aged mice. J Neuroinflammation 2024; 21:36. [PMID: 38287311 PMCID: PMC10826026 DOI: 10.1186/s12974-024-03028-4] [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: 09/16/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is an acute cerebral dysfunction caused by sepsis. Neuroinflammation induced by sepsis is considered a potential mechanism of SAE; however, very little is known about the role of the meningeal lymphatic system in SAE. METHODS Sepsis was established in male C57BL/6J mice by intraperitoneal injection of 5 mg/kg lipopolysaccharide, and the function of meningeal lymphatic drainage was assessed. Adeno-associated virus 1-vascular endothelial growth factor C (AAV1-VEGF-C) was injected into the cisterna magna to induce meningeal lymphangiogenesis. Ligation of deep cervical lymph nodes (dCLNs) was performed to induce pre-existing meningeal lymphatic dysfunction. Cognitive function was evaluated by a fear conditioning test, and inflammatory factors were detected by enzyme-linked immunosorbent assay. RESULTS The aged mice with SAE showed a significant decrease in the drainage of OVA-647 into the dCLNs and the coverage of the Lyve-1 in the meningeal lymphatic, indicating that sepsis impaired meningeal lymphatic drainage and morphology. The meningeal lymphatic function of aged mice was more vulnerable to sepsis in comparison to young mice. Sepsis also decreased the protein levels of caspase-3 and PSD95, which was accompanied by reductions in the activity of hippocampal neurons. Microglia were significantly activated in the hippocampus of SAE mice, which was accompanied by an increase in neuroinflammation, as indicated by increases in interleukin-1 beta, interleukin-6 and Iba1 expression. Cognitive function was impaired in aged mice with SAE. However, the injection of AAV1-VEGF-C significantly increased coverage in the lymphatic system and tracer dye uptake in dCLNs, suggesting that AAV1-VEGF-C promotes meningeal lymphangiogenesis and drainage. Furthermore, AAV1-VEGF-C reduced microglial activation and neuroinflammation and improved cognitive dysfunction. Improvement of meningeal lymphatics also reduced sepsis-induced expression of disease-associated genes in aged mice. Pre-existing lymphatic dysfunction by ligating bilateral dCLNs aggravated sepsis-induced neuroinflammation and cognitive impairment. CONCLUSION The meningeal lymphatic drainage is damaged in sepsis, and pre-existing defects in this drainage system exacerbate SAE-induced neuroinflammation and cognitive dysfunction. Promoting meningeal lymphatic drainage improves SAE. Manipulation of meningeal lymphangiogenesis could be a new strategy for the treatment of SAE.
Collapse
Affiliation(s)
- Hongquan Dong
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaonan Dai
- Department of Obstetrics, Nanjing Women and Children's Healthcare Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Yin Zhou
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chonglong Shi
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Piplu Bhuiyan
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhaochu Sun
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Nana Li
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenjie Jin
- Department of Anesthesiology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
28
|
Hu Z, Zhao X, Wu Z, Qu B, Yuan M, Xing Y, Song Y, Wang Z. Lymphatic vessel: origin, heterogeneity, biological functions, and therapeutic targets. Signal Transduct Target Ther 2024; 9:9. [PMID: 38172098 PMCID: PMC10764842 DOI: 10.1038/s41392-023-01723-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Lymphatic vessels, comprising the secondary circulatory system in human body, play a multifaceted role in maintaining homeostasis among various tissues and organs. They are tasked with a serious of responsibilities, including the regulation of lymph absorption and transport, the orchestration of immune surveillance and responses. Lymphatic vessel development undergoes a series of sophisticated regulatory signaling pathways governing heterogeneous-origin cell populations stepwise to assemble into the highly specialized lymphatic vessel networks. Lymphangiogenesis, as defined by new lymphatic vessels sprouting from preexisting lymphatic vessels/embryonic veins, is the main developmental mechanism underlying the formation and expansion of lymphatic vessel networks in an embryo. However, abnormal lymphangiogenesis could be observed in many pathological conditions and has a close relationship with the development and progression of various diseases. Mechanistic studies have revealed a set of lymphangiogenic factors and cascades that may serve as the potential targets for regulating abnormal lymphangiogenesis, to further modulate the progression of diseases. Actually, an increasing number of clinical trials have demonstrated the promising interventions and showed the feasibility of currently available treatments for future clinical translation. Targeting lymphangiogenic promoters or inhibitors not only directly regulates abnormal lymphangiogenesis, but improves the efficacy of diverse treatments. In conclusion, we present a comprehensive overview of lymphatic vessel development and physiological functions, and describe the critical involvement of abnormal lymphangiogenesis in multiple diseases. Moreover, we summarize the targeting therapeutic values of abnormal lymphangiogenesis, providing novel perspectives for treatment strategy of multiple human diseases.
