1
|
Kannan S, Phan TT, Creed HA, Reyna AJ, Baranwal G, Rich AL, Weiss DL, Rutkowski JM. Therapeutically Induced Lymphangiogenesis Is Ineffective in Resolving Established Kidney Disease in Mice. KIDNEY360 2025; 6:509-520. [PMID: 39689345 DOI: 10.34067/kid.0000000671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024]
Abstract
Key Points
CKD is a state of unresolved kidney inflammation.Lymphatic vessels and lymphangiogenesis regulate inflammation, and thus, more lymphatics could potentially resolve inflammation and CKD progression.Induction of kidney-specific lymphangiogenesis in three mouse CKD models did not improve kidney function and has the potential to worsen CKD.
Background
CKD counts AKI as one of its many underlying causes. Lymphatic vessels are important in modulating inflammation postinjury. Manipulating lymphatic vessel expansion thus has the potential to alter CKD progression. Previously, we demonstrated that renal lymphatic expansion before injury reduced CKD progression after an AKI. Here, we test whether inducing lymphangiogenesis affects established CKD.
Methods
After CKD progression, kidney lymphatics were expanded by transgenic induction of kidney-specific overexpression of vascular endothelial growth factor-D in aristolochic acid (AA) nephropathy and cisplatin injury aggravated with chronic high phosphate diet (CisPi) models or by infusion of kidney-targeting nanoparticles loaded with the vascular endothelial growth factor receptor-3 specific ligand vascular endothelial growth factor-C C156S in a progressive proteinuria (POD) model. Renal fibrosis and lymphatic density were determined by picrosirius red staining and immunofluorescence, respectively. Renal function was assessed by creatinine clearance rate, serum creatinine, BUN, and urinary albumin-creatinine ratio. Renal proinflammatory and fibrotic markers expression were measured by quantitative RT-PCR.
Results
Kidney-specific overexpression of vascular endothelial growth factor-D+ mice demonstrated expanded renal lymphatics, while nanoparticles treatment minimally expanded lymphatics. In neither the AA nor POD model did lymphangiogenesis improve renal function or fibrosis. AA mice showed decreased Tgfb1 expression and POD mice showed increased Col4a1 expression. Expansion worsened function in CisPi CKD and increased fibrosis. CisPi kidneys also demonstrated increased expression of Mcp-1, Il1b, Col1a1, and Tgfb1 and increased macrophage numbers.
Conclusions
Therapeutically induced lymphatic expansion is ineffective in resolving established CKD and has the potential to further worsen CKD progression.
Collapse
Affiliation(s)
- Saranya Kannan
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, Texas
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Zhong J, Liu J, Mutchler AL, Yang H, Kirabo A, Shelton EL, Kon V. Moving toward a better understanding of renal lymphatics: challenges and opportunities. Pediatr Nephrol 2025:10.1007/s00467-025-06692-7. [PMID: 39899153 DOI: 10.1007/s00467-025-06692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
The development of lymphatic-specific markers has enabled detailed visualization of the lymphatic vascular network that has greatly enhanced our ability to explore this often-overlooked system. Lymphatics remove fluid, solutes, macromolecules, and cells from the interstitium and return them to circulation. The kidneys have lymphatics. As in other organs, the kidney lymphatic vessels are highly sensitive to changes in the local microenvironment. The sensitivity to its milieu may be especially relevant in kidneys because they are central in regulating fluid homeostasis and clearance of metabolites delivered into and eliminated from the renal interstitial compartment. Numerous physiologic conditions and diseases modify the renal interstitial volume, pressure, and composition that can, in turn, influence the growth and function of the renal lymphatics. The impact of the renal microenvironment is further heightened by the fact that kidneys are encapsulated. This review considers the development, structure, and function of the renal lymphatic vessels and explores how factors within the kidney interstitial compartment modify their structure and functionality. Moreover, although currently there are no pharmaceutical agents that specifically target the lymphatic network, we highlight several medications currently used in children with kidney disease and hypertension that have significant but underappreciated effects on lymphatics.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jing Liu
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Nephrology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Ashley L Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Haichun Yang
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elaine L Shelton
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA
| | - Valentina Kon
- Department of Pediatrics, Division of Pediatric Nephrology, Vanderbilt University Medical Center, Medical Center North C-4204, 1161 21st Avenue South, Nashville, TN, 37232-2584, USA.
| |
Collapse
|
3
|
Mutchler AL, Zhong J, Yang HC, Zhao S, Crescenzi R, Taylor S, Rao RL, Shelton EL, Kirabo A, Kon V. ET-3/ETBR Mediates Na +-Activated Immune Signaling and Kidney Lymphatic Dynamics. Circ Res 2025; 136:194-208. [PMID: 39676651 PMCID: PMC11800760 DOI: 10.1161/circresaha.124.324890] [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: 05/08/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Lymphatic collecting vessels in the kidney are critical in clearing interstitial fluid, macromolecules, and infiltrating immune cells. Dysfunction of the lymphatic vessels can disrupt this process and exacerbate injury-associated inflammation in many disease conditions. We previously found that sodium accumulates within the kidney interstitium during proteinuric kidney injury and elevated sodium environments stimulate isolevuglandin production in antigen-presenting cells, stimulating T cells, and modulating inflammatory responses. In the present study, we investigated whether proteinuric injury increases production of isolevuglandin-adduct formation in antigen-presenting cells, their effects on lymphatic endothelial cells (LECs), and the role of the ET-3 (endothelin-3)/ETBR (endothelin type B receptor) on lymphatic vessel function. METHODS We used a mouse model of nephrotoxin-induced proteinuric injury to show that proteinuric injury expanded the kidney lymphatic network and to immunophenotype the infiltrating immune cells. To determine mechanisms, we analyzed the interaction of migratory immune cells and LECs using an in vitro transwell migration assay, bulk RNA sequencing, and flow cytometric analysis. To determine the effect of ET-3/ETBR axis on lymphatic vessel contractility, we analyzed microdissected lymphangions utilizing a vessel perfusion chamber. RESULTS We found that animals with proteinuric injury have increased kidney lymphangiogenesis, isolevuglandin-producing dendritic cells, and IFN (interferon)-γ-producing CD4+T cells. The sodium avid environment present in kidney injury enhances the interaction between LECs and migratory antigen-presenting cells and LEC production of isolevuglandin-adducts. Elevated sodium environment-induced isolevuglandin-adduct formation facilitates the ET-3/ETBR communication between LECs and dendritic cells. In addition, the ET-3/ETBR axis modulates lymphatic collecting vessel pumping dynamics. CONCLUSIONS These findings reveal a novel mechanism linking the isolevuglandin-mediated ET-3/ETBR axis with LECs and infiltrating dendritic cells. ET-3/ETBR signaling in lymphatic vessel dynamics is a novel pathogenic component and a possible therapeutic target in kidney disease.
