1
|
Tso P, Bernier-Latmani J, Petrova TV, Liu M. Transport functions of intestinal lymphatic vessels. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-00996-z. [PMID: 39496888 DOI: 10.1038/s41575-024-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
2
|
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
|
3
|
Kistner BM, Tian Y, Douglas ES, Caron KM. Cardiac lymphatics undergo distinct remodeling during hypertrophic and nonhypertrophic pregnancy. Am J Physiol Heart Circ Physiol 2024; 327:H1155-H1161. [PMID: 39269453 DOI: 10.1152/ajpheart.00459.2024] [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: 07/08/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024]
Abstract
Lymphatic vessels of the heart undergo dynamic remodeling in response to physiological and pathological cardiovascular events such as development, adult cardiac maintenance, and injury repair. During pregnancy, the cardiovascular system undergoes physiological changes to meet the increased demand for blood supply to the fetus. These extensive physiological changes make pregnancy and delivery a high-risk period in a woman's life. However, whether and how cardiac lymphatics change during pregnancy is largely undefined. Therefore, we used whole mount immunofluorescent labeling and quantitative morphometric analysis to characterize the changes in cardiac lymphatic vasculature during pregnancy using two genetically distinct inbred mouse strains, C57BL/6J and BALB/cJ. When compared with age-matched, nonpregnant C57BL/6J control mice, the hearts of C57BL/6J dams in late pregnancy [gestation day 17.5 (G17.5)] undergo physiological hypertrophy. However, there were no significant changes in the cardiac lymphatic vasculature. In contrast, BALB/cJ mice do not exhibit pregnancy-induced cardiac hypertrophy at G17.5 compared with age-matched, nonpregnant mice. Yet interestingly, the cardiac lymphatic vasculature of pregnant BALB/cJ dams undergoes extensive morphological changes, including decreased lymphatic length, number of end points, and vessel branch-point junctions on the ventral side of the heart. These findings underscore the complexity of genetic and physiological factors that contribute to the heterotypic remodeling of cardiac lymphatics during late pregnancy.NEW & NOTEWORTHY Cardiac lymphatics remodel in response to physiological and pathological stresses. This study is the first to investigate cardiac lymphatic vessel changes during pregnancy. BALB/cJ mice, which do not undergo pregnancy-induced cardiac hypertrophy, show decreased lymphatic length, number of end points, and junctions on the ventral side during pregnancy. In contrast, C57BL/6J mice, which undergo pregnancy-induced cardiac hypertrophy, had no such changes. These findings underscore the complexity of genetic and physiological factors contributing to cardiac lymphatic remodeling.
Collapse
Affiliation(s)
- Bryan M Kistner
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Yanna Tian
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Elizabeth S Douglas
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States
| |
Collapse
|
4
|
Lee H, Han DW, La H, Park C, Kang K, Kwon O, Uhm SJ, Song H, Do JT, Choi Y, Hong K. DOT1-like histone lysine methyltransferase is critical for adult vessel maintenance and functions. Anim Biosci 2024; 37:1635-1643. [PMID: 38665093 PMCID: PMC11366529 DOI: 10.5713/ab.23.0402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 09/03/2024] Open
Abstract
OBJECTIVE Disruptor of telomeric silencing 1-like (DOT1L) is the only known histone H3K79 methyltransferase essential for the development of the embryonic cardiovascular system, including the heart, blood vessels, and lymphatic vessels, through transcriptional regulation. Our previous study demonstrated that Dot1l deletion results in aberrant lymphatic development and function. However, its precise function in the postnatal cardiovascular system remains unknown. METHODS Using conditional and inducible Dot1l knockout (KO) mice, along with a reporter strain carrying the Geo gene at the Dot1l locus, DOT1L expression and its function in the vascular system during postnatal life were investigated. To assess vessel morphology and vascular permeability, we administered Latex or Evans blue dye to KO mice. In addition, in vitro tube formation and cell migration assays were performed using DOT1L-depleted human umbilical vein endothelial cells (HUVECs). Changes in the expression of vascular genes in HUVECs were measured by quantitative polymerase chain reaction. RESULTS Our findings demonstrate that conditional Dot1l knockout in the Tg (Tie2-cre) strain results in abnormal blood vessel formation and lymphatic anomalies in the intestine. In a mouse model of Rosa26-creER-mediated inducible Dot1l knockout, we observed vascular phenotypes, including increased vascular permeability and brain hemorrhage, when DOT1L was deleted in adulthood. Additionally, DOT1L depletion in cultured HUVECs led to impaired cell migration and tube formation, likely due to altered gene transcription. These findings highlight the essential role of DOT1L in maintaining vascular integrity and function during embryonic development and postnatal life. CONCLUSION Our study revealed that DOT1L is required for the maintenance of adult vascular function through the regulation of gene expression.
Collapse
Affiliation(s)
- HeeJi Lee
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen 529020,
China
| | - Hyeonwoo La
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Chanhyeok Park
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Kiye Kang
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Ohbeom Kwon
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Sang Jun Uhm
- Department of Animal Science, Sangji University, Wonju 26339,
Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Jeong Tae Do
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
5
|
Song G, Liu D, Ma J, Zhan Y, Ma F, Liu G. Cardiac Lymphatics and Therapeutic Prospects in Cardiovascular Disease: New Perspectives and Hopes. Cardiol Rev 2024:00045415-990000000-00289. [PMID: 39150263 DOI: 10.1097/crd.0000000000000743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The lymphatic system is the same reticular fluid system as the circulatory system found throughout the body in vascularized tissues. Lymphatic vessels are low-pressure, blind-ended tubular structures that play a crucial role in maintaining tissue fluid homeostasis, immune cell transport, and lipid absorption. The heart also has an extensive lymphatic network, and as research on cardiac lymphatics has progressed in recent years, more and more studies have found that cardiac lymphangiogenesis may ameliorate certain cardiovascular diseases, and therefore stimulation of cardiac lymphangiogenesis may be an important tool in the future treatment of cardiovascular diseases. This article briefly reviews the development and function of cardiac lymphatic vessels, the interaction of cardiac lymphatic vessels with cardiovascular diseases (including atrial fibrillation, coronary atherosclerosis, and heart failure), and finally discusses the therapeutic potential of targeted cardiac lymphatic therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Guoyuan Song
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Da Liu
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianwei Ma
- Gastrointestinal Disease Diagnosis and Treatment Center, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yinge Zhan
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fangfang Ma
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Gang Liu
- From the Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Phng LK, Hogan BM. Endothelial cell transitions in zebrafish vascular development. Dev Growth Differ 2024; 66:357-368. [PMID: 39072708 PMCID: PMC11457512 DOI: 10.1111/dgd.12938] [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: 05/07/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
In recent decades, developmental biologists have come to view vascular development as a series of progressive transitions. Mesoderm differentiates into endothelial cells; arteries, veins and lymphatic endothelial cells are specified from early endothelial cells; and vascular networks diversify and invade developing tissues and organs. Our understanding of this elaborate developmental process has benefitted from detailed studies using the zebrafish as a model system. Here, we review a number of key developmental transitions that occur in zebrafish during the formation of the blood and lymphatic vessel networks.
Collapse
Affiliation(s)
- Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Benjamin M Hogan
- Organogenesis and Cancer Programme, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology and the Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Turner DL, Amoozadeh S, Baric H, Stanley E, Werder RB. Building a human lung from pluripotent stem cells to model respiratory viral infections. Respir Res 2024; 25:277. [PMID: 39010108 PMCID: PMC11251358 DOI: 10.1186/s12931-024-02912-0] [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: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.
Collapse
Affiliation(s)
- Declan L Turner
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Sahel Amoozadeh
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Hannah Baric
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Ed Stanley
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, 3056, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia.
| |
Collapse
|
8
|
Kumar A, Kumar R, Narayan RK, Nath B, Datusalia AK, Rastogi AK, Jha RK, Kumar P, Pareek V, Prasoon P, Faiq MA, Agrawal P, Prasad SN, Kumari C, Asghar A. Anatomical correlates for the newly discovered meningeal layer in the existing literature: A systematic review. Anat Rec (Hoboken) 2024. [PMID: 38924700 DOI: 10.1002/ar.25524] [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: 02/23/2024] [Revised: 05/07/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
The existence of a previously unrecognized subarachnoid lymphatic-like membrane (SLYM) was reported in a recent study. SLYM is described as an intermediate leptomeningeal layer between the arachnoid and pia mater in mouse and human brains, which divides the subarachnoid space (SAS) into two functional compartments. Being a macroscopic structure, having missed detection in previous studies is surprising. We systematically reviewed the published reports in animals and humans to explore whether prior descriptions of this meningeal layer were reported in some way. A comprehensive search was conducted in PubMed/Medline, EMBASE, Google Scholar, Science Direct, and Web of Science databases using combinations of MeSH terms and keywords with Boolean operators from inception until 31 December 2023. We found at least eight studies that provided structural evidence of an intermediate leptomeningeal layer in the brain or spinal cord. However, unequivocal descriptions for this layer all along the central nervous system were scarce. Obscure names like the epipial, intermediate meningeal, outer pial layers, or intermediate lamella were used to describe it. Its microscopic/ultrastructural details closely resemble the recently reported SLYM. We further examined the counterarguments in current literature that are skeptical of the existence of this layer. The potential physiological and clinical implications of this new meningeal layer are significant, underscoring the urgent need for further exploration of its structural and functional details.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Rajesh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Ravi K Narayan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Banshi Nath
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Ashok K Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Ashok K Rastogi
- Department of Forensic Medicine and Toxicology, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Rakesh K Jha
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Pankaj Kumar
- Regional Institute of Ophthalmology, Indira Gandhi Institute of Medical Sciences, Patna, India
| | - Vikas Pareek
- Haskins Laboratories, Yale Child Study Centre, Yale School of Medicine, University of Connecticut, New Haven, Connecticut, USA
| | - Pranav Prasoon
- Department of Anatomy and Cell Biology, George Washington University, Washington, DC, USA
| | - Muneeb A Faiq
- New York University (NYU) Langone Health Center, NYU Robert I Grossman School of Medicine, New York, New York, USA
| | - Prabhat Agrawal
- Spine Surgery Clinic, Department of Orthopedics, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Surya Nandan Prasad
- Department of Radiodiagnosis, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Chiman Kumari
- Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Adil Asghar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| |
Collapse
|
9
|
Stasi E, Sciascia S, Naretto C, Baldovino S, Roccatello D. Lymphatic System and the Kidney: From Lymphangiogenesis to Renal Inflammation and Fibrosis Development. Int J Mol Sci 2024; 25:2853. [PMID: 38474100 DOI: 10.3390/ijms25052853] [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: 01/05/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The lymphatic kidney system plays a crucial role in managing interstitial fluid removal, regulating fluid balance, and tuning immune response. It also assists in the reabsorption of proteins, electrolytes, cytokines, growth factors, and immune cells. Pathological conditions, including tissue damage, excessive interstitial fluid, high blood glucose levels, and inflammation, can initiate lymphangiogenesis-the formation of new lymphatic vessels. This process is associated with various kidney diseases, including polycystic kidney disease, hypertension, ultrafiltration challenges, and complications post-organ transplantation. Although lymphangiogenesis has beneficial effects in removing excess fluid and immune cells, it may also contribute to inflammation and fibrosis within the kidneys. In this review, we aim to discuss the biology of the lymphatic system, from its development and function to its response to disease stimuli, with an emphasis on renal pathophysiology. Furthermore, we explore how innovative treatments targeting the lymphatic system could potentially enhance the management of kidney diseases.