Collapse
Affiliation(s)
- Zhaoliang Hu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Xushi Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Bicheng Qu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Minxian Yuan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Yanan Xing
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
29
|
Bernier-Latmani J, González-Loyola A, Petrova TV. Mechanisms and functions of intestinal vascular specialization. J Exp Med 2024; 221:e20222008. [PMID: 38051275 PMCID: PMC10697212 DOI: 10.1084/jem.20222008] [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: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The intestinal vasculature has been studied for the last 100 years, and its essential role in absorbing and distributing ingested nutrients is well known. Recently, fascinating new insights into the organization, molecular mechanisms, and functions of intestinal vessels have emerged. These include maintenance of intestinal epithelial cell function, coping with microbiota-induced inflammatory pressure, recruiting gut-specific immune cells, and crosstalk with other organs. Intestinal function is also regulated at the systemic and cellular levels, such that the postprandial hyperemic response can direct up to 30% of systemic blood to gut vessels, while micron-sized endothelial cell fenestrations are necessary for nutrient uptake. In this review, we will highlight past discoveries made about intestinal vasculature in the context of new findings of molecular mechanisms underpinning gut function. Such comprehensive understanding of the system will pave the way to breakthroughs in nutrient uptake optimization, drug delivery efficiency, and treatment of human diseases.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | | | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Subileau M, Vittet D. Ontogenesis of the Mouse Ocular Surface Lymphatic Vascular Network. Invest Ophthalmol Vis Sci 2023; 64:7. [PMID: 38054922 DOI: 10.1167/iovs.64.15.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
Purpose Ocular lymphatic vessels play major physiological role in eye homeostasis and their dysfunction can contribute to the progression of several eye diseases. In this study, we characterized their spatiotemporal development and the cellular mechanisms occurring during their ontogenesis in the mouse eye. Methods Whole mount immunofluorescent staining and imaging by standard or lightsheet fluorescence microscopy were performed on late embryonic and early postnatal eye mouse samples. Results We observed that the ocular surface lymphatic vascular network develops at the early postnatal stages (between P0 and P5) from two nascent trunks arising at the nasal side on both sides of the nictitating membrane. These nascent vessels further branch and encircle the whole eye surface by sprouting lymphangiogenesis. In addition, we got evidence for the existence of a transient lymphvasculogenesis process generating lymphatic vessel fragments that will mostly formed the corneolimbal lymphatic vasculature which further connect to the conjunctival lymphatic network. Our results also support that CD206-positive macrophages can transdifferentiate and then integrate into the lymphatic neovessels. Conclusions Several complementary cellular processes participate in the development of the lymphatic ocular surface vasculature. This knowledge paves the way for the design of new therapeutic strategies to interfere with ocular lymphatic vessel formation when needed.
Collapse
Affiliation(s)
- Mariela Subileau
- University Grenoble Alpes, CEA, Inserm, IRIG, UA13 BGE, Grenoble, France
| | - Daniel Vittet
- University Grenoble Alpes, CEA, Inserm, IRIG, UA13 BGE, Grenoble, France
| |
Collapse
|
31
|
Michalaki E, Surya VN, Rodríguez-Hakim M, Fuller GG, Dunn AR. Response of lymphatic endothelial cells to combined spatial and temporal variations in fluid flow. FASEB J 2023; 37:e23240. [PMID: 37902497 PMCID: PMC11863998 DOI: 10.1096/fj.201902205rrrr] [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: 08/29/2019] [Revised: 09/02/2023] [Accepted: 09/22/2023] [Indexed: 10/31/2023]
Abstract
One-way valves within lymphatic vessels are required for the efficient drainage of lymphatic fluids. Fluid flow is proposed to be a key cue in regulating both the formation and maintenance of lymphatic valves. However, to our knowledge, no previous study has systematically examined the response of LECs to the complex combination of spatially and temporally varying fluid flows that occur at lymphatic valves in vivo. We built an in vitro microfluidic device that reproduces key aspects of the flow environment found at lymphatic valves. Using this device, we found that a combination of spatially and temporally varying wall shear stresses (WSSs) led to upregulated transcription of PROX1 and FOXC2. In addition, we observed that combined spatial and temporal variations in WSS-modulated Ca2+ signaling and led to increased cellular levels of NFATc1. These observations suggest that the physical cues generated by the flow environment present within lymphatic valves may act to activate key regulatory pathways that contribute to valve maintenance.
Collapse
Affiliation(s)
- Eleftheria Michalaki
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Vinay N. Surya
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | | | - Gerald G. Fuller
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Alexander R. Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
32
|
Chen D, Wiggins D, Sevick EM, Davis MJ, King PD. An EPHB4-RASA1 signaling complex inhibits shear stress-induced Ras-MAPK activation in lymphatic endothelial cells to promote the development of lymphatic vessel valves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568378. [PMID: 38045382 PMCID: PMC10690291 DOI: 10.1101/2023.11.22.568378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
EPHB4 is a receptor protein tyrosine kinase that is required for the development of lymphatic vessel (LV) valves. We show here that EPHB4 is necessary for the specification of LV valves, their continued development after specification, and the maintenance of LV valves in adult mice. EPHB4 promotes LV valve development by inhibiting the activation of the Ras-MAPK pathway in LV endothelial cells (LEC). For LV specification, this role for EPHB4 depends on its ability to interact physically with the p120 Ras-GTPase-activating protein (RASA1) that acts as a negative regulator of Ras. Through physical interaction, EPHB4 and RASA1 dampen oscillatory shear stress (OSS)-induced Ras-MAPK activation in LEC, which is required for LV specification. We identify the Piezo1 OSS sensor as a focus of EPHB4-RASA1 regulation of OSS-induced Ras-MAPK signaling mediated through physical interaction. These findings contribute to an understanding of the mechanism by which EPHB4, RASA1 and Ras regulate lymphatic valvulogenesis.