Collapse
Affiliation(s)
- Ashley L. Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jianyong Zhong
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rachelle Crescenzi
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Shannon Taylor
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Roy L. Rao
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elaine L. Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
4
|
Du YX, Li X, Ji SW, Niu N. Hypertension toxicity of VEGFR-TKIs in cancer treatment: incidence, mechanisms, and management strategies. Arch Toxicol 2025; 99:67-81. [PMID: 39347999 DOI: 10.1007/s00204-024-03874-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) are a class of targeted anticancer agents that include pazopanib, sunitinib, axitinib, and others. Currently, VEGFR-TKIs are widely used in the clinical treatment of various tumors, which can prolong patients' survival and even cure tumors. However, the use of VEGFR-TKIs is frequently associated with the occurrence of cardiovascular adverse events, with hypertension being the most prevalent. Hypertension and its complications can significantly impact the prognosis of patients, potentially jeopardizing their lives and resulting in the reduction or even cessation of treatment in severe cases. This review addresses the incidence of hypertension due to VEGFR-TKIs, mechanisms of toxicity, management strategies, and future research directions. In addition, hypertension due to VEGFR-TKIs may be associated with salt sensitivity, and possible mechanisms of hypertensive side effects are vasodilator imbalance, decreased capillary density, renal injury, impaired endothelial function due to oxidative stress, decreased lymphatic vascular density, and "off-target effect". A comprehensive understanding of hypertension toxicity due to cancer treatment with VEGFR-TKIs, can enhance clinical practice, thereby improving the prognostic outcomes of VEGFR-TKIs in oncology patients.
Collapse
Affiliation(s)
- Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Si-Wen Ji
- Office of Academic Affairs, North Sichuan Medical College, Nanchong, 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
5
|
Márquez-Nogueras KM, Elliott B, Thuo P, DiNello E, Knutila RM, Fritzmann GE, Vuchkovska V, Flury S, Willis M, Chapman AB, Cao Q, Barefield DY, Kuo IY. Cardiac Localized Polycystin-2 in the Natriuretic Peptide Signaling Pathway and Hypertension. J Am Soc Nephrol 2025; 36:34-47. [PMID: 39302726 PMCID: PMC11706566 DOI: 10.1681/asn.0000000000000490] [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: 01/04/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Key Points Cardiac localized polycystin facilitates natriuretic peptide signaling pathways. Hypertension associated with autosomal dominant polycystic kidney disease may arise from impaired cardiac natriuretic peptide signaling. Background Hypertension is seen in 70% of patients with autosomal dominant polycystic kidney disease by age of 30 years before decline in kidney function. However, cardiac origins of hypertension, such as the natriuretic peptide signaling pathway, have not been fully investigated. We hypothesized that cardiomyocyte localized polycystin proteins contribute to production of natriuretic peptides, and loss of this pathway would contribute to hypertension. Methods Telemetry, echocardiography, and a molecular analysis of the natriuretic peptide pathway from left ventricular tissue of cardiomyocyte specific knockout models of polycystin-2 (cPC2-KO) mice and Cre control littermates were conducted. Complementary studies were conducted in ex vivo murine hearts, engineered heart tissue with human iPSCs driven into cardiomyocytes with CRISPR/Cas9 knockout of PKD2 and in in vitro cell lines. Results cPC2-KO mice demonstrated diurnal hypertension. Circulating atrial natriuretic peptide (ANP) and brain natriuretic peptide were unchanged between cPC2-KO and Cre mice. Analysis of the pathways involved in production, maturation, and activity of natriuretic peptides identified decreased transcription of chromogranin B, PCSK6, NPR1, and NFAT genes in cPC2-KOs. Human iPSC-derived cardiomyocytes with PC2-KO failed to produce ANP. Re-expression of polycystin-2 in a myoblast cell line, but not pathogenic forms of polycystin-2, restored ANP production. Conclusions Natriuretic peptide production required cardiac localized polycystin-2, and loss of this pathway may contribute to the development of hypertension in autosomal dominant polycystic kidney disease. Podcast This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2024_10_08_ASN0000000000000490.mp3
Collapse
Affiliation(s)
- Karla M. Márquez-Nogueras
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Brandon Elliott
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Paula Thuo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Elisabeth DiNello
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Ryne M. Knutila
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Geena E. Fritzmann
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - Virdjinija Vuchkovska
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Sarah Flury
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Monte Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Arlene B. Chapman
- Section of Nephrology, Biological Sciences Division, Department of Medicine and Institute for Translational Medicine, University of Chicago, Chicago, Illinois
| | - Quan Cao
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - David Y. Barefield
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| | - Ivana Y. Kuo
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
- Stritch School of Medicine, Cardiovascular Research Institute, Loyola University Chicago, Maywood, Illinois
| |
Collapse
|
6
|
Lu X, Ma K, Ren J, Peng H, Wang J, Wang X, Nasser MI, Liu C. The immune regulatory role of lymphangiogenesis in kidney disease. J Transl Med 2024; 22:1053. [PMID: 39578812 PMCID: PMC11583545 DOI: 10.1186/s12967-024-05859-4] [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: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
The renal lymphatic system is critical for maintaining kidney homeostasis and regulating the immune response inside the kidney. In various kidney pathological situations, the renal lymphatic network experiences lymphangiogenesis, which is defined as the creation of new lymphatic vessels. Kidney lymphangiogenesis controls immunological response inside the kidney by controlling lymphatic flow, immune cell trafficking, and immune cell regulation. Ongoing study reveals lymphangiogenesis's different architecture and functions in numerous tissues and organs. New research suggests that lymphangiogenesis in kidney disorders may regulate the renal immune response in various ways. The flexibility of lymphatic endothelial cells (LECs) improves the kidney's immunological regulatory function of lymphangiogenesis. Furthermore, current research has shown disparate findings regarding its impact on distinct renal diseases, resulting in contradictory outcomes even within the same kidney condition. The fundamental causes of the various effects of lymphangiogenesis on renal disorders remain unknown. In this thorough review, we explore the dual impacts of renal lymphangiogenesis on several kidney pathologies, with a particular emphasis on existing empirical data and new developments in understanding its immunological regulatory function in kidney disease. An improved understanding of the immunological regulatory function of lymphangiogenesis in kidney diseases might help design novel medicines targeting lymphatics to treat kidney pathologies.