Collapse
Affiliation(s)
- Elodie Stasi
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, ASL Città di Torino and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy
| | - Savino Sciascia
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, ASL Città di Torino and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy
| | - Carla Naretto
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, ASL Città di Torino and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy
| | - Simone Baldovino
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, ASL Città di Torino and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy
| | - Dario Roccatello
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley, ASL Città di Torino and Department of Clinical and Biological Sciences, University of Turin, 10154 Turin, Italy
| |
Collapse
|
10
|
Yamaguchi S, Minamide N, Imai H, Ikeda T, Watanabe M, Imanaka-Yoshida K, Maruyama K. The development of early human lymphatic vessels as characterized by lymphatic endothelial markers. EMBO J 2024; 43:868-885. [PMID: 38351385 PMCID: PMC10907744 DOI: 10.1038/s44318-024-00045-0] [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: 11/09/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 03/03/2024] Open
Abstract
Lymphatic vessel development studies in mice and zebrafish models have demonstrated that lymphatic endothelial cells (LECs) predominantly differentiate from venous endothelial cells via the expression of the transcription factor Prox1. However, LECs can also be generated from undifferentiated mesoderm, suggesting potential diversity in their precursor cell origins depending on the organ or anatomical location. Despite these advances, recapitulating human lymphatic malformations in animal models has been difficult, and considering lymphatic vasculature function varies widely between species, analysis of development directly in humans is needed. Here, we examined early lymphatic development in humans by analyzing the histology of 31 embryos and three 9-week-old fetuses. We found that human embryonic cardinal veins, which converged to form initial lymph sacs, produce Prox1-expressing LECs. Furthermore, we describe the lymphatic vessel development in various organs and observe organ-specific differences. These characterizations of the early development of human lymphatic vessels should help to better understand the evolution and phylogenetic relationships of lymphatic systems, and their roles in human disease.
Collapse
Affiliation(s)
- Shoichiro Yamaguchi
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Natsuki Minamide
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Hiroshi Imai
- Pathology Division, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Masatoshi Watanabe
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan
| | - Kazuaki Maruyama
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-0001, Japan.
| |
Collapse
|
11
|
Weinberger M, Riley PR. Animal models to study cardiac regeneration. Nat Rev Cardiol 2024; 21:89-105. [PMID: 37580429 DOI: 10.1038/s41569-023-00914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/16/2023]
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
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: 9] [Impact Index Per Article: 9.0] [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
|
13
|
Dijkhuis L, Johns A, Ragusa D, van den Brink SC, Pina C. Haematopoietic development and HSC formation in vitro: promise and limitations of gastruloid models. Emerg Top Life Sci 2023; 7:439-454. [PMID: 38095554 PMCID: PMC10754337 DOI: 10.1042/etls20230091] [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: 08/22/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Haematopoietic stem cells (HSCs) are the most extensively studied adult stem cells. Yet, six decades after their first description, reproducible and translatable generation of HSC in vitro remains an unmet challenge. HSC production in vitro is confounded by the multi-stage nature of blood production during development. Specification of HSC is a late event in embryonic blood production and depends on physical and chemical cues which remain incompletely characterised. The precise molecular composition of the HSC themselves is incompletely understood, limiting approaches to track their origin in situ in the appropriate cellular, chemical and mechanical context. Embryonic material at the point of HSC emergence is limiting, highlighting the need for an in vitro model of embryonic haematopoietic development in which current knowledge gaps can be addressed and exploited to enable HSC production. Gastruloids are pluripotent stem cell-derived 3-dimensional (3D) cellular aggregates which recapitulate developmental events in gastrulation and early organogenesis with spatial and temporal precision. Gastruloids self-organise multi-tissue structures upon minimal and controlled external cues, and are amenable to live imaging, screening, scaling and physicochemical manipulation to understand and translate tissue formation. In this review, we consider the haematopoietic potential of gastruloids and review early strategies to enhance blood progenitor and HSC production. We highlight possible strategies to achieve HSC production from gastruloids, and discuss the potential of gastruloid systems in illuminating current knowledge gaps in HSC specification.
Collapse
Affiliation(s)
- Liza Dijkhuis
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Ayona Johns
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | - Denise Ragusa
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | | | - Cristina Pina
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| |
Collapse
|
14
|
Montenegro-Navarro N, García-Báez C, García-Caballero M. Molecular and metabolic orchestration of the lymphatic vasculature in physiology and pathology. Nat Commun 2023; 14:8389. [PMID: 38104163 PMCID: PMC10725466 DOI: 10.1038/s41467-023-44133-x] [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: 03/03/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023] Open
Abstract
Lymphangiogenesis refers to the generation of new lymphatic vessels from pre-existing ones. During development and particular adult states, lymphatic endothelial cells (LEC) undergo reprogramming of their transcriptomic and signaling networks to support the high demands imposed by cell proliferation and migration. Although there has been substantial progress in identifying growth factors and signaling pathways controlling lymphangiogenesis in the last decades, insights into the role of metabolism in lymphatic cell functions are just emerging. Despite numerous similarities between the main metabolic pathways existing in LECs, blood ECs (BEC) and other cell types, accumulating evidence has revealed that LECs acquire a unique metabolic signature during lymphangiogenesis, and their metabolic engine is intertwined with molecular regulatory networks, resulting in a tightly regulated and interconnected process. Considering the implication of lymphatic dysfunction in cancer and lymphedema, alongside other pathologies, recent findings hold promising opportunities to develop novel therapeutic approaches. In this review, we provide an overview of the status of knowledge in the molecular and metabolic network regulating the lymphatic vasculature in health and disease.
Collapse
Affiliation(s)
- Nieves Montenegro-Navarro
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
| | - Claudia García-Báez
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain.
| |
Collapse
|
15
|
Kobayashi S, Cox AG, Harvey KF, Hogan BM. Vasculature is getting Hip(po): Hippo signaling in vascular development and disease. Dev Cell 2023; 58:2627-2640. [PMID: 38052179 DOI: 10.1016/j.devcel.2023.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/29/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023]
Abstract
The Hippo signaling pathway regulates developmental organ growth, regeneration, and cell fate decisions. Although the role of the Hippo pathway, and its transcriptional effectors YAP and TAZ, has been well documented in many cell types and species, only recently have the roles for this pathway come to light in vascular development and disease. Experiments in mice, zebrafish, and in vitro have uncovered roles for the Hippo pathway, YAP, and TAZ in vasculogenesis, angiogenesis, and lymphangiogenesis. In addition, the Hippo pathway has been implicated in vascular cancers and cardiovascular diseases, thus identifying it as a potential therapeutic target for the treatment of these conditions. However, despite recent advances, Hippo's role in the vasculature is still underappreciated compared with its role in epithelial tissues. In this review, we appraise our current understanding of the Hippo pathway in blood and lymphatic vessel development and highlight the current knowledge gaps and opportunities for further research.
Collapse
Affiliation(s)
- Sakurako Kobayashi
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew G Cox
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kieran F Harvey
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
16
|
Jain A, Ang PS, Matrongolo MJ, Tischfield MA. Understanding the development, pathogenesis, and injury response of meningeal lymphatic networks through the use of animal models. Cell Mol Life Sci 2023; 80:332. [PMID: 37872442 PMCID: PMC11072018 DOI: 10.1007/s00018-023-04984-5] [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: 08/18/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023]
Abstract
Meningeal lymphatic vessels (MLVs) help maintain central nervous system (CNS) homeostasis via their ability to facilitate macromolecule waste clearance and neuroimmune trafficking. Although these vessels were overlooked for centuries, they have now been characterized in humans, non-human primates, and rodents. Recent studies in mice have explored the stereotyped growth and expansion of MLVs in dura mater, the various transcriptional, signaling, and environmental factors regulating their development and long-term maintenance, and the pathological changes these vessels undergo in injury, disease, or with aging. Key insights gained from these studies have also been leveraged to develop therapeutic approaches that help augment or restore MLV functions to improve brain health and cognition. Here, we review fundamental processes that control the development of peripheral lymphatic networks and how these might apply to the growth and expansion of MLVs in their unique meningeal environment. We also emphasize key findings in injury and disease models that may reveal additional insights into the plasticity of these vessels throughout the lifespan. Finally, we highlight unanswered questions and future areas of study that can further reveal the exciting therapeutic potential of meningeal lymphatics.
Collapse
Affiliation(s)
- Aditya Jain
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Phillip S Ang
- University of Chicago Pritzker School of Medicine, Chicago, IL, 60637, USA
| | - Matthew J Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Max A Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA.