Collapse
|
33
|
Shin M, Yin HM, Shih YH, Nozaki T, Portman D, Toles B, Kolb A, Luk K, Isogai S, Ishida K, Hanasaka T, Parsons MJ, Wolfe SA, Burns CE, Burns CG, Lawson ND. Generation and application of endogenously floxed alleles for cell-specific knockout in zebrafish. Dev Cell 2023; 58:2614-2626.e7. [PMID: 37633272 PMCID: PMC10840978 DOI: 10.1016/j.devcel.2023.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/30/2023] [Accepted: 07/28/2023] [Indexed: 08/28/2023]
Abstract
The zebrafish is amenable to a variety of genetic approaches. However, lack of conditional deletion alleles limits stage- or cell-specific gene knockout. Here, we applied an existing protocol to establish a floxed allele for gata2a but failed to do so due to off-target integration and incomplete knockin. To address these problems, we applied simultaneous co-targeting with Cas12a to insert loxP sites in cis, together with transgenic counterscreening and comprehensive molecular analysis, to identify off-target insertions and confirm targeted knockins. We subsequently used our approach to establish endogenously floxed alleles of foxc1a, rasa1a, and ruvbl1, each in a single generation. We demonstrate the utility of these alleles by verifying Cre-dependent deletion, which yielded expected phenotypes in each case. Finally, we used the floxed gata2a allele to demonstrate an endothelial autonomous requirement in lymphatic valve development. Together, our results provide a framework for routine generation and application of endogenously floxed alleles in zebrafish.
Collapse
Affiliation(s)
- Masahiro Shin
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hui-Min Yin
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Huan Shih
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Takayuki Nozaki
- Technical Support Center for Life Science Research, Iwate Medical University, Shiwa, Iwate 028-3694, Japan
| | - Daneal Portman
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Benjamin Toles
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Amy Kolb
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kevin Luk
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sumio Isogai
- Department of Medical Education, Iwate Medical University, Shiwa, Iwate 028-3694, Japan
| | - Kinji Ishida
- Technical Support Center for Life Science Research, Iwate Medical University, Shiwa, Iwate 028-3694, Japan
| | - Tomohito Hanasaka
- Technical Support Center for Life Science Research, Iwate Medical University, Shiwa, Iwate 028-3694, Japan
| | - Michael J Parsons
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Scot A Wolfe
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
34
|
Saha S, Fan F, Alderfer L, Graham F, Hall E, Hanjaya-Putra D. Synthetic hyaluronic acid coating preserves the phenotypes of lymphatic endothelial cells. Biomater Sci 2023; 11:7346-7357. [PMID: 37789798 PMCID: PMC10628678 DOI: 10.1039/d3bm00873h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/14/2023] [Indexed: 10/05/2023]
Abstract
Lymphatic endothelial cells (LECs) play a critical role in the formation and maintenance of the lymphatic vasculature, which is essential for the immune system, fluid balance, and tissue repair. However, LECs are often difficult to study in vivo and in vitro models that accurately mimic their behaviors and phenotypes are limited. In particular, LECs have been shown to lose their lymphatic markers over time while being cultured in vitro, which reflect their plasticity and heterogeneity in vivo. Since LECs uniquely express lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), we hypothesized that surface coating with hyaluronic acid (HA) can preserve LEC phenotypes and functionalities. Dopamine conjugated hyaluronic acid (HA-DP) was synthesized with 42% degree of substitution to enable surface modification and conjugation onto standard tissue culture plates. Compared to fibronectin coating and tissue culture plate controls, surface coating with HA-DP was able to preserve lymphatic markers, such as prospero homeobox protein 1 (Prox1), podoplanin (PDPN), and LYVE-1 over several passages in vitro. LECs cultured on HA-DP expressed lower levels of focal adhesion kinase (FAK) and YAP/TAZ, which may be responsible for the maintenance of the lymphatic characteristics. Collectively, the HA-DP coating may provide a novel method for culturing human LECs in vitro toward more representative studies in basic lymphatic biology and lymphatic regeneration.
Collapse
Affiliation(s)
- Sanjoy Saha
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Fei Fan
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Laura Alderfer
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Francine Graham
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
| | - Eva Hall
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
| | - Donny Hanjaya-Putra
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, IN 46556, USA.
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
| |
Collapse
|
35
|
Iyer D, Mastrogiacomo DM, Li K, Banerjee R, Yang Y, Scallan JP. eNOS Regulates Lymphatic Valve Specification by Controlling β-Catenin Signaling During Embryogenesis in Mice. Arterioscler Thromb Vasc Biol 2023; 43:2197-2212. [PMID: 37767708 PMCID: PMC10655861 DOI: 10.1161/atvbaha.123.319405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Lymphatic valves play a critical role in ensuring unidirectional lymph transport. Loss of lymphatic valves or dysfunctional valves are associated with several diseases including lymphedema, lymphatic malformations, obesity, and ileitis. Lymphatic valves first develop during embryogenesis in response to mechanotransduction signaling pathways triggered by oscillatory lymph flow. In blood vessels, eNOS (endothelial NO synthase; gene name: Nos3) is a well-characterized shear stress signaling effector, but its role in lymphatic valve development remains unexplored. METHODS We used global Nos3-/- mice and cultured human dermal lymphatic endothelial cells to investigate the role of eNOS in lymphatic valve development, which requires oscillatory shear stress signaling. RESULTS Our data reveal a 45% reduction in lymphatic valve specification cell clusters and that loss of eNOS protein inhibited activation of β-catenin and its nuclear translocation. Genetic knockout or knockdown of eNOS led to downregulation of β-catenin target proteins in vivo and in vitro. However, pharmacological inhibition of NO production did not reproduce these effects. Co-immunoprecipitation and proximity ligation assays reveal that eNOS directly binds to β-catenin and their binding is enhanced by oscillatory shear stress. Finally, genetic ablation of the Foxo1 gene enhanced FOXC2 expression and partially rescued the loss of valve specification in the eNOS knockouts. CONCLUSIONS In conclusion, we demonstrate a novel, NO-independent role for eNOS in regulating lymphatic valve specification and propose a mechanism by which eNOS directly binds β-catenin to regulate its nuclear translocation and thereby transcriptional activity.