Collapse
Affiliation(s)
- Xiangheng Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jia Wang
- General Practice Center, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Moussa Ide Nasser
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, 510100, Guangdong, China.
| | - Chi Liu
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, China.
| |
Collapse
|
7
|
McDermott JG, Goodlett BL, Creed HA, Navaneethabalakrishnan S, Rutkowski JM, Mitchell BM. Inflammatory Alterations to Renal Lymphatic Endothelial Cell Gene Expression in Mouse Models of Hypertension. Kidney Blood Press Res 2024; 49:588-604. [PMID: 38972305 PMCID: PMC11345939 DOI: 10.1159/000539721] [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: 10/16/2023] [Accepted: 06/02/2024] [Indexed: 07/09/2024] Open
Abstract
INTRODUCTION Hypertension (HTN) is a major cardiovascular disease that can cause and be worsened by renal damage and inflammation. We previously reported that renal lymphatic endothelial cells (LECs) increase in response to HTN and that augmenting lymphangiogenesis in the kidneys reduces blood pressure and renal pro-inflammatory immune cells in mice with various forms of HTN. Our aim was to evaluate the specific changes that renal LECs undergo in HTN. METHODS We performed single-cell RNA sequencing. Using the angiotensin II-induced and salt-sensitive mouse models of HTN, we isolated renal CD31+ and podoplanin+ cells. RESULTS Sequencing of these cells revealed three distinct cell types with unique expression profiles, including LECs. The number and transcriptional diversity of LECs increased in samples from mice with HTN, as demonstrated by 597 differentially expressed genes (p < 0.01), 274 significantly enriched pathways (p < 0.01), and 331 regulons with specific enrichment in HTN LECs. These changes demonstrate a profound inflammatory response in renal LECs in HTN, leading to an increase in genes and pathways associated with inflammation-driven growth and immune checkpoint activity in LECs. CONCLUSION These results reinforce and help to further explain the benefits of renal LECs and lymphangiogenesis in HTN.
Collapse
Affiliation(s)
- Justin G. McDermott
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Heidi A. Creed
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | | | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807
| |
Collapse
|
8
|
Fowler JWM, Song L, Tam K, Roth Flach RJ. Targeting lymphatic function in cardiovascular-kidney-metabolic syndrome: preclinical methods to analyze lymphatic function and therapeutic opportunities. Front Cardiovasc Med 2024; 11:1412857. [PMID: 38915742 PMCID: PMC11194411 DOI: 10.3389/fcvm.2024.1412857] [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: 04/05/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
The lymphatic vascular system spans nearly every organ in the body and serves as an important network that maintains fluid, metabolite, and immune cell homeostasis. Recently, there has been a growing interest in the role of lymphatic biology in chronic disorders outside the realm of lymphatic abnormalities, lymphedema, or oncology, such as cardiovascular-kidney-metabolic syndrome (CKM). We propose that enhancing lymphatic function pharmacologically may be a novel and effective way to improve quality of life in patients with CKM syndrome by engaging multiple pathologies at once throughout the body. Several promising therapeutic targets that enhance lymphatic function have already been reported and may have clinical benefit. However, much remains unclear of the discreet ways the lymphatic vasculature interacts with CKM pathogenesis, and translation of these therapeutic targets to clinical development is challenging. Thus, the field must improve characterization of lymphatic function in preclinical mouse models of CKM syndrome to better understand molecular mechanisms of disease and uncover effective therapies.
Collapse
Affiliation(s)
| | | | | | - Rachel J. Roth Flach
- Internal Medicine Research Unit, Pfizer Research and Development, Cambridge, MA, United States
| |
Collapse
|
9
|
Cooper STE, Lokman AB, Riley PR. Role of the Lymphatics in Cardiac Disease. Arterioscler Thromb Vasc Biol 2024; 44:1181-1190. [PMID: 38634279 DOI: 10.1161/atvbaha.124.319854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Cardiovascular diseases remain the largest cause of death worldwide with recent evidence increasingly attributing the development and progression of these diseases to an exacerbated inflammatory response. As a result, significant research is now focused on modifying the immune environment to prevent the disease progression. This in turn has highlighted the lymphatic system in the pathophysiology of cardiovascular diseases owing, in part, to its established function in immune cell surveillance and trafficking. In this review, we highlight the role of the cardiac lymphatic system and its potential as an immunomodulatory therapeutic target in selected cardiovascular diseases.
Collapse
Affiliation(s)
- Susanna T E Cooper
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Adam B Lokman
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Paul R Riley
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| |
Collapse
|
10
|
Bai L, Wang Y, Du S, Si Y, Chen L, Li L, Li Y. Lymphangiogenesis: A new strategy for heart disease treatment (Review). Int J Mol Med 2024; 53:35. [PMID: 38391009 PMCID: PMC10903933 DOI: 10.3892/ijmm.2024.5359] [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: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Heart disease remains a global health challenge, contributing notably to morbidity and mortality. The lymphatic vasculature, an integral component of the cardiovascular system, plays a crucial role in regulating essential physiological processes, including fluid balance, transportation of extravasated proteins and immune cell trafficking, all of which are important for heart function. Through thorough scientometric analysis and extensive research, the present review identified lymphangiogenesis as a hotspot in cardiovascular disease research, and the mechanisms underlying impaired cardiac lymphangiogenesis and inadequate lymph drainage in various cardiovascular diseases are discussed. Furthermore, the way used to improve lymphangiogenesis to effectively regulate a variety of heart diseases and associated signaling pathways was investigated. Notably, the current review also highlights the impact of Traditional Chinese Medicine (TCM) on lymphangiogenesis, aiming to establish a clinical basis for the potential of TCM to improve cardiovascular diseases by promoting lymphangiogenesis.