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
17
|
Phan QM, Salz L, Kindl SS, Lopez JS, Thompson SM, Makkar J, Driskell IM, Driskell RR. Lineage commitment of dermal fibroblast progenitors is controlled by Kdm6b-mediated chromatin demethylation. EMBO J 2023; 42:e113880. [PMID: 37602956 PMCID: PMC10548174 DOI: 10.15252/embj.2023113880] [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: 03/02/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Dermal Fibroblast Progenitors (DFPs) differentiate into distinct fibroblast lineages during skin development. However, the epigenetic mechanisms that regulate DFP differentiation are not known. Our objective was to use multimodal single-cell approaches, epigenetic assays, and allografting techniques to define a DFP state and the mechanism that governs its differentiation potential. Our initial results indicated that the overall transcription profile of DFPs is repressed by H3K27me3 and has inaccessible chromatin at lineage-specific genes. Surprisingly, the repressive chromatin profile of DFPs renders them unable to reform the skin in allograft assays despite their multipotent potential. We hypothesized that chromatin derepression was modulated by the H3K27me3 demethylase, Kdm6b/Jmjd3. Dermal fibroblast-specific deletion of Kdm6b/Jmjd3 in mice resulted in adipocyte compartment ablation and inhibition of mature dermal papilla functions, confirmed by additional single-cell RNA-seq, ChIP-seq, and allografting assays. We conclude that DFPs are functionally derepressed during murine skin development by Kdm6b/Jmjd3. Our studies therefore reveal a multimodal understanding of how DFPs differentiate into distinct fibroblast lineages and provide a novel publicly available multiomics search tool.
Collapse
Affiliation(s)
- Quan M Phan
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Lucia Salz
- North Rhine‐Westphalia Technical University of AachenAachenGermany
| | - Sam S Kindl
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Jayden S Lopez
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Sean M Thompson
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Jasson Makkar
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Iwona M Driskell
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Ryan R Driskell
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
- Center for Reproductive BiologyWashington State UniversityPullmanWAUSA
| |
Collapse
|
18
|
Travisano SI, Harrison MRM, Thornton ME, Grubbs BH, Quertermous T, Lien CL. Single-nuclei multiomic analyses identify human cardiac lymphatic endothelial cells associated with coronary arteries in the epicardium. Cell Rep 2023; 42:113106. [PMID: 37676760 DOI: 10.1016/j.celrep.2023.113106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/31/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Cardiac lymphatic vessels play important roles in fluid homeostasis, inflammation, disease, and regeneration of the heart. The developing cardiac lymphatics in human fetal hearts are closely associated with coronary arteries, similar to those in zebrafish hearts. We identify a population of cardiac lymphatic endothelial cells (LECs) that reside in the epicardium. Single-nuclei multiomic analysis of the human fetal heart reveals the plasticity and heterogeneity of the cardiac endothelium. Furthermore, we find that VEGFC is highly expressed in arterial endothelial cells and epicardium-derived cells, providing a molecular basis for the arterial association of cardiac lymphatic development. Using a cell-type-specific integrative analysis, we identify a population of cardiac lymphatic endothelial cells marked by the PROX1 and the lymphangiocrine RELN and enriched in binding motifs of erythroblast transformation specific (ETS) variant (ETV) transcription factors. We report the in vivo molecular characterization of human cardiac lymphatics and provide a valuable resource to understand fetal heart development.
Collapse
Affiliation(s)
| | - Michael R M Harrison
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Matthew E Thornton
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brendan H Grubbs
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and the Cardiovascular Institute, School of Medicine, Stanford University, Falk CVRC, Stanford, CA 94305, USA
| | - Ching-Ling Lien
- The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Departments of Surgery, Biochemistry, and Molecular Medicine, Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
19
|
Lotto J, Stephan TL, Hoodless PA. Fetal liver development and implications for liver disease pathogenesis. Nat Rev Gastroenterol Hepatol 2023; 20:561-581. [PMID: 37208503 DOI: 10.1038/s41575-023-00775-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/21/2023]
Abstract
The metabolic, digestive and homeostatic roles of the liver are dependent on proper crosstalk and organization of hepatic cell lineages. These hepatic cell lineages are derived from their respective progenitors early in organogenesis in a spatiotemporally controlled manner, contributing to the liver's specialized and diverse microarchitecture. Advances in genomics, lineage tracing and microscopy have led to seminal discoveries in the past decade that have elucidated liver cell lineage hierarchies. In particular, single-cell genomics has enabled researchers to explore diversity within the liver, especially early in development when the application of bulk genomics was previously constrained due to the organ's small scale, resulting in low cell numbers. These discoveries have substantially advanced our understanding of cell differentiation trajectories, cell fate decisions, cell lineage plasticity and the signalling microenvironment underlying the formation of the liver. In addition, they have provided insights into the pathogenesis of liver disease and cancer, in which developmental processes participate in disease emergence and regeneration. Future work will focus on the translation of this knowledge to optimize in vitro models of liver development and fine-tune regenerative medicine strategies to treat liver disease. In this Review, we discuss the emergence of hepatic parenchymal and non-parenchymal cells, advances that have been made in in vitro modelling of liver development and draw parallels between developmental and pathological processes.
Collapse
Affiliation(s)
- Jeremy Lotto
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Tabea L Stephan
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada
| | - Pamela A Hoodless
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, Canada.
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Mehrara BJ, Radtke AJ, Randolph GJ, Wachter BT, Greenwel P, Rovira II, Galis ZS, Muratoglu SC. The emerging importance of lymphatics in health and disease: an NIH workshop report. J Clin Invest 2023; 133:e171582. [PMID: 37655664 PMCID: PMC10471172 DOI: 10.1172/jci171582] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
The lymphatic system (LS) is composed of lymphoid organs and a network of vessels that transport interstitial fluid, antigens, lipids, cholesterol, immune cells, and other materials in the body. Abnormal development or malfunction of the LS has been shown to play a key role in the pathophysiology of many disease states. Thus, improved understanding of the anatomical and molecular characteristics of the LS may provide approaches for disease prevention or treatment. Recent advances harnessing single-cell technologies, clinical imaging, discovery of biomarkers, and computational tools have led to the development of strategies to study the LS. This Review summarizes the outcomes of the NIH workshop entitled "Yet to be Charted: Lymphatic System in Health and Disease," held in September 2022, with emphasis on major areas for advancement. International experts showcased the current state of knowledge regarding the LS and highlighted remaining challenges and opportunities to advance the field.
Collapse
Affiliation(s)
- Babak J. Mehrara
- Department of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrea J. Radtke
- Lymphocyte Biology Section and Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Gwendalyn J. Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brianna T. Wachter
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Patricia Greenwel
- Division of Digestive Diseases & Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, and
| | - Ilsa I. Rovira
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Zorina S. Galis
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Selen C. Muratoglu
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Yoo H, La H, Park C, Yoo S, Lee H, Song H, Do JT, Choi Y, Hong K. Common and distinct functions of mouse Dot1l in the regulation of endothelial transcriptome. Front Cell Dev Biol 2023; 11:1176115. [PMID: 37397258 PMCID: PMC10311421 DOI: 10.3389/fcell.2023.1176115] [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: 02/28/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
Epigenetic mechanisms are mandatory for endothelial called lymphangioblasts during cardiovascular development. Dot1l-mediated gene transcription in mice is essential for the development and function of lymphatic ECs (LECs). The role of Dot1l in the development and function of blood ECs blood endothelial cells is unclear. RNA-seq datasets from Dot1l-depleted or -overexpressing BECs and LECs were used to comprehensively analyze regulatory networks of gene transcription and pathways. Dot1l depletion in BECs changed the expression of genes involved in cell-to-cell adhesion and immunity-related biological processes. Dot1l overexpression modified the expression of genes involved in different types of cell-to-cell adhesion and angiogenesis-related biological processes. Genes involved in specific tissue development-related biological pathways were altered in Dot1l-depleted BECs and LECs. Dot1l overexpression altered ion transportation-related genes in BECs and immune response regulation-related genes in LECs. Importantly, Dot1l overexpression in BECs led to the expression of genes related to the angiogenesis and increased expression of MAPK signaling pathways related was found in both Dot1l-overexpressing BECs and LECs. Therefore, our integrated analyses of transcriptomics in Dot1l-depleted and Dot1l-overexpressed ECs demonstrate the unique transcriptomic program of ECs and the differential functions of Dot1l in the regulation of gene transcription in BECs and LECs.
Collapse
|
22
|
Kenney HM, Peng Y, de Mesy Bentley KL, Xing L, Ritchlin CT, Schwarz EM. The Enigmas of Lymphatic Muscle Cells: Where Do They Come From, How Are They Maintained, and Can They Regenerate? Curr Rheumatol Rev 2023; 19:246-259. [PMID: 36705238 PMCID: PMC10257750 DOI: 10.2174/1573397119666230127144711] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/29/2022] [Accepted: 12/02/2022] [Indexed: 01/28/2023]
Abstract
Lymphatic muscle cell (LMC) contractility and coverage of collecting lymphatic vessels (CLVs) are integral to effective lymphatic drainage and tissue homeostasis. In fact, defects in lymphatic contractility have been identified in various conditions, including rheumatoid arthritis, inflammatory bowel disease, and obesity. However, the fundamental role of LMCs in these pathologic processes is limited, primarily due to the difficulty in directly investigating the enigmatic nature of this poorly characterized cell type. LMCs are a unique cell type that exhibit dual tonic and phasic contractility with hybrid structural features of both vascular smooth muscle cells (VSMCs) and cardiac myocytes. While advances have been made in recent years to better understand the biochemistry and function of LMCs, central questions regarding their origins, investiture into CLVs, and homeostasis remain unanswered. To summarize these discoveries, unexplained experimental results, and critical future directions, here we provide a focused review of current knowledge and open questions related to LMC progenitor cells, recruitment, maintenance, and regeneration. We also highlight the high-priority research goal of identifying LMC-specific genes towards genetic conditional- inducible in vivo gain and loss of function studies. While our interest in LMCs has been focused on understanding lymphatic dysfunction in an arthritic flare, these concepts are integral to the broader field of lymphatic biology, and have important potential for clinical translation through targeted therapeutics to control lymphatic contractility and drainage.