Collapse
Affiliation(s)
- Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Diandra M Mastrogiacomo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Kunyu Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
36
|
Meng Y, Lv T, Zhang J, Shen W, Li L, Li Y, Liu X, Lei X, Lin X, Xu H, Meng A, Jia S. Temporospatial inhibition of Erk signaling is required for lymphatic valve formation. Signal Transduct Target Ther 2023; 8:342. [PMID: 37691058 PMCID: PMC10493226 DOI: 10.1038/s41392-023-01571-9] [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] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 09/12/2023] Open
Abstract
Intraluminal lymphatic valves (LVs) and lymphovenous valves (LVVs) are critical to ensure the unidirectional flow of lymphatic fluid. Morphological abnormalities in these valves always cause lymph or blood reflux, and result in lymphedema. However, the underlying molecular mechanism of valve development remains poorly understood. We here report the implication of Efnb2-Ephb4-Rasa1 regulated Erk signaling axis in lymphatic valve development with identification of two new valve structures. Dynamic monitoring of phospho-Erk activity indicated that Erk signaling is spatiotemporally inhibited in some lymphatic endothelial cells (LECs) during the valve cell specification. Inhibition of Erk signaling via simultaneous depletion of zygotic erk1 and erk2 or treatment with MEK inhibitor selumetinib causes lymphatic vessel hypoplasia and lymphatic valve hyperplasia, suggesting opposite roles of Erk signaling during these two processes. ephb4b mutants, efnb2a;efnb2b or rasa1a;rasa1b double mutants all have defective LVs and LVVs and exhibit blood reflux into lymphatic vessels with an edema phenotype. Importantly, the valve defects in ephb4b or rasa1a;rasa1b mutants are mitigated with high-level gata2 expression in the presence of MEK inhibitors. Therefore, Efnb2-Ephb4 signaling acts to suppress Erk activation in valve-forming cells to promote valve specification upstream of Rasa1. Not only do our findings reveal a molecular mechanism of lymphatic valve formation, but also provide a basis for the treatment of lymphatic disorders.
Collapse
Affiliation(s)
- Yaping Meng
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tong Lv
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junfeng Zhang
- Guangzhou Laboratory, Guangzhou, 510320, Guangdong Province, China
| | - Weimin Shen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lifang Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yaqi Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xin Liu
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xing Lei
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuguang Lin
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Hanfang Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Anming Meng
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Guangzhou Laboratory, Guangzhou, 510320, Guangdong Province, China.
| | - Shunji Jia
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
37
|
Ho YC, Geng X, O’Donnell A, Ibarrola J, Fernandez-Celis A, Varshney R, Subramani K, Azartash-Namin ZJ, Kim J, Silasi R, Wylie-Sears J, Alvandi Z, Chen L, Cha B, Chen H, Xia L, Zhou B, Lupu F, Burkhart HM, Aikawa E, Olson LE, Ahamed J, López-Andrés N, Bischoff J, Yutzey KE, Srinivasan RS. PROX1 Inhibits PDGF-B Expression to Prevent Myxomatous Degeneration of Heart Valves. Circ Res 2023; 133:463-480. [PMID: 37555328 PMCID: PMC10487359 DOI: 10.1161/circresaha.123.323027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Cardiac valve disease is observed in 2.5% of the general population and 10% of the elderly people. Effective pharmacological treatments are currently not available, and patients with severe cardiac valve disease require surgery. PROX1 (prospero-related homeobox transcription factor 1) and FOXC2 (Forkhead box C2 transcription factor) are transcription factors that are required for the development of lymphatic and venous valves. We found that PROX1 and FOXC2 are expressed in a subset of valvular endothelial cells (VECs) that are located on the downstream (fibrosa) side of cardiac valves. Whether PROX1 and FOXC2 regulate cardiac valve development and disease is not known. METHODS We used histology, electron microscopy, and echocardiography to investigate the structure and functioning of heart valves from Prox1ΔVEC mice in which Prox1 was conditionally deleted from VECs. Isolated valve endothelial cells and valve interstitial cells were used to identify the molecular mechanisms in vitro, which were tested in vivo by RNAScope, additional mouse models, and pharmacological approaches. The significance of our findings was tested by evaluation of human samples of mitral valve prolapse and aortic valve insufficiency. RESULTS Histological analysis revealed that the aortic and mitral valves of Prox1ΔVEC mice become progressively thick and myxomatous. Echocardiography revealed that the aortic valves of Prox1ΔVEC mice are stenotic. FOXC2 was downregulated and PDGF-B (platelet-derived growth factor-B) was upregulated in the VECs of Prox1ΔVEC mice. Conditional knockdown of FOXC2 and conditional overexpression of PDGF-B in VECs recapitulated the phenotype of Prox1ΔVEC mice. PDGF-B was also increased in mice lacking FOXC2 and in human mitral valve prolapse and insufficient aortic valve samples. Pharmacological inhibition of PDGF-B signaling with imatinib partially ameliorated the valve defects of Prox1ΔVEC mice. CONCLUSIONS PROX1 antagonizes PDGF-B signaling partially via FOXC2 to maintain the extracellular matrix composition and prevent myxomatous degeneration of cardiac valves.