Collapse
Affiliation(s)
- Liding Bai
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yanyan Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Siqi Du
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yumeng Si
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Lu Chen
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Lin Li
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yuhong Li
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin 301617, P.R. China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
11
|
Rossitto G, Bertoldi G, Rutkowski JM, Mitchell BM, Delles C. Sodium, Interstitium, Lymphatics and Hypertension-A Tale of Hydraulics. Hypertension 2024; 81:727-737. [PMID: 38385255 PMCID: PMC10954399 DOI: 10.1161/hypertensionaha.123.17942] [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] [Indexed: 02/23/2024]
Abstract
Blood pressure is regulated by vascular resistance and intravascular volume. However, exchanges of electrolytes and water between intra and extracellular spaces and filtration of fluid and solutes in the capillary beds blur the separation between intravascular, interstitial and intracellular compartments. Contemporary paradigms of microvascular exchange posit filtration of fluids and solutes along the whole capillary bed and a prominent role of lymphatic vessels, rather than its venous end, for their reabsorption. In the last decade, these concepts have stimulated greater interest in and better understanding of the lymphatic system as one of the master regulators of interstitial volume homeostasis. Here, we describe the anatomy and function of the lymphatic system and focus on its plasticity in relation to the accumulation of interstitial sodium in hypertension. The pathophysiological relevance of the lymphatic system is exemplified in the kidneys, which are crucially involved in the control of blood pressure, but also hypertension-mediated cardiac damage. Preclinical modulation of the lymphatic reserve for tissue drainage has demonstrated promise, but has also generated conflicting results. A better understanding of the hydraulic element of hypertension and the role of lymphatics in maintaining fluid balance can open new approaches to prevent and treat hypertension and its consequences, such as heart failure.
Collapse
Affiliation(s)
- Giacomo Rossitto
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
- Emergency Medicine and Hypertension, DIMED; Università degli Studi di Padova, Italy
| | - Giovanni Bertoldi
- Emergency Medicine and Hypertension, DIMED; Università degli Studi di Padova, Italy
| | | | - Brett M. Mitchell
- Dept. of Medical Physiology, Texas A&M University School of Medicine, USA
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| |
Collapse
|
12
|
Wu Q, Meng W, Zhu B, Chen X, Fu J, Zhao C, Liu G, Luo X, Lv Y, Zhao W, Wang F, Hu S, Zhang S. VEGFC ameliorates salt-sensitive hypertension and hypertensive nephropathy by inhibiting NLRP3 inflammasome via activating VEGFR3-AMPK dependent autophagy pathway. Cell Mol Life Sci 2023; 80:327. [PMID: 37837447 PMCID: PMC11072217 DOI: 10.1007/s00018-023-04978-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/29/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Salt-sensitivity hypertension (SSHTN) is an independent predictor for cardiovascular mortality. VEGFC has been reported to be a protective role in SSHTN and hypertensive kidney injury. However, the underlying mechanisms remain largely unclear. The current study aimed to explore the protective effects and mechanisms of VEGFC against SSHTN and hypertensive nephropathy. Here, we reported that VEGFC attenuated high blood pressure as well as protected against renal inflammation and fibrosis in SSHTN mice. Moreover, VEGFC suppressed the activation of renal NLRP3 inflammasome in SSHTN mice. In vitro, we found VEGFC inhibited NLRP3 inflammasome activation, meanwhile, upregulated autophagy in high-salt-induced macrophages, while these effects were reversed by an autophagy inhibitor 3MA. Furthermore, in vivo, 3MA pretreatment weakened the protective effects of VEGFC on SSHTN and hypertensive nephropathy. Mechanistically, VEGF receptor 3 (VEGFR3) kinase domain activated AMPK by promoting the phosphorylation at Thr183 via binding to AMPK, thus enhancing autophagy activity in the context of high-salt-induced macrophages. These findings indicated that VEGFC inhibited NLRP3 inflammasome activation by promoting VEGFR3-AMPK-dependent autophagy pathway in high-salt-induced macrophages, which provided a mechanistic basis for the therapeutic target in SSHTN and hypertensive kidney injury.
Collapse
Affiliation(s)
- Qiuwen Wu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Wei Meng
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Bin Zhu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Xi Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jiaxin Fu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Chunyu Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Gang Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xing Luo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Wenqi Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Sining Hu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| | - Shuo Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
13
|
Bertoldi G, Caputo I, Calò L, Rossitto G. Lymphatic vessels and the renin-angiotensin-system. Am J Physiol Heart Circ Physiol 2023; 325:H837-H855. [PMID: 37565265 DOI: 10.1152/ajpheart.00023.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
The lymphatic system is an integral part of the circulatory system and plays an important role in the fluid homeostasis of the human body. Accumulating evidence has recently suggested the involvement of lymphatic dysfunction in the pathogenesis of cardio-reno-vascular (CRV) disease. However, how the sophisticated contractile machinery of lymphatic vessels is modulated and, possibly impaired in CRV disease, remains largely unknown. In particular, little attention has been paid to the effect of the renin-angiotensin-system (RAS) on lymphatics, despite the high concentration of RAS mediators that these tissue-draining vessels are exposed to and the established role of the RAS in the development of classic microvascular dysfunction and overt CRV disease. We herein review recent studies linking RAS to lymphatic function and/or plasticity and further highlight RAS-specific signaling pathways, previously shown to drive adverse arterial remodeling and CRV organ damage that have potential for direct modulation of the lymphatic system.
Collapse
Affiliation(s)
- Giovanni Bertoldi
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
- Nephrology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Ilaria Caputo
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Lorenzo Calò
- Nephrology Unit, DIMED, Università degli Studi di Padova, Padova, Italy
| | - Giacomo Rossitto
- Emergency and Hypertension Unit, DIMED, Università degli Studi di Padova, Padova, Italy
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
14
|
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: 12] [Impact Index Per Article: 6.0] [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
|
15
|
Baker ML, Cantley LG. The Lymphatic System in Kidney Disease. KIDNEY360 2023; 4:e841-e850. [PMID: 37019177 PMCID: PMC10371377 DOI: 10.34067/kid.0000000000000120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 04/07/2023]
Abstract
The high-capacity vessels of the lymphatic system drain extravasated fluid and macromolecules from nearly every part of the body. However, far from merely a passive conduit for fluid removal, the lymphatic system also plays a critical and active role in immune surveillance and immune response modulation through the presentation of fluid, macromolecules, and trafficking immune cells to surveillance cells in regional draining lymph nodes before their return to the systemic circulation. The potential effect of this system in numerous disease states both within and outside of the kidney is increasingly being explored for their therapeutic potential. In the kidneys, the lymphatics play a critical role in both fluid and macromolecule removal to maintain oncotic and hydrostatic pressure gradients for normal kidney function, as well as in shaping kidney immunity, and potentially in balancing physiological pathways that promote healthy organ maintenance and responses to injury. In many states of kidney disease, including AKI, the demand on the preexisting lymphatic network increases for clearance of injury-related tissue edema and inflammatory infiltrates. Lymphangiogenesis, stimulated by macrophages, injured resident cells, and other drivers in kidney tissue, is highly prevalent in settings of AKI, CKD, and transplantation. Accumulating evidence points toward lymphangiogenesis being possibly harmful in AKI and kidney allograft rejection, which would potentially position lymphatics as another target for novel therapies to improve outcomes. However, the extent to which lymphangiogenesis is protective rather than maladaptive in the kidney in various settings remains poorly understood and thus an area of active research.