Collapse
Grants
- R01AG059775,R01AG059775,R01AG059775 NIA NIH HHS
- R01AR056702,R01AR069000,T32AR076950,P30AR069655,R01AR056702,R01AR069000,P30AR069655,T32AR076950,R01AR056702,R01AR069000,T32AR076950,P30AR069655 NIAMS NIH HHS
- P30 AR069655 NIAMS NIH HHS
- R01 AR069000 NIAMS NIH HHS
- T32 GM007356 NIGMS NIH HHS
- R01 AG059775 NIA NIH HHS
- T32GM007356,T32GM007356,T32GM007356,T32GM007356 NIGMS NIH HHS
- T32 AR076950 NIAMS NIH HHS
- R01 AR056702 NIAMS NIH HHS
- F30 AG076326 NIA NIH HHS
Collapse
Affiliation(s)
- H. Mark Kenney
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Yue Peng
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher T. Ritchlin
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy, Immunology, Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
23
|
Grimm L, Mason E, Yu H, Dudczig S, Panara V, Chen T, Bower NI, Paterson S, Rondon Galeano M, Kobayashi S, Senabouth A, Lagendijk AK, Powell J, Smith KA, Okuda KS, Koltowska K, Hogan BM. Single-cell analysis of lymphatic endothelial cell fate specification and differentiation during zebrafish development. EMBO J 2023:e112590. [PMID: 36912146 DOI: 10.15252/embj.2022112590] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/24/2023] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
During development, the lymphatic vasculature forms as a second network derived chiefly from blood vessels. The transdifferentiation of embryonic venous endothelial cells (VECs) into lymphatic endothelial cells (LECs) is a key step in this process. Specification, differentiation and maintenance of LEC fate are all driven by the transcription factor Prox1, yet the downstream mechanisms remain to be elucidated. We here present a single-cell transcriptomic atlas of lymphangiogenesis in zebrafish, revealing new markers and hallmarks of LEC differentiation over four developmental stages. We further profile single-cell transcriptomic and chromatin accessibility changes in zygotic prox1a mutants that are undergoing a LEC-VEC fate shift. Using maternal and zygotic prox1a/prox1b mutants, we determine the earliest transcriptomic changes directed by Prox1 during LEC specification. This work altogether reveals new downstream targets and regulatory regions of the genome controlled by Prox1 and presents evidence that Prox1 specifies LEC fate primarily by limiting blood vascular and haematopoietic fate. This extensive single-cell resource provides new mechanistic insights into the enigmatic role of Prox1 and the control of LEC differentiation in development.
Collapse
Affiliation(s)
- Lin Grimm
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Elizabeth Mason
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Hujun Yu
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Stefanie Dudczig
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Virginia Panara
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Tyrone Chen
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Neil I Bower
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Scott Paterson
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Maria Rondon Galeano
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sakurako Kobayashi
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Anne Senabouth
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia.,Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Joseph Powell
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia.,Garvan Institute of Medical Research, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Kensington, Sydney, NSW, Australia.,Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Kelly A Smith
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Kazuhide S Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, Australia.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Phan QM, Salz L, Kindl SS, Lopez JS, Thompson SM, Makkar J, Driskell IM, Driskell RR. Lineage Commitment of Dermal Fibroblast Progenitors is Mediated by Chromatin De-repression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531478. [PMID: 36945417 PMCID: PMC10028926 DOI: 10.1101/2023.03.07.531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Dermal Fibroblast Progenitors (DFPs) differentiate into distinct fibroblast lineages during skin development. However, the mechanisms that regulate lineage commitment of naive dermal progenitors to form niches around the hair follicle, dermis, and hypodermis, are unknown. In our study, we used multimodal single-cell approaches, epigenetic assays, and allografting techniques to define a DFP state and the mechanisms that govern its differentiation potential. Our results indicate that the overall chromatin profile of DFPs is repressed by H3K27me3 and has inaccessible chromatin at lineage specific genes. Surprisingly, the repressed chromatin profile of DFPs renders them unable to reform skin in allograft assays despite their multipotent potential. Distinct fibroblast lineages, such as the dermal papilla and adipocytes contained specific chromatin profiles that were de-repressed during late embryogenesis by the H3K27-me3 demethylase, Kdm6b/Jmjd3. Tissue-specific deletion of Kdm6b/Jmjd3 resulted in ablating the adipocyte compartment and inhibiting mature dermal papilla functions in single-cell-RNA-seq, ChIPseq, and allografting assays. Altogether our studies reveal a mechanistic multimodal understanding of how DFPs differentiate into distinct fibroblast lineages, and we provide a novel multiomic search-tool within skinregeneration.org.
Collapse
Affiliation(s)
- Quan M. Phan
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Lucia Salz
- North Rhine-Westphalia Technical University of Aachen, Aachen, Germany
| | - Sam S. Kindl
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Jayden S. Lopez
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Sean M. Thompson
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Jasson Makkar
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Iwona M. Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Ryan R. Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA
- Center for Reproductive Biology, Washington State University, Pullman, WA
| |
Collapse
|
25
|
Nakajima H, Ishikawa H, Yamamoto T, Chiba A, Fukui H, Sako K, Fukumoto M, Mattonet K, Kwon HB, Hui SP, Dobreva GD, Kikuchi K, Helker CSM, Stainier DYR, Mochizuki N. Endoderm-derived islet1-expressing cells differentiate into endothelial cells to function as the vascular HSPC niche in zebrafish. Dev Cell 2023; 58:224-238.e7. [PMID: 36693371 DOI: 10.1016/j.devcel.2022.12.013] [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: 11/20/2021] [Revised: 10/26/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023]
Abstract
Endothelial cells (ECs) line blood vessels and serve as a niche for hematopoietic stem and progenitor cells (HSPCs). Recent data point to tissue-specific EC specialization as well as heterogeneity; however, it remains unclear how ECs acquire these properties. Here, by combining live-imaging-based lineage-tracing and single-cell transcriptomics in zebrafish embryos, we identify an unexpected origin for part of the vascular HSPC niche. We find that islet1 (isl1)-expressing cells are the progenitors of the venous ECs that constitute the majority of the HSPC niche. These isl1-expressing cells surprisingly originate from the endoderm and differentiate into ECs in a process dependent on Bmp-Smad signaling and subsequently requiring npas4l (cloche) function. Single-cell RNA sequencing analyses show that isl1-derived ECs express a set of genes that reflect their distinct origin. This study demonstrates that endothelial specialization in the HSPC niche is determined at least in part by the origin of the ECs.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; AMED-CREST, AMED, Tokyo 100-0004, Japan; Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Keisuke Sako
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Moe Fukumoto
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Hyouk-Bum Kwon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Subhra P Hui
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata 700019, India
| | - Gergana D Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Kazu Kikuchi
- Department of Cardiac Regeneration Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Christian S M Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany; Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg 35043, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
26
|
Filosa A, Sawamiphak S. Heart development and regeneration-a multi-organ effort. FEBS J 2023; 290:913-930. [PMID: 34894086 DOI: 10.1111/febs.16319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Development of the heart, from early morphogenesis to functional maturation, as well as maintenance of its homeostasis are tasks requiring collaborative efforts of cardiac tissue and different extra-cardiac organ systems. The brain, lymphoid organs, and gut are among the interaction partners that can communicate with the heart through a wide array of paracrine signals acting at local or systemic level. Disturbance of cardiac homeostasis following ischemic injury also needs immediate response from these distant organs. Our hearts replace dead muscles with non-contractile fibrotic scars. We have learned from animal models capable of scarless repair that regenerative capability of the heart does not depend only on competency of the myocardium and cardiac-intrinsic factors but also on long-range molecular signals originating in other parts of the body. Here, we provide an overview of inter-organ signals that take part in development and regeneration of the heart. We highlight recent findings and remaining questions. Finally, we discuss the potential of inter-organ modulatory approaches for possible therapeutic use.
Collapse
Affiliation(s)
- Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany
| |
Collapse
|
27
|
Leboulanger N, Bisdorff A, Boccara O, Dompmartin A, Guibaud L, Labreze C, Lagier J, Lebrun-Vignes B, Herbreteau D, Joly A, Malloizel-Delaunay J, Martel A, Munck S, Saint-Aubin F, Maruani A. French national diagnosis and care protocol (PNDS, protocole national de diagnostic et de soins): cystic lymphatic malformations. Orphanet J Rare Dis 2023; 18:10. [PMID: 36639640 PMCID: PMC9837920 DOI: 10.1186/s13023-022-02608-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023] Open
Abstract
Cystic lymphatic malformations (LMs) are rare chronic conditions which management differs according to the type (macrocystic LMs, microcystic LMs or both). Studies are lacking due to rarity of the pathology. We aimed to establish a French National Diagnosis and Care Protocol (PNDS: Protocole National de Diagnostic et de Soins), to provide health professionals with free open access synthesis on optimal management and care of patients with LMs ( https://www.has-sante.fr/upload/docs/application/pdf/2021-03/malformations_lymphatiques_kystiques_-_pnds.pdf ). The process included a critical review of the literature and multidisciplinary expert consensus. LMs are congenital but are not always discovered at birth. Nearly 75% of them are located in the head and neck because of the highly dense lymphatic system in this region. Physical examination (showing painless masses with normal skin color and depressible consistency, or cutaneous/mucosal lymphangiectasia) and color Doppler ultrasonography, usually allow for diagnosis. MRI (involving T2 sequences with fat saturation in at least two spatial planes) is the tool of choice for evaluating anatomical extension, characterizing lesions (microcystic and macrocystic), and before considering therapeutic management. A biopsy, coupled to a blood sample, can also be used for molecular biology analyses, to search for activating mutations of the PIK3CA gene, particularly with LM integrating in a syndromic form (CLOVES or Klippel-Trenaunay syndrome) but also in certain isolated (or common) LMs. The spontaneous evolution of LMs, in particular microcystic forms, is often toward progressive aggravation, with an increase in the number of vesicles, thickening, increased oozing and bleeding, while pure macrocystic LMs may regress due to "natural sclerosis", i.e. fibrosis secondary to an inflammatory reorganization after common infantile infections. In case of voluminous LMs or syndromic forms, functional and psychological repercussions can be major, deteriorating the patient's quality of life. LMs must be treated by physicians integrated in multidisciplinary teams, and be personalized. Management is a life-long process that involves one or several of these therapies: conservative management, physical therapy (compression), sclerotherapy, surgery, drugs such as mTOR inhibitors (sirolimus), that has shown efficacy in decreasing the volume of LMs, and, more recently, PI3K-inhibitors in syndromic forms. Psychological and social support is necessary, taking into account the patient and his family.