Collapse
Affiliation(s)
- Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
- Now with Sanegene Bio, Woburn, MA (X.G.)
| | - Anna O’Donnell
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (A.O., K.E.Y.)
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (J.I.)
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A., R.S.S.)
| | - Amaya Fernandez-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A., R.S.S.)
| | - Rohan Varshney
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Kumar Subramani
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Zheila J. Azartash-Namin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Jang Kim
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center (J.K.)
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Jill Wylie-Sears
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (J.W.-S., Z.A., H.C., J.B.)
| | - Zahra Alvandi
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (J.W.-S., Z.A., H.C., J.B.)
| | - Lijuan Chen
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
- Now with Daegu Gyeongbuk Medical Innovation Foundation, Republic of Korea (B.C.)
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (J.W.-S., Z.A., H.C., J.B.)
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY (B.Z.)
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Harold M. Burkhart
- Oklahoma Children’s Hospital, University of Oklahoma Health Heart Center, Oklahoma City, OK (H.M.B.)
| | - Elena Aikawa
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA (E.A.)
| | - Lorin E. Olson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A., R.S.S.)
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children's Hospital, Boston, MA (J.W.-S., Z.A., H.C., J.B.)
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (A.O., K.E.Y.)
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK (Y.-C.H., X.G., R.V., K.S., Z.J.A.-N., J.K., R.S., L.C., B.C., L.X., F.L., L.E.O., J.A., R.S.S.)
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A., R.S.S.)
| |
Collapse
|
38
|
Angeli V, Lim HY. Biomechanical control of lymphatic vessel physiology and functions. Cell Mol Immunol 2023; 20:1051-1062. [PMID: 37264249 PMCID: PMC10469203 DOI: 10.1038/s41423-023-01042-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 06/03/2023] Open
Abstract
The ever-growing research on lymphatic biology has clearly identified lymphatic vessels as key players that maintain human health through their functional roles in tissue fluid homeostasis, immunosurveillance, lipid metabolism and inflammation. It is therefore not surprising that the list of human diseases associated with lymphatic malfunctions has grown larger, including issues beyond lymphedema, a pathology traditionally associated with lymphatic drainage insufficiency. Thus, the discovery of factors and pathways that can promote optimal lymphatic functions may offer new therapeutic options. Accumulating evidence indicates that aside from biochemical factors, biomechanical signals also regulate lymphatic vessel expansion and functions postnatally. Here, we review how mechanical forces induced by fluid shear stress affect the behavior and functions of lymphatic vessels and the mechanisms lymphatic vessels employ to sense and transduce these mechanical cues into biological signals.
Collapse
Affiliation(s)
- Veronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
39
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
40
|
O’Donnell A, Gonzalez BA, Mukherjee S, Wilson R, Alfieri CM, Swoboda CO, Millay DP, Zorn AM, Yutzey KE. Localized Prox1 Regulates Aortic Valve Endothelial Cell Diversity and Extracellular Matrix Stratification in Mice. Arterioscler Thromb Vasc Biol 2023; 43:1478-1493. [PMID: 37381982 PMCID: PMC10528305 DOI: 10.1161/atvbaha.123.319424] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Specialized valve endothelial cell (VEC) populations are localized oriented to blood flow in developing aortic and mitral valves, but their roles in valve development and disease are unknown. In the aortic valve (AoV), a population of VECs on the fibrosa side expresses the transcription factor Prox1 together with genes found in lymphatic ECs. In this study, we examine Prox1's role in regulating a lymphatic-like gene network and promoting VEC diversity required for the development of the stratified trilaminar extracellular matrix (ECM) of murine AoV leaflets. METHODS To determine whether disruption of Prox1 localization affects heart valve development, we generated mice (NFATc1enCre Prox1 gain-of-function) in which Prox1 is overexpressed on the ventricularis side of the AoV beginning in embryonic development. To identify potential targets of Prox1, we performed cleavage under targets and release using nuclease on wild-type and NFATc1enCre Prox1 gain-of-function AoVs with validation by colocalization in vivo using RNA in situ hybridization in NFATc1enCre Prox1 gain-of-function AoVs. Natural induction of Prox1 and target gene expression was evaluated in myxomatous AoVs in a mouse model of Marfan syndrome (Fbn1C1039G/+). RESULTS The overexpression of Prox1 is sufficient to cause enlargement of AoVs by postnatal day (P)0, as well as a decrease in ventricularis-specific gene expression and disorganized interstitial ECM layers at P7. We identified potential targets of Prox1 known to play roles in lymphatic ECs including Flt1, Efnb2, Egfl7, and Cx37. Ectopic Prox1 colocalized with induced Flt1, Efnb2, and Cx37 expression in NFATc1enCre Prox1 gain-of-function AoVs. Moreover, in Marfan syndrome myxomatous AoVs, endogenous Prox1, and its identified targets, were ectopically induced in ventricularis side VECs. CONCLUSIONS Our results support a role for Prox1 in localized lymphatic-like gene expression on the fibrosa side of the AoV. Furthermore, localized VEC specialization is required for development of the stratified trilaminar ECM critical for AoV function and is dysregulated in congenitally malformed valves.