Collapse
Affiliation(s)
- Megan L Baker
- Section of Nephrology, Yale School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
16
|
Chignalia AZ. Novel avenues to control blood pressure: targeting the renal lymphatic system. Clin Sci (Lond) 2023; 137:597-601. [PMID: 37075761 PMCID: PMC10116342 DOI: 10.1042/cs20220775] [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: 01/03/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/21/2023]
Abstract
Hypertension is associated with the activation of the immune and lymphatic systems as well as lymphangiogenesis. The changes in the lymphatic system are considered an adaptive response to mitigate the deleterious effects of immune and inflammatory cells on the cardiovascular system. In the article recently published in Clinical Science by Goodlett and collaborators, evidence is shown that inducing renal lymphangiogenesis after the establishment of hypertension in mice is an effective maneuver to reduce systemic arterial blood pressure. In this commentary, we will briefly review what is known about the relationship between the activation of the immune and lymphatic systems, and the resulting effects on systemic blood pressure, summarize the findings published by Goodlett and collaborators, and discuss the impact of their findings on the field.
Collapse
Affiliation(s)
- Andreia Zago Chignalia
- Department of Anesthesiology, The University of Arizona, College of Medicine - Tucson, AZ, U.S.A
- Department of Physiology, The University of Arizona, College of Medicine - Tucson, AZ, U.S.A
- Department of Pharmacology and Toxicology, The University of Arizona, College of Pharmacy - Tucson, AZ, U.S.A
- Sarver Heart Center, The University of Arizona, College of Medicine - Tucson, AZ, U.S.A
| |
Collapse
|
17
|
Liu X, Cui K, Wu H, Li KS, Peng Q, Wang D, Cowan DB, Dixon JB, Sathish Srinivasan R, 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: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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 (X.L.)
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | | | - Kathryn S Li
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | - Qianman Peng
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | - Donghai Wang
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | - J Brandon Dixon
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta (J.B.D.)
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.S.S.)
| | - Diane R Bielenberg
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| | - Kaifu Chen
- Department of Cardiology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, MA (K.C.)
| | - Da-Zhi Wang
- USF Heart Institute, Center for Regenerative Medicine, College of Medicine Internal Medicine, University of South Florida, Tampa (D.Z.W.)
| | - Yabing Chen
- Department of Pathology, Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham (Y.C.)
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, MA (K.C., K.S.L., Q.P., D.W., D.B.C., D.R.B., H.C.)
| |
Collapse
|
18
|
Goodlett BL, Balasubbramanian D, Navaneethabalakrishnan S, Love SE, Luera EM, Konatham S, Chiasson VL, Wedgeworth S, Rutkowski JM, Mitchell BM. Genetically inducing renal lymphangiogenesis attenuates hypertension in mice. Clin Sci (Lond) 2022; 136:1759-1772. [PMID: 36345993 PMCID: PMC10586591 DOI: 10.1042/cs20220547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Hypertension (HTN) is associated with renal proinflammatory immune cell infiltration and increased sodium retention. We reported previously that renal lymphatic vessels, which are responsible for trafficking immune cells from the interstitial space to draining lymph nodes, increase in density under hypertensive conditions. We also demonstrated that augmenting renal lymphatic density can prevent HTN in mice. Whether renal lymphangiogenesis can treat HTN in mice is unknown. We hypothesized that genetically inducing renal lymphangiogenesis after the establishment of HTN would attenuate HTN in male and female mice from three different HTN models. METHODS Mice with inducible kidney-specific overexpression of VEGF-D (KidVD) experience renal lymphangiogenesis upon doxycycline administration. HTN was induced in KidVD+ and KidVD- mice by subcutaneous release of angiotensin II, administration of the nitric oxide synthase inhibitor L-NAME, or consumption of a 4% salt diet following a L-NAME priming and washout period. After a week of HTN stimuli treatment, doxycycline was introduced. Systolic blood pressure (SBP) readings were taken weekly. Kidney function was determined from urine and serum measures. Kidneys were processed for RT-qPCR, flow cytometry, and imaging. RESULTS Mice that underwent renal-specific lymphangiogenesis had significantly decreased SBP and renal proinflammatory immune cells. Additionally, renal lymphangiogenesis was associated with a decrease in sodium transporter expression and increased fractional excretion of sodium, indicating improved sodium handling efficiency. CONCLUSIONS These findings demonstrate that augmenting renal lymphangiogenesis can treat HTN in male and female mice by improving renal immune cell trafficking and sodium handling.
Collapse
Affiliation(s)
- Bethany L Goodlett
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | | | | | - Sydney E Love
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Emily M Luera
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Sunitha Konatham
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Valorie L Chiasson
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Sophie Wedgeworth
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Joseph M Rutkowski
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| |
Collapse
|
19
|
Rossitto G, Delles C. Mechanisms of sodium-mediated injury in cardiovascular disease: old play, new scripts. FEBS J 2022; 289:7260-7273. [PMID: 34355504 DOI: 10.1111/febs.16155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 01/13/2023]
Abstract
There is a strong association between salt intake and cardiovascular diseases, particularly hypertension, on the population level. The mechanisms that explain this association remain incompletely understood and appear to extend beyond blood pressure. In this review, we describe some of the 'novel' roles of Na+ in cardiovascular health and disease: energetic implications of sodium handling in the kidneys; local accumulation in tissue; fluid dynamics; and the role of the microvasculature, with particular focus on the lymphatic system. We describe the interplay between these factors that involves body composition, metabolic signatures, inflammation and composition of the extracellular and intracellular milieus.