Collapse
Affiliation(s)
- Nicolas Leboulanger
- Otolaryngology - Head and Neck Surgery Department. National Reference Center for Rare Otolaryngological Malformations (MALO), Necker Enfants Malades Hospital, 149 Rue de Sèvres, 75015, Paris, France. .,INSERM U955, Paris Cité University. ERN Cranio, Paris, France.
| | - Annouk Bisdorff
- grid.411296.90000 0000 9725 279XDepartment of Interventional Radiology, Lariboisière Hospital, Paris, France
| | - Olivia Boccara
- grid.412134.10000 0004 0593 9113Department of Dermatology and Reference Center for Rare Diseases and Vascular Malformations (MAGEC), Necker Enfants Malades Hospital, Paris, France
| | - Anne Dompmartin
- grid.411149.80000 0004 0472 0160Department of Dermatology, CHU Côte de Nacre, Caen, France
| | - Laurent Guibaud
- grid.413852.90000 0001 2163 3825Department of Radiology, Hôpital Mère-Enfant, CHU de Lyon, Lyon, France
| | - Christine Labreze
- grid.42399.350000 0004 0593 7118Department of Dermatology, Pellegrin Hospital, CHU de Bordeaux, Bordeaux, France
| | - Jacques Lagier
- grid.410528.a0000 0001 2322 4179Department of Ophthalmology, CHU de Nice, Nice, France
| | - Bénédicte Lebrun-Vignes
- grid.411439.a0000 0001 2150 9058Pharmacovigilance Unit, AP-HP, Department of Pharmacology, Pitié-Salpêtrière Hospital, Paris, France
| | - Denis Herbreteau
- grid.411167.40000 0004 1765 1600Department of Neuroradiology and Interventional Radiology - Reference Center for Rare Diseases and Vascular Malformations (MAGEC), CHRU de Tours, Tours, France
| | - Aline Joly
- grid.411167.40000 0004 1765 1600Department of Maxillofacial Surgery - Reference Center for Rare Diseases and Vascular Malformations (MAGEC), CHRU de Tours, 37044 Tours, Cedex 9 France
| | - Julie Malloizel-Delaunay
- grid.411175.70000 0001 1457 2980Department of Vascular Medicine, Rangueil Hospital, CHU de Toulouse, Toulouse, France
| | - Arnaud Martel
- grid.410528.a0000 0001 2322 4179Department of Ophthalmology, CHU de Nice, Nice, France
| | - Stéphane Munck
- grid.460782.f0000 0004 4910 6551Département d’enseignement et de Recherche en Médecine Générale, Retines, Healthy, Université Côte d’Azur, 06000 Nice, France
| | | | - Annabel Maruani
- grid.411167.40000 0004 1765 1600Department of Dermatology and Reference Center for Rare Diseases and Vascular Malformations (MAGEC), CHRU de Tours, Tours, France ,grid.12366.300000 0001 2182 6141INSERM 1246 ‑ SPHERE, Universities of Tours and Nantes, 37000 Tours, France
| |
Collapse
|
28
|
González-Hernández S, Mukouyama YS. Lymphatic vasculature in the central nervous system. Front Cell Dev Biol 2023; 11:1150775. [PMID: 37091974 PMCID: PMC10119411 DOI: 10.3389/fcell.2023.1150775] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The central nervous system (CNS) is considered as an immune privilege organ, based on experiments in the mid 20th century showing that the brain fails to mount an efficient immune response against an allogeneic graft. This suggests that in addition to the presence of the blood-brain barrier (BBB), the apparent absence of classical lymphatic vasculature in the CNS parenchyma limits the capacity for an immune response. Although this view is partially overturned by the recent discovery of the lymphatic-like hybrid vessels in the Schlemm's canal in the eye and the lymphatic vasculature in the outmost layer of the meninges, the existence of lymphatic vessels in the CNS parenchyma has not been reported. Two potential mechanisms by which lymphatic vasculature may arise in the organs are: 1) sprouting and invasion of lymphatic vessels from the surrounding tissues into the parenchyma and 2) differentiation of blood endothelial cells into lymphatic endothelial cells in the parenchyma. Considering these mechanisms, we here discuss what causes the dearth of lymphatic vessels specifically in the CNS parenchyma.
Collapse
|
29
|
Liu X, Cui K, Wu H, Li KS, Peng Q, Wang D, Cowan DB, Dixon JB, Srinivasan RS, Bielenberg DR, Chen K, Wang DZ, Chen Y, Chen H. Promoting Lymphangiogenesis and Lymphatic Growth and Remodeling to Treat Cardiovascular and Metabolic Diseases. Arterioscler Thromb Vasc Biol 2023; 43:e1-e10. [PMID: 36453280 PMCID: PMC9780193 DOI: 10.1161/atvbaha.122.318406] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022]
Abstract
Lymphatic vessels are low-pressure, blind-ended tubular structures that play a crucial role in the maintenance of tissue fluid homeostasis, immune cell trafficking, and dietary lipid uptake and transport. Emerging research has indicated that the promotion of lymphatic vascular growth, remodeling, and function can reduce inflammation and diminish disease severity in several pathophysiologic conditions. In particular, recent groundbreaking studies have shown that lymphangiogenesis, which describes the formation of new lymphatic vessels from the existing lymphatic vasculature, can be beneficial for the alleviation and resolution of metabolic and cardiovascular diseases. Therefore, promoting lymphangiogenesis represents a promising therapeutic approach. This brief review summarizes the most recent findings related to the modulation of lymphatic function to treat metabolic and cardiovascular diseases such as obesity, myocardial infarction, atherosclerosis, and hypertension. We also discuss experimental and therapeutic approaches to enforce lymphatic growth and remodeling as well as efforts to define the molecular and cellular mechanisms underlying these processes.
Collapse
Affiliation(s)
- Xiaolei Liu
- Lemole Center for Integrated Lymphatics Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Kathryn S. Li
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Qianman Peng
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Donghai Wang
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Douglas B. Cowan
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - J. Brandon Dixon
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Diane R. Bielenberg
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Kaifu Chen
- Department of Cardiology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Da-Zhi Wang
- USF Heart Institute, Center for Regenerative Medicine, College of Medicine Internal Medicine, University of South Florida, Tampa, FL
| | - Yabing Chen
- Department of Pathology, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, AL
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Harris NR, Bálint L, Dy DM, Nielsen NR, Méndez HG, Aghajanian A, Caron KM. The ebb and flow of cardiac lymphatics: a tidal wave of new discoveries. Physiol Rev 2023; 103:391-432. [PMID: 35953269 PMCID: PMC9576179 DOI: 10.1152/physrev.00052.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The heart is imbued with a vast lymphatic network that is responsible for fluid homeostasis and immune cell trafficking. Disturbances in the forces that regulate microvascular fluid movement can result in myocardial edema, which has profibrotic and proinflammatory consequences and contributes to cardiovascular dysfunction. This review explores the complex relationship between cardiac lymphatics, myocardial edema, and cardiac disease. It covers the revised paradigm of microvascular forces and fluid movement around the capillary as well as the arsenal of preclinical tools and animal models used to model myocardial edema and cardiac disease. Clinical studies of myocardial edema and their prognostic significance are examined in parallel to the recent elegant animal studies discerning the pathophysiological role and therapeutic potential of cardiac lymphatics in different cardiovascular disease models. This review highlights the outstanding questions of interest to both basic scientists and clinicians regarding the roles of cardiac lymphatics in health and disease.
Collapse
Affiliation(s)
- Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - László Bálint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle M Dy
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hernán G Méndez
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Amir Aghajanian
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
31
|
The Impact of Stem/Progenitor Cells on Lymphangiogenesis in Vascular Disease. Cells 2022; 11:cells11244056. [PMID: 36552820 PMCID: PMC9776475 DOI: 10.3390/cells11244056] [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: 10/23/2022] [Revised: 12/03/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022] Open
Abstract
Lymphatic vessels, as the main tube network of fluid drainage and leukocyte transfer, are responsible for the maintenance of homeostasis and pathological repairment. Recently, by using genetic lineage tracing and single-cell RNA sequencing techniques, significant cognitive progress has been made about the impact of stem/progenitor cells during lymphangiogenesis. In the embryonic stage, the lymphatic network is primarily formed through self-proliferation and polarized-sprouting from the lymph sacs. However, the assembly of lymphatic stem/progenitor cells also guarantees the sustained growth of lymphvasculogenesis to obtain the entire function. In addition, there are abundant sources of stem/progenitor cells in postnatal tissues, including circulating progenitors, mesenchymal stem cells, and adipose tissue stem cells, which can directly differentiate into lymphatic endothelial cells and participate in lymphangiogenesis. Specifically, recent reports indicated a novel function of lymphangiogenesis in transplant arteriosclerosis and atherosclerosis. In the present review, we summarized the latest evidence about the diversity and incorporation of stem/progenitor cells in lymphatic vasculature during both the embryonic and postnatal stages, with emphasis on the impact of lymphangiogenesis in the development of vascular diseases to provide a rational guidance for future research.
Collapse
|
32
|
Itoh F, Watabe T. TGF-β signaling in lymphatic vascular vessel formation and maintenance. Front Physiol 2022; 13:1081376. [PMID: 36589453 PMCID: PMC9799095 DOI: 10.3389/fphys.2022.1081376] [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: 10/27/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor (TGF)-β and its family members, including bone morphogenetic proteins (BMPs), nodal proteins, and activins, are implicated in the development and maintenance of various organs. Here, we review its role in the lymphatic vascular system (the secondary vascular system in vertebrates), which plays a crucial role in various physiological and pathological processes, participating in the maintenance of the normal tissue fluid balance, immune cell trafficking, and fatty acid absorption in the gut. The lymphatic system is associated with pathogenesis in multiple diseases, including lymphedema, inflammatory diseases, and tumor metastasis. Lymphatic vessels are composed of lymphatic endothelial cells, which differentiate from blood vascular endothelial cells (BECs). Although TGF-β family signaling is essential for maintaining blood vessel function, little is known about the role of TGF-β in lymphatic homeostasis. Recently, we reported that endothelial-specific depletion of TGF-β signaling affects lymphatic function. These reports suggest that TGF-β signaling in lymphatic endothelial cells maintains the structure of lymphatic vessels and lymphatic homeostasis, and promotes tumor lymphatic metastasis. Suppression of TGF-β signaling in lymphatic endothelial cells may therefore be effective in inhibiting cancer metastasis. We highlight recent advances in understanding the roles of TGF-β signaling in the formation and maintenance of the lymphatic system.