Collapse
Affiliation(s)
- Anna O’Donnell
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Brittany A. Gonzalez
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Shreyasi Mukherjee
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ruby Wilson
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Christina M. Alfieri
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O. Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Douglas P. Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Aaron M. Zorn
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
41
|
Hoofnagle MH, Hess A, Nalugo M, Ghosh S, Hughes SW, Fuchs A, Welsh JD, Kahn ML, Bochicchio GV, Randolph GJ, Leonard JM, Turnbull IR. Defects in vein valve PROX1/FOXC2 antithrombotic pathway in endothelial cells drive the hypercoagulable state induced by trauma and critical illness. J Trauma Acute Care Surg 2023; 95:197-204. [PMID: 37072887 PMCID: PMC10524206 DOI: 10.1097/ta.0000000000003945] [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: 04/20/2023]
Abstract
OBJECTIVES Deep venous thrombosis (DVT) causes significant morbidity and mortality after trauma. Recently, we have shown that blood flow patterns at vein valves induce oscillatory stress genes, which maintain an anticoagulant endothelial phenotype that inhibits spontaneous clotting at vein valves and sinuses, is lost in the presence of DVT in human pathological samples, and is dependent on expression of the transcription factor FOXC2. We describe an assay, modifying our mouse multiple injury system, which shows evidence of clinically relevant microthrombosis and hypercoagulability applicable to the study of spontaneous DVT in trauma without requiring direct vascular injury or ligation. Finally, we investigated whether these model findings are relevant to a human model of critical illness by examining gene expression changes by quantitative polymerase chain reaction and immunofluorescence in veins collected from critically ill. METHODS C57/Bl6 mice were subjected to a modified mouse multiple injury model with liver crush injury, crush and pseudofracture of a single lower extremity, and 15% total blood volume hemorrhage. Serum was assayed for d-dimer at 2, 6, 24, and 48 hours after injury by enzyme-linked immunosorbent assay. For the thrombin clotting assay, veins of the leg were exposed, 100 μL of 1 mM rhodamine (6 g) was injected retro-orbitally, and 450 μg/mL thrombin was then applied to the surface of the vein with examination of real-time clot formation via in vivo immunofluorescence microscopy. Images were then examined for percentage area of clot coverage of visible mouse saphenous and common femoral vein. Vein valve specific knockout of FOXC2 was induced with tamoxifen treatment in PROX1 Ert2Cre FOXC2 fl/fl mice as previously described. Animals were then subjected to a modified mouse multiple injury model with liver crush injury, crush and pseudofracture of a single lower extremity, and 15% total blood volume hemorrhage. Twenty-four hours after injury, we examined the valve phenotype in naive versus multiple injury animals, with and without loss of the FOXC2 gene from the vein valve (FOXC2 del ) via the thrombin assay. Images were then examined for proximity of clot formation to the valve present at the junction of the mouse saphenous, tibial, and superficial femoral vein and presence of spontaneous microthrombi present in the veins before exposure to thrombin. Human vein samples were obtained from excess tissue preserved after harvest for elective cardiac surgery and from organ donors after organ procurement. Sections were submitted for paraffin embedding and then assayed by immunofluorescence for PROX1, FOXC2, thrombomodulin, endothelial protein C receptor, and von Willebrand's factor. All animal studies were reviewed and approved by the Institutional Animal Care and Use Committee, and all human studies reviewed and approved by the institutional review board. RESULTS After mouse multiple injuries, enzyme-linked immunosorbent assay for d-dimer showed evidence of products of fibrin breakdown consistent with formation of clot related to injury, fibrinolysis, and/or microthrombosis. The thrombin clotting assay demonstrated higher percentage area of vein covered with clot when exposed to thrombin in the multiple injury animals compared with uninjured (45% vs. 27% p = 0.0002) consistent with a phenotype of hypercoagulable state after trauma in our model system. Unmanipulated FoxC2 knockout mice manifest increased clotting at the vein valve as compared with unmanipulated wild type animals. After multiple injuries, wild type mice manifest increase clotting at the vein after thrombin exposure ( p = 0.0033), and equivalent to that of valvular knockout of FoxC2 (FoxC2del), recapitulating the phenotype seen in FoxC2 knockout animals. The combination of multiple injuries and FoxC2 knockout resulted in spontaneous microthrombi in 50% of the animals, a phenotype not observed with either multiple injuries or FoxC2 deficiency alone (χ 2 , p = 0.017). Finally, human vein samples demonstrated the protective vein valve phenotype of increased FOXC2 and PROX1 and showed decreased expression in the critically ill organ donor population by immunofluorescence imaging in organ donor samples. CONCLUSION We have established a novel model of posttrauma hypercoagulation that does not require direct restriction of venous flow or direct injury to the vessel endothelium to assay for hypercoagulability and can generate spontaneous microthrombosis when combined with valve-specific FOXC2 knockout. We find that multiple injuries induce a procoagulant phenotype that recapitulates the valvular hypercoagulability seen in FOXC2 knockout and, in critically ill human specimens, find evidence for loss of oscillatory shear stress-induced gene expression of FOXC2 and PROX1 in the valvular endothelium consistent with potential loss of DVT-protective valvular phenotype.