Collapse
Affiliation(s)
- Giacomo Rossitto
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK.,Department of Medicine (DIMED), University of Padua, Italy
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| |
Collapse
|
20
|
Thowsen IM, Reikvam T, Skogstrand T, Samuelsson AM, Müller DN, Tenstad O, Alitalo K, Karlsen T, Wiig H. Genetic Engineering of Lymphangiogenesis in Skin Does Not Affect Blood Pressure in Mouse Models of Salt-Sensitive Hypertension. Hypertension 2022; 79:2451-2462. [DOI: 10.1161/hypertensionaha.122.19777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background:
Recent studies have indicated that sodium storage is influenced by macrophages that secrete VEGF-C (vascular endothelial growth factor) during salt stress thus stimulating lymphangiogenesis, thereby acting as a buffer against increased blood pressure (BP). We aimed to explore the role of dermal lymphatics in BP and sodium homeostasis. Our hypothesis was that mice with reduced dermal lymphatic vessels were more prone to develop salt-sensitive hypertension, and that mice with hyperplastic vessels were protected.
Methods:
Mice with either hypoplastic (Chy), absent (K14-VEGFR3 [vascular endothelial growth factor receptor 3]-Ig), or hyperplastic (K14-VEGF-C) dermal lymphatic vessels and littermate controls were given high-salt diet (4% NaCl in the chow), deoxycorticosterone acetate (DOCA)-salt diet and 1% saline to drink or nitric oxide blocker diet L-N
G
-nitro arginine methyl ester (followed by high salt diet). BP was measured by telemetric recording, and tissue sodium content by ion chromatography.
Results:
In contrast to previous studies, high salt diet did not induce an increase in BP or sodium storage in any of the mouse strains investigated. DOCA-salt, on the other hand, gave an increase in BP in Chy and K14-VEGFR3-Ig not different from their corresponding WT controls. DOCA induced salt storage in skin and muscle, but to the same extent in mice with dysfunctional lymphatic vessels and WT controls. Lymph flow as assessed by tracer washout was not affected by the diet in any of the mouse strains.
Conclusions:
Our results suggest that dermal lymphatic vessels are not involved in salt storage or blood pressure regulation in these mouse models of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Irene Matre Thowsen
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Tore Reikvam
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Trude Skogstrand
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Anne-Maj Samuelsson
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
- Department of Medicine, Haukeland University Hospital, Bergen, Norway (A.-M.S.)
| | - Dominik N. Müller
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany (D.N.M.)
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Finland (K.A.)
| | - Tine Karlsen
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| |
Collapse
|
21
|
Mattson DL. Lymphatics and Blood Pressure. Hypertension 2022; 79:2463-2464. [DOI: 10.1161/hypertensionaha.122.20166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Abstract
The lymphatic vessels play an essential role in maintaining immune and fluid homeostasis and in the transport of dietary lipids. The discovery of lymphatic endothelial cell-specific markers facilitated the visualization and mechanistic analysis of lymphatic vessels over the past two decades. As a result, lymphatic vessels have emerged as a crucial player in the pathogenesis of several cardiovascular diseases, as demonstrated by worsened disease progression caused by perturbations to lymphatic function. In this review, we discuss the major findings on the role of lymphatic vessels in cardiovascular diseases such as hypertension, obesity, atherosclerosis, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Dakshnapriya Balasubbramanian
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas 77807, USA
| |
Collapse
|
23
|
Geng X, Srinivasan RS. Molecular Mechanisms Driving Lymphedema and Other Lymphatic Anomalies. Cold Spring Harb Perspect Med 2022; 12:a041272. [PMID: 35817543 PMCID: PMC9341459 DOI: 10.1101/cshperspect.a041272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lymphatic vasculature regulates fluid homeostasis by absorbing interstitial fluid and returning it to blood. Lymphatic vasculature is also critical for lipid absorption and inflammatory response. Lymphatic vasculature is composed of lymphatic capillaries, collecting lymphatic vessels, lymphatic valves, and lymphovenous valves. Defects in any of these structures could lead to lymphatic anomalies such as lymphedema, cystic lymphatic malformation, and Gorham-Stout disease. Basic research has led to a deeper understanding of the stepwise development of the lymphatic vasculature. VEGF-C and shear stress signaling pathways have evolved as critical regulators of lymphatic vascular development. Loss-of-function and gain-of-function mutations in genes that are involved in these signaling pathways are associated with lymphatic anomalies. Importantly, drugs that target these molecules are showing outstanding efficacy in treating certain lymphatic anomalies. In this article, we summarize these exciting developments and highlight the future challenges.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73013, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73013, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, USA
| |
Collapse
|
24
|
Liu J, Shelton EL, Crescenzi R, Colvin DC, Kirabo A, Zhong J, Delpire EJ, Yang HC, Kon V. Kidney Injury Causes Accumulation of Renal Sodium That Modulates Renal Lymphatic Dynamics. Int J Mol Sci 2022; 23:ijms23031428. [PMID: 35163352 PMCID: PMC8836121 DOI: 10.3390/ijms23031428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 01/02/2023] Open
Abstract
Lymphatic vessels are highly responsive to changes in the interstitial environment. Previously, we showed renal lymphatics express the Na-K-2Cl cotransporter. Since interstitial sodium retention is a hallmark of proteinuric injury, we examined whether renal sodium affects NKCC1 expression and the dynamic pumping function of renal lymphatic vessels. Puromycin aminonucleoside (PAN)-injected rats served as a model of proteinuric kidney injury. Sodium 23Na/1H-MRI was used to measure renal sodium and water content in live animals. Renal lymph, which reflects the interstitial composition, was collected, and the sodium analyzed. The contractile dynamics of isolated renal lymphatic vessels were studied in a perfusion chamber. Cultured lymphatic endothelial cells (LECs) were used to assess direct sodium effects on NKCC1. MRI showed elevation in renal sodium and water in PAN. In addition, renal lymph contained higher sodium, although the plasma sodium showed no difference between PAN and controls. High sodium decreased contractility of renal collecting lymphatic vessels. In LECs, high sodium reduced phosphorylated NKCC1 and SPAK, an upstream activating kinase of NKCC1, and eNOS, a downstream effector of lymphatic contractility. The NKCC1 inhibitor furosemide showed a weaker effect on ejection fraction in isolated renal lymphatics of PAN vs controls. High sodium within the renal interstitium following proteinuric injury is associated with impaired renal lymphatic pumping that may, in part, involve the SPAK-NKCC1-eNOS pathway, which may contribute to sodium retention and reduce lymphatic responsiveness to furosemide. We propose that this lymphatic vessel dysfunction is a novel mechanism of impaired interstitial clearance and edema in proteinuric kidney disease.