Collapse
Affiliation(s)
- Fumiko Itoh
- Laboratory of Stem Cells Regulations, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,*Correspondence: Fumiko Itoh, ; Tetsuro Watabe,
| |
Collapse
|
33
|
Ujiie N, Kume T. Mechanical forces in lymphatic vessel development: Focus on transcriptional regulation. Front Physiol 2022; 13:1066460. [PMID: 36439271 PMCID: PMC9685408 DOI: 10.3389/fphys.2022.1066460] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
The lymphatic system is crucial for the maintenance of interstitial fluid and protein homeostasis. It has important roles in collecting excess plasma and interstitial fluid leaked from blood vessels, lipid absorption and transportation in the digestive system, and immune surveillance and response. The development of lymphatic vessels begins during fetal life as lymphatic endothelial progenitor cells first differentiate into lymphatic endothelial cells (LECs) by expressing the master lymphatic vascular regulator, prospero-related homeobox 1 (PROX1). The lymphatic vasculature forms a hierarchical network that consists of blind-ended and unidirectional vessels. Although much progress has been made in the elucidation of the cellular and molecular mechanisms underlying the formation of the lymphatic vascular system, the causes of lymphatic vessel abnormalities and disease are poorly understood and complicated; specifically, the mechanistic basis for transcriptional dysregulation in lymphatic vessel development remains largely unclear. In this review, we discuss the recent advances in our understanding of the molecular and cellular mechanisms of lymphatic vascular development, including LEC differentiation, lymphangiogenesis, and valve formation, and the significance of mechanical forces in lymphatic vessels, with a focus on transcriptional regulation. We also summarize the current knowledge on epigenetic mechanisms of lymphatic gene expression.
Collapse
|
34
|
Zhang Y, Ortsäter H, Martinez-Corral I, Mäkinen T. Cdh5-lineage-independent origin of dermal lymphatics shown by temporally restricted lineage tracing. Life Sci Alliance 2022; 5:5/11/e202201561. [PMID: 35961777 PMCID: PMC9375154 DOI: 10.26508/lsa.202201561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/24/2022] Open
Abstract
Temporally restricted lineage tracing reveals a non-venous source of dermal lymphatic vessels and highlights Cre induction strategy as a critical parameter for stage-specific cell labeling. The developmental origins of lymphatic endothelial cells (LECs) have been under intense research after a century-long debate. Although previously thought to be of solely venous endothelial origin, additional sources of LECs were recently identified in multiple tissues in mice. Here, we investigated the regional differences in the origin(s) of the dermal lymphatic vasculature by lineage tracing using the pan-endothelial Cdh5-CreERT2 line. Tamoxifen-induced labeling of blood ECs at E9.5, before initiation of lymphatic development, traced most of the dermal LECs but with lower efficiency in the lumbar compared with the cervical skin. By contrast, when used at E9.5 but not at E11.5, 4-hydroxytamoxifen, the active metabolite of tamoxifen that provides a tighter window of Cre activity, revealed low labeling frequency of LECs, and lymphvasculogenic clusters in the lumbar skin in particular. Temporally restricted lineage tracing thus reveals contribution of LECs of Cdh5-lineage–independent origin to dermal lymphatic vasculature. Our results further highlight Cre induction strategy as a critical parameter in defining the temporal window for stage-specific lineage tracing during early developmental stages of rapid tissue differentiation.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Henrik Ortsäter
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ines Martinez-Corral
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Mattonet K, Riemslagh FW, Guenther S, Prummel KD, Kesavan G, Hans S, Ebersberger I, Brand M, Burger A, Reischauer S, Mosimann C, Stainier DYR. Endothelial versus pronephron fate decision is modulated by the transcription factors Cloche/Npas4l, Tal1, and Lmo2. SCIENCE ADVANCES 2022; 8:eabn2082. [PMID: 36044573 PMCID: PMC9432843 DOI: 10.1126/sciadv.abn2082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/11/2022] [Indexed: 05/17/2023]
Abstract
Endothelial specification is a key event during embryogenesis; however, when, and how, endothelial cells separate from other lineages is poorly understood. In zebrafish, Npas4l is indispensable for endothelial specification by inducing the expression of the transcription factor genes etsrp, tal1, and lmo2. We generated a knock-in reporter in zebrafish npas4l to visualize endothelial progenitors and their derivatives in wild-type and mutant embryos. Unexpectedly, we find that in npas4l mutants, npas4l reporter-expressing cells contribute to the pronephron tubules. Single-cell transcriptomics and live imaging of the early lateral plate mesoderm in wild-type embryos indeed reveals coexpression of endothelial and pronephron markers, a finding confirmed by creERT2-based lineage tracing. Increased contribution of npas4l reporter-expressing cells to pronephron tubules is also observed in tal1 and lmo2 mutants and is reversed in npas4l mutants injected with tal1 mRNA. Together, these data reveal that Npas4l/Tal1/Lmo2 regulate the fate decision between the endothelial and pronephron lineages.
Collapse
Affiliation(s)
- Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- DZL (German Center for Lung Research), partner site, 43, D-61231 Bad Nauheim
| | - Fréderike W. Riemslagh
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Stefan Guenther
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Karin D. Prummel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Gokul Kesavan
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Ingo Ebersberger
- Goethe University Frankfurt am Main, Institute of Cell Biology and Neuroscience, Frankfurt 60438, Germany
- Senckenberg Biodiversity and Climate Research Center (S-BIKF), Frankfurt 60325, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt 60325, Germany
| | - Michael Brand
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- DZL (German Center for Lung Research), partner site, 43, D-61231 Bad Nauheim
| |
Collapse
|
36
|
Koui Y, Ideue T, Boylan M, Anderson MJ, Osato M, Suda T, Yokomizo T, Mukouyama YS. Hepatic leukemia factor-expressing paraxial mesoderm cells contribute to the developing brain vasculature. Biol Open 2022; 11:276428. [PMID: 36017733 PMCID: PMC9493726 DOI: 10.1242/bio.059510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022] Open
Abstract
Recent genetic lineage tracing studies reveal heterogeneous origins of vascular endothelial cells and pericytes in the developing brain vasculature, despite classical experimental evidence for a mesodermal origin. Here we provide evidence through a genetic lineage tracing experiment that cephalic paraxial mesodermal cells give rise to endothelial cells and pericytes in the developing mouse brain. We show that Hepatic leukemia factor (Hlf) is transiently expressed by cephalic paraxial mesenchyme at embryonic day (E) 8.0-9.0 and the genetically-marked E8.0 Hlf-expressing cells mainly contribute to the developing brain vasculature. Interestingly, the genetically-marked E10.5 Hlf-expressing cells, which have been previously reported to contain embryonic hematopoietic stem cells, fail to contribute to the vascular cells. Combined, our genetic lineage tracing data demonstrate that a transient expression of Hlf marks a cephalic paraxial mesenchyme contributing to the developing brain vasculature.
Collapse
Affiliation(s)
- Yuta Koui
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Takako Ideue
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Michael Boylan
- Cancer and Developmental Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Motomi Osato
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Toshio Suda
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.,Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Tomomasa Yokomizo
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.,Microscopic and Developmental Anatomy, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
37
|
Ho VW, Grainger DE, Chagraoui H, Porcher C. Specification of the haematopoietic stem cell lineage: From blood-fated mesodermal angioblasts to haemogenic endothelium. Semin Cell Dev Biol 2022; 127:59-67. [PMID: 35125239 DOI: 10.1016/j.semcdb.2022.01.008] [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: 11/06/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Haematopoietic stem and progenitor cells emerge from specialized haemogenic endothelial cells in select vascular beds during embryonic development. Specification and commitment to the blood lineage, however, occur before endothelial cells are endowed with haemogenic competence, at the time of mesoderm patterning and production of endothelial cell progenitors (angioblasts). Whilst early blood cell fate specification has long been recognized, very little is known about the mechanisms that induce endothelial cell diversification and progressive acquisition of a blood identity by a subset of these cells. Here, we review the endothelial origin of the haematopoietic system and the complex developmental journey of blood-fated angioblasts. We discuss how recent technological advances will be instrumental to examine the diversity of the embryonic anatomical niches, signaling pathways and downstream epigenetic and transcriptional processes controlling endothelial cell heterogeneity and blood cell fate specification. Ultimately, this will give essential insights into the ontogeny of the cells giving rise to haematopoietic stem cells, that may aid in the development of novel strategies for their in vitro production for clinical purposes.
Collapse
Affiliation(s)
- Vivien W Ho
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - David E Grainger
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hedia Chagraoui
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Catherine Porcher
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Abstract
The lymphatic system, composed of initial and collecting lymphatic vessels as well as lymph nodes that are present in almost every tissue of the human body, acts as an essential transport system for fluids, biomolecules and cells between peripheral tissues and the central circulation. Consequently, it is required for normal body physiology but is also involved in the pathogenesis of various diseases, most notably cancer. The important role of tumor-associated lymphatic vessels and lymphangiogenesis in the formation of lymph node metastasis has been elucidated during the last two decades, whereas the underlying mechanisms and the relation between lymphatic and peripheral organ dissemination of cancer cells are incompletely understood. Lymphatic vessels are also important for tumor-host communication, relaying molecular information from a primary or metastatic tumor to regional lymph nodes and the circulatory system. Beyond antigen transport, lymphatic endothelial cells, particularly those residing in lymph node sinuses, have recently been recognized as direct regulators of tumor immunity and immunotherapy responsiveness, presenting tumor antigens and expressing several immune-modulatory signals including PD-L1. In this review, we summarize recent discoveries in this rapidly evolving field and highlight strategies and challenges of therapeutic targeting of lymphatic vessels or specific lymphatic functions in cancer patients.