Collapse
Affiliation(s)
- Mark H Hoofnagle
- From the Section of Acute and Critical Care Surgery, Department of Surgery (M.H.H., A.H., S.G., S.-W.H., A.F., G.V.B., J.M.L., I.R.T.), Section of Vascular Surgery, Department of Surgery (M.N.), and Department of Pathology (G.J.R.), School of Medicine, Washington University in Saint Louis, St. Louis, Missouri; Osciflex LLC (J.D.W.); and Department of Medicine (M.L.K.), Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rajan KC, Patel NR, Shenoy A, Scallan JP, Chiang MY, Galazo MJ, Meadows SM. Zmiz1 is a novel regulator of lymphatic endothelial cell gene expression and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.22.550165. [PMID: 37503058 PMCID: PMC10370198 DOI: 10.1101/2023.07.22.550165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of Zmiz1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.
Collapse
Affiliation(s)
- K C Rajan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Nehal R Patel
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Anoushka Shenoy
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Joshua P Scallan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mark Y Chiang
- Division of Hematology-Oncology, Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI
| | - Maria J Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Stryder M Meadows
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
43
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Scallan JP, Yang Y. Rictor induces AKT signaling to regulate lymphatic valve formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544698. [PMID: 37397997 PMCID: PMC10312634 DOI: 10.1101/2023.06.12.544698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lymphatic valves are specialized structures of the collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress (OSS) from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves throughout life. Conventionally, in other tissue types, AKT activation requires dual kinase activity and the mammalian target of rapamycin complex 2 (mTORC2) commands this process by phosphorylating AKT at Ser473. Here we showed that embryonic and postnatal lymphatic deletion of Rictor , a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human lymphatic endothelial cells (hdLECs) not only significantly reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions, but also abolished the upregulation of AKT activity and valve-forming genes in response to flow. We further showed that the AKT target, FOXO1, a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric LECs, in vivo . Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in both mesenteric and ear lymphatics. Our work revealed a novel role of RICTOR signaling in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately allows the formation and maintenance of a normal lymphatic valve.
Collapse
|
44
|
Saygili Demir C, Sabine A, Gong M, Dormond O, Petrova TV. Mechanosensitive mTORC1 signaling maintains lymphatic valves. J Cell Biol 2023; 222:e202207049. [PMID: 37036444 PMCID: PMC10097975 DOI: 10.1083/jcb.202207049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/26/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
Homeostatic maintenance and repair of lymphatic vessels are essential for health. We investigated the dynamics and the molecular mechanisms of lymphatic endothelial cell (LEC) renewal in adult mesenteric quiescent lymphatic vasculature using label-retention, lineage tracing, and cell ablation strategies. Unlike during development, adult LEC turnover and proliferation was confined to the valve regions of collecting vessels, with valve cells displaying the shortest lifespan. Proliferating valve sinus LECs were the main source for maintenance and repair of lymphatic valves. We identified mechanistic target of rapamycin complex 1 (mTORC1) as a mechanoresponsive pathway activated by fluid shear stress in LECs. Depending on the shear stress level, mTORC1 activity drives division of valve cells or dictates their mechanic resilience through increased protein synthesis. Overactivation of lymphatic mTORC1 in vivo promoted supernumerary valve formation. Our work provides insights into the molecular mechanisms of maintenance of healthy lymphatic vascular system.
Collapse
Affiliation(s)
- Cansaran Saygili Demir
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Amélie Sabine
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Muyun Gong
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
45
|
Abstract
Kidney disease is associated with adverse consequences in many organs beyond the kidney, including the heart, lungs, brain, and intestines. The kidney-intestinal cross talk involves intestinal epithelial damage, dysbiosis, and generation of uremic toxins. Recent studies reveal that kidney injury expands the intestinal lymphatics, increases lymphatic flow, and alters the composition of mesenteric lymph. The intestinal lymphatics, like blood vessels, are a route for transporting potentially harmful substances generated by the intestines. The lymphatic architecture and actions are uniquely suited to take up and transport large macromolecules, functions that differentiate them from blood vessels, allowing them to play a distinct role in a variety of physiological and pathological processes. Here, we focus on the mechanisms by which kidney diseases result in deleterious changes in intestinal lymphatics and consider a novel paradigm of a vicious cycle of detrimental organ cross talk. This concept involves kidney injury-induced modulation of intestinal lymphatics that promotes production and distribution of harmful factors, which in turn contributes to disease progression in distant organ systems.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics (A.K.), Vanderbilt University Medical Center, Nashville, TN
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN (A.K.)