Collapse
Affiliation(s)
- Jing Liu
- Department of Nephrology, Tongji University School of Medicine, Shanghai 200070, China;
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Elaine L. Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachelle Crescenzi
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (R.C.); (D.C.C.)
| | - Daniel C. Colvin
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (R.C.); (D.C.C.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinal Pharmacology and Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (A.K.); (J.Z.)
| | - Jianyong Zhong
- Department of Medicine, Division of Clinal Pharmacology and Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (A.K.); (J.Z.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Eric J. Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hai-Chun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: (H.-C.Y.); (V.K.); Tel.: +1-615-343-0110 (H.-C.Y.); +1-615-322-7416 (V.K.)
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: (H.-C.Y.); (V.K.); Tel.: +1-615-343-0110 (H.-C.Y.); +1-615-322-7416 (V.K.)
| |
Collapse
|
25
|
Goodlett BL, Kang CS, Yoo E, Navaneethabalakrishnan S, Balasubbramanian D, Love SE, Sims BM, Avilez DL, Tate W, Chavez DR, Baranwal G, Nabity MB, Rutkowski JM, Kim D, Mitchell BM. A Kidney-Targeted Nanoparticle to Augment Renal Lymphatic Density Decreases Blood Pressure in Hypertensive Mice. Pharmaceutics 2021; 14:pharmaceutics14010084. [PMID: 35056980 PMCID: PMC8780399 DOI: 10.3390/pharmaceutics14010084] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/07/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic interstitial inflammation and renal infiltration of activated immune cells play an integral role in hypertension. Lymphatics regulate inflammation through clearance of immune cells and excess interstitial fluid. Previously, we demonstrated increasing renal lymphangiogenesis prevents hypertension in mice. We hypothesized that targeted nanoparticle delivery of vascular endothelial growth factor-C (VEGF-C) to the kidney would induce renal lymphangiogenesis, lowering blood pressure in hypertensive mice. A kidney-targeting nanoparticle was loaded with a VEGF receptor-3-specific form of VEGF-C and injected into mice with angiotensin II-induced hypertension or LNAME-induced hypertension every 3 days. Nanoparticle-treated mice exhibited increased renal lymphatic vessel density and width compared to hypertensive mice injected with VEGF-C alone. Nanoparticle-treated mice exhibited decreased systolic blood pressure, decreased pro-inflammatory renal immune cells, and increased urinary fractional excretion of sodium. Our findings demonstrate that pharmacologically expanding renal lymphatics decreases blood pressure and is associated with favorable alterations in renal immune cells and increased sodium excretion.
Collapse
Affiliation(s)
- Bethany L. Goodlett
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Chang Sun Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (C.S.K.); (E.Y.); (D.K.)
| | - Eunsoo Yoo
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (C.S.K.); (E.Y.); (D.K.)
| | - Shobana Navaneethabalakrishnan
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Dakshnapriya Balasubbramanian
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Sydney E. Love
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Braden M. Sims
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Daniela L. Avilez
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Winter Tate
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Delilah R. Chavez
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Gaurav Baranwal
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Mary B. Nabity
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Science, Texas A&M University, College Station, TX 77843, USA;
| | - Joseph M. Rutkowski
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (C.S.K.); (E.Y.); (D.K.)
| | - Brett M. Mitchell
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77807, USA; (B.L.G.); (S.N.); (D.B.); (S.E.L.); (B.M.S.); (D.L.A.); (W.T.); (D.R.C.); (G.B.); (J.M.R.)
- Correspondence: ; Tel.:+1-979-436-0751
| |
Collapse
|
26
|
Black LM, Farrell ER, Barwinska D, Osis G, Zmijewska AA, Traylor AM, Esman SK, Bolisetty S, Whipple G, Kamocka MM, Winfree S, Spangler DR, Khan S, Zarjou A, El-Achkar TM, Agarwal A. VEGFR3 tyrosine kinase inhibition aggravates cisplatin nephrotoxicity. Am J Physiol Renal Physiol 2021; 321:F675-F688. [PMID: 34658261 PMCID: PMC8714977 DOI: 10.1152/ajprenal.00186.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elisa R Farrell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daria Barwinska
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Gunars Osis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anna A Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephanie K Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Grace Whipple
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Malgorzata M Kamocka
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Seth Winfree
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Daryll R Spangler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
- Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
27
|
Liu H, Hiremath C, Patterson Q, Vora S, Shang Z, Jamieson AR, Fiolka R, Dean KM, Dellinger MT, Marciano DK. Heterozygous Mutation of Vegfr3 Reduces Renal Lymphatics without Renal Dysfunction. J Am Soc Nephrol 2021; 32:3099-3113. [PMID: 34551997 PMCID: PMC8638391 DOI: 10.1681/asn.2021010061] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Lymphatic abnormalities are observed in several types of kidney disease, but the relationship between the renal lymphatic system and renal function is unclear. The discovery of lymphatic-specific proteins, advances in microscopy, and available genetic mouse models provide the tools to help elucidate the role of renal lymphatics in physiology and disease. METHODS We utilized a mouse model containing a missense mutation in Vegfr3 (dubbed Chy ) that abrogates its kinase ability. Vegfr3 Chy/+ mice were examined for developmental abnormalities and kidney-specific outcomes. Control and Vegfr3 Chy/+ mice were subjected to cisplatin-mediated injury. We characterized renal lymphatics using tissue-clearing, light-sheet microscopy, and computational analyses. RESULTS In the kidney, VEGFR3 is expressed not only in lymphatic vessels but also, in various blood capillaries. Vegfr3 Chy/+ mice had severely reduced renal lymphatics with 100% penetrance, but we found no abnormalities in BP, serum creatinine, BUN, albuminuria, and histology. There was no difference in the degree of renal injury after low-dose cisplatin (5 mg/kg), although Vegfr3 Chy/+ mice developed perivascular inflammation. Cisplatin-treated controls had no difference in total cortical lymphatic volume and length but showed increased lymphatic density due to decreased cortical volume. CONCLUSIONS We demonstrate that VEGFR3 is required for development of renal lymphatics. Our studies reveal that reduced lymphatic density does not impair renal function at baseline and induces only modest histologic changes after mild injury. We introduce a novel quantification method to evaluate renal lymphatics in 3D and demonstrate that accurate measurement of lymphatic density in CKD requires assessment of changes to cortical volume.