Collapse
Affiliation(s)
- Lothar C Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Department of Biosciences, University of Milan, Milan, Italy
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Matsuoka RL, Buck LD, Vajrala KP, Quick RE, Card OA. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol Life Sci 2022; 79:372. [PMID: 35726097 PMCID: PMC9209386 DOI: 10.1007/s00018-022-04403-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Dynamic brain activity requires timely communications between the brain parenchyma and circulating blood. Brain-blood communication is facilitated by intricate networks of brain vasculature, which display striking heterogeneity in structure and function. This vascular cell heterogeneity in the brain is fundamental to mediating diverse brain functions and has long been recognized. However, the molecular basis of this biological phenomenon has only recently begun to be elucidated. Over the past century, various animal species and in vitro systems have contributed to the accumulation of our fundamental and phylogenetic knowledge about brain vasculature, collectively advancing this research field. Historically, dye tracer and microscopic observations have provided valuable insights into the anatomical and functional properties of vasculature across the brain, and these techniques remain an important approach. Additionally, recent advances in molecular genetics and omics technologies have revealed significant molecular heterogeneity within brain endothelial and perivascular cell types. The combination of these conventional and modern approaches has enabled us to identify phenotypic differences between healthy and abnormal conditions at the single-cell level. Accordingly, our understanding of brain vascular cell states during physiological, pathological, and aging processes has rapidly expanded. In this review, we summarize major historical advances and current knowledge on blood endothelial cell heterogeneity in the brain, and discuss important unsolved questions in the field.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Keerti P Vajrala
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.,Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| |
Collapse
|
40
|
Role of Transcriptional and Epigenetic Regulation in Lymphatic Endothelial Cell Development. Cells 2022; 11:cells11101692. [PMID: 35626729 PMCID: PMC9139870 DOI: 10.3390/cells11101692] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 12/04/2022] Open
Abstract
The lymphatic system is critical for maintaining the homeostasis of lipids and interstitial fluid and regulating the immune cell development and functions. Developmental anomaly-induced lymphatic dysfunction is associated with various pathological conditions, including lymphedema, inflammation, and cancer. Most lymphatic endothelial cells (LECs) are derived from a subset of endothelial cells in the cardinal vein. However, recent studies have reported that the developmental origin of LECs is heterogeneous. Multiple regulatory mechanisms, including those mediated by signaling pathways, transcription factors, and epigenetic pathways, are involved in lymphatic development and functions. Recent studies have demonstrated that the epigenetic regulation of transcription is critical for embryonic LEC development and functions. In addition to the chromatin structures, epigenetic modifications may modulate transcriptional signatures during the development or differentiation of LECs. Therefore, the understanding of the epigenetic mechanisms involved in the development and function of the lymphatic system can aid in the management of various congenital or acquired lymphatic disorders. Future studies must determine the role of other epigenetic factors and changes in mammalian lymphatic development and function. Here, the recent findings on key factors involved in the development of the lymphatic system and their epigenetic regulation, LEC origins from different organs, and lymphatic diseases are reviewed.
Collapse
|
41
|
Lenti E, Genovese L, Bianchessi S, Maurizio A, Sain SB, di Lillo A, Mattavelli G, Harel I, Bernassola F, Hehlgans T, Pfeffer K, Crosti M, Abrignani S, Evans SM, Sitia G, Guimarães-Camboa N, Russo V, van de Pavert SA, Garcia-Manteiga JM, Brendolan A. Fate mapping and scRNA sequencing reveal origin and diversity of lymph node stromal precursors. Immunity 2022; 55:606-622.e6. [PMID: 35358427 DOI: 10.1016/j.immuni.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2021] [Accepted: 03/03/2022] [Indexed: 11/25/2022]
Abstract
Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.
Collapse
Affiliation(s)
- Elisa Lenti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Genovese
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bianchessi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simona Baghai Sain
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia di Lillo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Greta Mattavelli
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Itamar Harel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Thomas Hehlgans
- Leibniz Institute of Immunotherapy (LIT), Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus Pfeffer
- Institute of Medical, Microbiology and Hospital Hygiene, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Mariacristina Crosti
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy; Department of Clinical Science and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Sylvia M Evans
- Skaggs School of Pharmacy, University of California at San Diego, La Jolla, CA 92093, USA
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nuno Guimarães-Camboa
- Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt 60590, Germany; German Center for Cardiovascular Research, Berlin (partner site Frankfurt Rhine-Main), Germany
| | - Vincenzo Russo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serge A van de Pavert
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | | | - Andrea Brendolan
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
42
|
Cardiac lymphatics: state of the art. Curr Opin Hematol 2022; 29:156-165. [PMID: 35220321 DOI: 10.1097/moh.0000000000000713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The beneficial role of cardiac lymphatics in health and disease has begun to be recognized, with both preclinical and clinical evidence demonstrating that lymphangiogenesis is activated in cardiovascular diseases. This review aims to summarize our current understanding of the regulation and impact of cardiac lymphatic remodeling during development and in adult life, highlighting emerging concepts regarding distinguishing traits of cardiac lymphatic endothelial cells (LEC). RECENT FINDINGS Genetic lineage-tracing and clonal analyses have revealed that a proportion of cardiac LECs originate from nonvenous sources. Further, these sources may vary between different regions of the heart, and could translate to differences in LEC sensitivity to molecular regulators. Several therapeutic approaches have been applied to investigate how lymphatics contribute to resolution of myocardial edema and inflammation in cardiovascular diseases. From these studies have emerged novel insights, notably concerning the cross-talk between lymphatics and cardiac interstitial cells, especially immune cells. SUMMARY Recent years have witnessed a significant expansion in our knowledge of the molecular characteristics and regulation of cardiac lymphatics. The current body of work is in support of critical contributions of cardiac lymphatics to maintain both fluid and immune homeostasis in the heart.
Collapse
|
43
|
Klaourakis K, Riley PR, Vieira JM. Quantitative Three-Dimensional Analysis of the Lymphatic Vasculature in the Postnatal Mouse Heart. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:171-181. [PMID: 35099736 DOI: 10.1007/978-1-0716-2059-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The development and maturation of the lymphatic vasculature are essential for organ function with disruption leading to severe phenotypes. For example, malfunction of cardiac lymphatics results in myocardial oedema, persistent inflammation and reduced cardiac output. Thus, it is important to study the process of cardiac lymphatic formation and growth from the early stages of fetal development to adulthood. In the murine heart the lymphatics continue to develop and expand postnatally with extensive growth and patterning occurring up to at least 2 weeks after birth. Here, we describe a protocol for whole-mount, multi-view imaging and quantification of lymphatic vessel parameters, including vessel junction number (i.e., branching density), vessel length, and number of vessel end points in the murine postnatal heart. This protocol is based on the use of reliable antibodies against key markers of lymphatic endothelial cells (LECs), specifically the glycoprotein lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1), the vascular endothelial growth factor receptor 3 (VEGFR3; also known as Fms-related receptor tyrosine kinase 4, FLT4), the mucin-type protein podoplanin (PDPN), and the co-receptor neuropilin 2 (NRP2). For imaging and quantitative analysis of the sub-epicardial network in neonatal hearts, VEGFR3 was selected given its exclusive expression in the lymphatic endothelium. In addition to LECs, LYVE1 expression was detected in tissue-resident macrophages, PDPN in the epicardium, and NRP2 in the autonomic nervous system of the heart. Overall, we characterized the expression patterns of commonly used lymphatic markers in the context of the neonatal heart and provide an image analysis pipeline that can be adapted to study other organs and systems (e.g., blood vasculature and nerve system).
Collapse
Affiliation(s)
- Konstantinos Klaourakis
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, and British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK
| | - Paul R Riley
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, and British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK.
| | - Joaquim Miguel Vieira
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, and British Heart Foundation-Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford, UK.
| |
Collapse
|
44
|
Harris NR, Nielsen NR, Pawlak JB, Aghajanian A, Rangarajan K, Serafin DS, Farber G, Dy DM, Nelson-Maney NP, Xu W, Ratra D, Hurr SH, Qian L, Scallan JP, Caron KM. VE-Cadherin Is Required for Cardiac Lymphatic Maintenance and Signaling. Circ Res 2022; 130:5-23. [PMID: 34789016 PMCID: PMC8756423 DOI: 10.1161/circresaha.121.318852] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The adherens protein VE-cadherin (vascular endothelial cadherin) has diverse roles in organ-specific lymphatic vessels. However, its physiological role in cardiac lymphatics and its interaction with lymphangiogenic factors has not been fully explored. We sought to determine the spatiotemporal functions of VE-cadherin in cardiac lymphatics and mechanistically elucidate how VE-cadherin loss influences prolymphangiogenic signaling pathways, such as adrenomedullin and VEGF (vascular endothelial growth factor)-C/VEGFR3 (vascular endothelial growth factor receptor 3) signaling. METHODS Cdh5flox/flox;Prox1CreERT2 mice were used to delete VE-cadherin in lymphatic endothelial cells across life stages, including embryonic, postnatal, and adult. Lymphatic architecture and function was characterized using immunostaining and functional lymphangiography. To evaluate the impact of temporal and functional regression of cardiac lymphatics in Cdh5flox/flox;Prox1CreERT2 mice, left anterior descending artery ligation was performed and cardiac function and repair after myocardial infarction was evaluated by echocardiography and histology. Cellular effects of VE-cadherin deletion on lymphatic signaling pathways were assessed by knockdown of VE-cadherin in cultured lymphatic endothelial cells. RESULTS Embryonic deletion of VE-cadherin produced edematous embryos with dilated cardiac lymphatics with significantly altered vessel tip morphology. Postnatal deletion of VE-cadherin caused complete disassembly of cardiac lymphatics. Adult deletion caused a temporal regression of the quiescent epicardial lymphatic network which correlated with significant dermal and cardiac lymphatic dysfunction, as measured by fluorescent and quantum dot lymphangiography, respectively. Surprisingly, despite regression of cardiac lymphatics, Cdh5flox/flox;Prox1CreERT2 mice exhibited preserved cardiac function, both at baseline and following myocardial infarction, compared with control mice. Mechanistically, loss of VE-cadherin leads to aberrant cellular internalization of VEGFR3, precluding the ability of VEGFR3 to be either canonically activated by VEGF-C or noncanonically transactivated by adrenomedullin signaling, impairing downstream processes such as cellular proliferation. CONCLUSIONS VE-cadherin is an essential scaffolding protein to maintain prolymphangiogenic signaling nodes at the plasma membrane, which are required for the development and adult maintenance of cardiac lymphatics, but not for cardiac function basally or after injury.