| | - Hai-Chun Yang
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Agnes B. Fogo
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine (A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Elaine L. Shelton
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Valentina Kon
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
46
|
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
|
47
|
Davis MJ, Castorena-Gonzalez JA, Kim HJ, Li M, Remedi M, Nichols CG. Lymphatic contractile dysfunction in mouse models of Cantú Syndrome with K ATP channel gain-of-function. FUNCTION 2023; 4:zqad017. [PMID: 37214333 PMCID: PMC10194823 DOI: 10.1093/function/zqad017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/01/2023] [Accepted: 04/07/2023] [Indexed: 05/24/2023] Open
Abstract
Cantú Syndrome (CS) is an autosomal dominant disorder caused by gain-of-function (GoF) mutations in the Kir6.1 and SUR2 subunits of KATP channels. KATP overactivity results in a chronic reduction in arterial tone and hypotension, leading to other systemic cardiovascular complications. However, the underlying mechanism of lymphedema, developed by >50% of CS patients, is unknown. We investigated whether lymphatic contractile dysfunction occurs in mice expressing CS mutations in Kir6.1 (Kir6.1[V65M]) or SUR2 (SUR2[A478V], SUR2[R1154Q]). Pressure myograph tests of contractile function of popliteal lymphatic vessels over the physiological pressure range revealed significantly impaired contractile strength and reduced frequency of spontaneous contractions at all pressures in heterozygous Kir6.1[V65M] vessels, compared to control littermates. Contractile dysfunction of intact popliteal lymphatics in vivo was confirmed using near-infrared fluorescence microscopy. Homozygous SUR2[A478V] vessels exhibited profound contractile dysfunction ex vivo, but heterozygous SUR2[A478V] vessels showed essentially normal contractile function. However, further investigation of vessels from all three GoF mouse strains revealed significant disruption in contraction wave entrainment, decreased conduction speed and distance, multiple pacemaker sites, and reversing wave direction. Tests of 2-valve lymphatic vessels forced to pump against an adverse pressure gradient revealed that all CS-associated genotypes were essentially incapable of pumping under an imposed outflow load. Our results show that varying degrees of lymphatic contractile dysfunction occur in proportion to the degree of molecular GoF in Kir6.1 or SUR2. This is the first example of lymphatic contractile dysfunction caused by a smooth muscle ion channel mutation and potentially explains the susceptibility of CS patients to lymphedema.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia MO 65212, USA
| | | | - Hae Jin Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia MO 65212, USA
| | - Min Li
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia MO 65212, USA
| | - Maria Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Iyer D, Mastrogiacomo D, Li K, Banerjee R, Yang Y, Scallan JP. Endothelial Nitric Oxide Synthase Regulates Lymphatic Valve Specification By Controlling β - catenin Signaling During Embryogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536303. [PMID: 37090551 PMCID: PMC10120724 DOI: 10.1101/2023.04.10.536303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Objective Lymphatic valves play a critical role in ensuring unidirectional lymph transport. Loss of lymphatic valves or dysfunctional valves are associated with several diseases including lymphedema, lymphatic malformations, obesity, and ileitis. Lymphatic valves first develop during embryogenesis in response to mechanotransduction signaling pathways triggered by oscillatory lymph flow. In blood vessels, eNOS (gene name: Nos3 ) is a well characterized shear stress signaling effector, but its role in lymphatic valve development remains unexplored. Approach and Results We used global Nos3 -/- mice and cultured hdLECs to investigate the role of eNOS in lymphatic valve development, which requires oscillatory shear stress signaling. Our data reveal a 45% reduction in lymphatic valve specification cell clusters and that loss of eNOS protein inhibited activation of β-catenin and its nuclear translocation. Genetic knockout or knockdown of eNOS led to downregulation of β-catenin target proteins in vivo and in vitro . However, pharmacological inhibition of NO production did not reproduce these effects. Coimmunoprecipitation experiments reveal that eNOS forms a complex with β-catenin and their association is enhanced by oscillatory shear stress. Finally, genetic ablation of the Foxo1 gene enhanced FOXC2 expression and rescued the loss of valve specification in the eNOS knockouts. Conclusion In conclusion, we demonstrate a novel, nitric oxide-independent role for eNOS in regulating lymphatic valve specification and propose a mechanism by which eNOS forms a complex with β-catenin to regulate its nuclear translocation and thereby transcriptional activity.
Collapse
Affiliation(s)
- Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Diandra Mastrogiacomo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Kunyu Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| |
Collapse
|
49
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
50
|
Qiu X, Deng Z, Wang M, Feng Y, Bi L, Li L. Piezo protein determines stem cell fate by transmitting mechanical signals. Hum Cell 2023; 36:540-553. [PMID: 36580272 DOI: 10.1007/s13577-022-00853-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Piezo ion channel is a mechanosensitive protein on the cell membrane, which contains Piezo1 and Piezo2. Piezo channels are activated by mechanical forces, including stretch, matrix stiffness, static pressure, and shear stress. Piezo channels transmit mechanical signals that cause different downstream responses in the differentiation process, including integrin signaling pathway, ERK1/2 MAPK signaling pathway, Notch signaling, and WNT signaling pathway. In the fate of stem cell differentiation, scientists found differences in Piezo channel expression and found that Piezo channel expression is related to developmental diseases. Here, we briefly review the structure and function of Piezo channels and the relationship between Piezo and mechanical signals, discussing the current understanding of the role of Piezo channels in stem cell fate and associated molecules and developmental diseases. Ultimately, we believe this review will help identify the association between Piezo channels and stem cell fate.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhuoyue Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Meijing Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yuqi Feng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|