Collapse
Affiliation(s)
- Natalie R. Harris
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Natalie R. Nielsen
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - John B. Pawlak
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Amir Aghajanian
- Department of Medicine Division of Cardiology, University
of North Carolina at Chapel Hill; 160 Dental Circle, Chapel Hill, North Carolina,
USA 27599
| | - Krsna Rangarajan
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - D. Stephen Serafin
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Gregory Farber
- Department of Pathology and Laboratory Medicine, University
of North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina,
USA 27599,McAllister Heart Institute, University of North Carolina,
Chapel Hill, North Carolina, USA 27599
| | - Danielle M. Dy
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Nathan P. Nelson-Maney
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Wenjing Xu
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Disha Ratra
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Sophia H. Hurr
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University
of North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina,
USA 27599
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology,
University of South Florida, Tampa, Florida, USA 33612
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of
North Carolina at Chapel Hill; 111 Mason Farm Road, Chapel Hill, North Carolina, USA
27599
| |
Collapse
|
45
|
Oliver G. Lymphatic endothelial cell fate specification in the mammalian embryo: An historical perspective. Dev Biol 2021; 482:44-54. [PMID: 34915023 DOI: 10.1016/j.ydbio.2021.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
Development of the mammalian lymphatic vasculature is a stepwise process requiring the specification of lymphatic endothelial cell progenitors in the embryonic veins, and their subsequent budding to give rise to most of the mature lymphatic vasculature. In mice, formation of the lymphatic vascular network starts inside the cardinal vein at around E9.5 when a subpopulation of venous endothelial cells gets committed into the lymphatic lineage by their acquisition of Prox1 expression. Identification of critical genes regulating lymphatic development facilitated the detailed cellular and molecular characterization of some of the cellular and molecular mechanisms regulating the early steps leading to the formation of the mammalian lymphatic vasculature. A better understanding of basic aspects of early lymphatic development, and the availability of novel tools and animal models has been instrumental in the identification of important novel functional roles of this vasculature network.
Collapse
Affiliation(s)
- Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
46
|
Mao A, Zhang P, Zhang K, Kan H, He D, Han X, Wang Z, Tang C, Ma X. Endothelial TRPV4-eNOS coupling as a vital therapy target for treatment of hypertension. Br J Pharmacol 2021; 179:2297-2312. [PMID: 34822720 DOI: 10.1111/bph.15755] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Reduced nitric oxide (NO) level and activity are signs of endothelial dysfunction, which is important in mediating blood pressure up-regulation. Previously, we demonstrated that transient receptor potential channel V4 (TRPV4) could form functional complex with other proteins to mediate vasodilation in the Endothelial cells (ECs). But how TRPV4 interacts with the NO pathway in larger arteries requires further exploration. EXPERIMENTAL APPROACH We used single-cell RNA-sequencing to find the CD106+ TRPV4high NOS3high ECs. The TRPV4-eNOS interaction was verified by co-immunoprecipitation and Immunofluorescence resonance energy transfer (FRET), and their binding site was found by site-directed mutagenesis. Endothelium-specific TRPV4 knockout (TRPV4EC -/- ) mice were used to study the effect of the TRPV4-eNOS interaction on blood pressure. A small molecule, JNc-463 was designed through molecular docking technology. KEY RESULTS We uncovered CD106+ TRPV4high NOS3high ECs in the mouse aorta, which they could regulate vasodilation via a TRPV4-eNOS interaction, and they were essential to regulate blood pressure. The TRPV4-eNOS interaction markedly decreased during the process of hypertension. We further attempted to identify the molecules re-join the TRPV4-eNOS interaction and develop a small-molecule drug, JNc-463, which could increase the TRPV4-eNOS interaction to enhance vasodilation, and exert antihypertensive effects in mice. CONCLUSION AND IMPLICATIONS This is the first study integrating single-cell RNA-Seq, single-cell functional study and drug screening in aorta. We identified a subpopulation of CD106+ TRPV4high NOS3high ECs, in which an impaired TRPV4-eNOS interaction was important in the progress of hypertension and we designed a small molecule, JNc-463 to improve the impaired TRPV4-eNOS interaction in hypertension.
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Peng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiping Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
47
|
Tabrizi ZB, Ahmed NS, Horder JL, Storr SJ, Benest AV. Transcription Factor Control of Lymphatic Quiescence and Maturation of Lymphatic Neovessels in Development and Physiology. Front Physiol 2021; 12:672987. [PMID: 34795596 PMCID: PMC8593113 DOI: 10.3389/fphys.2021.672987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/30/2021] [Indexed: 11/25/2022] Open
Abstract
The lymphatic system is a vascular system comprising modified lymphatic endothelial cells, lymph nodes and other lymphoid organs. The system has diverse, but critical functions in both physiology and pathology, and forms an interface between the blood vascular and immune system. It is increasingly evident that remodelling of the lymphatic system occurs alongside remodelling of the blood microvascular system, which is now considered a hallmark of most pathological conditions as well as being critical for normal development. Much attention has focussed on how the blood endothelium undergoes phenotypic switching in development and disease, resulting in over two decades of research to probe the mechanisms underlying the resulting heterogeneity. The lymphatic system has received less attention, and consequently there are fewer descriptions of functional and molecular heterogeneity, but differential transcription factor activity is likely an important control mechanism. Here we introduce and discuss significant transcription factors of relevance to coordinating cellular responses during lymphatic remodelling as the lymphatic endothelium dynamically changes from quiescence to actively remodelling.
Collapse
Affiliation(s)
- Zarah B Tabrizi
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Nada S Ahmed
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Joseph L Horder
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, Centre for Cancer Sciences School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Andrew V Benest
- Endothelial Quiescence Group, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
48
|
Brouillard P, Witte MH, Erickson RP, Damstra RJ, Becker C, Quéré I, Vikkula M. Primary lymphoedema. Nat Rev Dis Primers 2021; 7:77. [PMID: 34675250 DOI: 10.1038/s41572-021-00309-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 11/09/2022]
Abstract
Lymphoedema is the swelling of one or several parts of the body owing to lymph accumulation in the extracellular space. It is often chronic, worsens if untreated, predisposes to infections and causes an important reduction in quality of life. Primary lymphoedema (PLE) is thought to result from abnormal development and/or functioning of the lymphatic system, can present in isolation or as part of a syndrome, and can be present at birth or develop later in life. Mutations in numerous genes involved in the initial formation of lymphatic vessels (including valves) as well as in the growth and expansion of the lymphatic system and associated pathways have been identified in syndromic and non-syndromic forms of PLE. Thus, the current hypothesis is that most cases of PLE have a genetic origin, although a causative mutation is identified in only about one-third of affected individuals. Diagnosis relies on clinical presentation, imaging of the structure and functionality of the lymphatics, and in genetic analyses. Management aims at reducing or preventing swelling by compression therapy (with manual drainage, exercise and compressive garments) and, in carefully selected cases, by various surgical techniques. Individuals with PLE often have a reduced quality of life owing to the psychosocial and lifelong management burden associated with their chronic condition. Improved understanding of the underlying genetic origins of PLE will translate into more accurate diagnosis and prognosis and personalized treatment.
Collapse
Affiliation(s)
- Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Marlys H Witte
- Department of Surgery, Neurosurgery, and Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Robert P Erickson
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Robert J Damstra
- VASCERN PPL European Reference Centre; Department of Dermatology, Phlebology and Lymphology, Nij Smellinghe Hospital, Drachten, Netherlands
| | | | - Isabelle Quéré
- Department of Vascular Medicine, Centre de référence des Maladies Lymphatiques et Vasculaires Rares, Inserm IDESP, CHU Montpellier, Université de Montpellier, Montpellier, France
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium. .,VASCERN VASCA European Reference Centre; Center for Vascular Anomalies, Division of Plastic Surgery, University Clinics Saint-Luc, University of Louvain, Brussels, Belgium. .,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), de Duve Institute, University of Louvain, Brussels, Belgium.
| |
Collapse
|
49
|
Donnan MD, Kenig-Kozlovsky Y, Quaggin SE. The lymphatics in kidney health and disease. Nat Rev Nephrol 2021; 17:655-675. [PMID: 34158633 DOI: 10.1038/s41581-021-00438-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
The mammalian vascular system consists of two networks: the blood vascular system and the lymphatic vascular system. Throughout the body, the lymphatic system contributes to homeostatic mechanisms by draining extravasated interstitial fluid and facilitating the trafficking and activation of immune cells. In the kidney, lymphatic vessels exist mainly in the kidney cortex. In the medulla, the ascending vasa recta represent a hybrid lymphatic-like vessel that performs lymphatic-like roles in interstitial fluid reabsorption. Although the lymphatic network is mainly derived from the venous system, evidence supports the existence of lymphatic beds that are of non-venous origin. Following their development and maturation, lymphatic vessel density remains relatively stable; however, these vessels undergo dynamic functional changes to meet tissue demands. Additionally, new lymphatic growth, or lymphangiogenesis, can be induced by pathological conditions such as tissue injury, interstitial fluid overload, hyperglycaemia and inflammation. Lymphangiogenesis is also associated with conditions such as polycystic kidney disease, hypertension, ultrafiltration failure and transplant rejection. Although lymphangiogenesis has protective functions in clearing accumulated fluid and immune cells, the kidney lymphatics may also propagate an inflammatory feedback loop, exacerbating inflammation and fibrosis. Greater understanding of lymphatic biology, including the developmental origin and function of the lymphatics and their response to pathogenic stimuli, may aid the development of new therapeutic agents that target the lymphatic system.
Collapse
Affiliation(s)
- Michael D Donnan
- Feinberg Cardiovascular & Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Susan E Quaggin
- Feinberg Cardiovascular & Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
50
|
Martin-Almedina S, Mortimer PS, Ostergaard P. Development and physiological functions of the lymphatic system: insights from human genetic studies of primary lymphedema. Physiol Rev 2021; 101:1809-1871. [PMID: 33507128 DOI: 10.1152/physrev.00006.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Primary lymphedema is a long-term (chronic) condition characterized by tissue lymph retention and swelling that can affect any part of the body, although it usually develops in the arms or legs. Due to the relevant contribution of the lymphatic system to human physiology, while this review mainly focuses on the clinical and physiological aspects related to the regulation of fluid homeostasis and edema, clinicians need to know that the impact of lymphatic dysfunction with a genetic origin can be wide ranging. Lymphatic dysfunction can affect immune function so leading to infection; it can influence cancer development and spread, and it can determine fat transport so impacting on nutrition and obesity. Genetic studies and the development of imaging techniques for the assessment of lymphatic function have enabled the recognition of primary lymphedema as a heterogenic condition in terms of genetic causes and disease mechanisms. In this review, the known biological functions of several genes crucial to the development and function of the lymphatic system are used as a basis for understanding normal lymphatic biology. The disease conditions originating from mutations in these genes are discussed together with a detailed clinical description of the phenotype and the up-to-date knowledge in terms of disease mechanisms acquired from in vitro and in vivo research models.
Collapse
Affiliation(s)
- Silvia Martin-Almedina
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Peter S Mortimer
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
- Dermatology and Lymphovascular Medicine, St. George's Universities NHS Foundation Trust, London, United Kingdom
| | - Pia Ostergaard
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|