Collapse
Affiliation(s)
- Hao Liu
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quinten Patterson
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Saumya Vora
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiguo Shang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew R. Jamieson
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Reto Fiolka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas,Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kevin M. Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael T. Dellinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K. Marciano
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
28
|
Baranwal G, Creed HA, Black LM, Auger A, Quach AM, Vegiraju R, Eckenrode HE, Agarwal A, Rutkowski JM. Expanded renal lymphatics improve recovery following kidney injury. Physiol Rep 2021; 9:e15094. [PMID: 34806312 PMCID: PMC8606868 DOI: 10.14814/phy2.15094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) is a major cause of patient mortality and a major risk multiplier for the progression to chronic kidney disease (CKD). The mechanism of the AKI to CKD transition is complex but is likely mediated by the extent and length of the inflammatory response following the initial injury. Lymphatic vessels help to maintain tissue homeostasis through fluid, macromolecule, and immune modulation. Increased lymphatic growth, or lymphangiogenesis, often occurs during inflammation and plays a role in acute and chronic disease processes. What roles renal lymphatics and lymphangiogenesis play in AKI recovery and CKD progression remains largely unknown. To determine if the increased lymphatic density is protective in the response to kidney injury, we utilized a transgenic mouse model with inducible, kidney-specific overexpression of the lymphangiogenic protein vascular endothelial growth factor-D to expand renal lymphatics. "KidVD" mouse kidneys were injured using inducible podocyte apoptosis and proteinuria (POD-ATTAC) or bilateral ischemia reperfusion. In the acute injury phase of both models, KidVD mice demonstrated a similar loss of function measured by serum creatinine and glomerular filtration rate compared to their littermates. While the initial inflammatory response was similar, KidVD mice demonstrated a shift toward more CD4+ and fewer CD8+ T cells in the kidney. Reduced collagen deposition and improved functional recovery over time was also identified in KidVD mice. In KidVD-POD-ATTAC mice, an increased number of podocytes were counted at 28 days post-injury. These data demonstrate that increased lymphatic density prior to injury alters the injury recovery response and affords protection from CKD progression.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Heidi A. Creed
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Laurence M. Black
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Alexa Auger
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Alexander M. Quach
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Rahul Vegiraju
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| | - Han E. Eckenrode
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anupam Agarwal
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Nephrology Research and Training CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Department of Veterans AffairsBirmingham Veterans Administration Medical CenterBirminghamAlabamaUSA
| | - Joseph M. Rutkowski
- Division of Lymphatic BiologyDepartment of Medical PhysiologyTexas A&M University College of MedicineBryanTexasUSA
| |
Collapse
|
29
|
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: 34] [Impact Index Per Article: 8.5] [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
|
30
|
Baranwal G, Pilla R, Goodlett BL, Coleman AK, Arenaz CM, Jayaraman A, Rutkowski JM, Alaniz RC, Mitchell BM. Common Metabolites in Two Different Hypertensive Mouse Models: A Serum and Urine Metabolome Study. Biomolecules 2021; 11:1387. [PMID: 34572600 PMCID: PMC8467937 DOI: 10.3390/biom11091387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 11/24/2022] Open
Abstract
Recent metabolomics studies have identified a wide array of microbial metabolites and metabolite pathways that are significantly altered in hypertension. However, whether these metabolites play an active role in pathogenesis of hypertension or are altered because of this has yet to be determined. In the current study, we hypothesized that metabolite changes common between hypertension models may unify hypertension's pathophysiology with respect to metabolites. We utilized two common mouse models of experimental hypertension: L-arginine methyl ester hydrochloride (L-NAME)/high-salt-diet-induced hypertension (LSHTN) and angiotensin II induced hypertension (AHTN). To identify common metabolites that were altered across both models, we performed untargeted global metabolomics analysis in serum and urine and the resulting data were analyzed using MetaboAnalyst software and compared to control mice. A total of 41 serum metabolites were identified as being significantly altered in any hypertensive model compared to the controls. Of these compounds, 14 were commonly changed in both hypertensive groups, with 4 significantly increased and 10 significantly decreased. In the urine, six metabolites were significantly altered in any hypertensive group with respect to the control; however, none of them were common between the hypertensive groups. These findings demonstrate that a modest, but potentially important, number of serum metabolites are commonly altered between experimental hypertension models. Further studies of the newly identified metabolites from this untargeted metabolomics analysis may lead to a greater understanding of the association between gut dysbiosis and hypertension.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| | - Rachel Pilla
- Department of Small Animal Clinical Science, College of Veterinary Medicine & Biomedical Science, Texas A&M University, College Station, TX 77843, USA;
| | - Bethany L. Goodlett
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| | - Aja K. Coleman
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| | - Cristina M. Arenaz
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| | - Arul Jayaraman
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (A.J.); (R.C.A.)
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Joseph M. Rutkowski
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (A.J.); (R.C.A.)
| | - Brett M. Mitchell
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX 77847, USA; (G.B.); (B.L.G.); (A.K.C.); (C.M.A.); (J.M.R.)
| |
Collapse
|
31
|
Geng X, Ho YC, Srinivasan RS. Biochemical and mechanical signals in the lymphatic vasculature. Cell Mol Life Sci 2021; 78:5903-5923. [PMID: 34240226 PMCID: PMC11072415 DOI: 10.1007/s00018-021-03886-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
Lymphatic vasculature is an integral part of the cardiovascular system where it maintains interstitial fluid balance. Additionally, lymphatic vasculature regulates lipid assimilation and inflammatory response. Lymphatic vasculature is composed of lymphatic capillaries, collecting lymphatic vessels and valves that function in synergy to absorb and transport fluid against gravitational and pressure gradients. Defects in lymphatic vessels or valves leads to fluid accumulation in tissues (lymphedema), chylous ascites, chylothorax, metabolic disorders and inflammation. The past three decades of research has identified numerous molecules that are necessary for the stepwise development of lymphatic vasculature. However, approaches to treat lymphatic disorders are still limited to massages and compression bandages. Hence, better understanding of the mechanisms that regulate lymphatic vascular development and function is urgently needed to develop efficient therapies. Recent research has linked mechanical signals such as shear stress and matrix stiffness with biochemical pathways that regulate lymphatic vessel growth, patterning and maturation and valve formation. The goal of this review article is to highlight these innovative developments and speculate on unanswered questions.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73013, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
32
|
Donnan MD. Kidney lymphatics: new insights in development and disease. Curr Opin Nephrol Hypertens 2021; 30:450-455. [PMID: 34027907 DOI: 10.1097/mnh.0000000000000717] [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 This review will highlight recent advances in our understanding of the kidney lymphatics regarding their development, physiologic function, and their potential role in the progression of kidney disease. RECENT FINDINGS Although sparse in comparison to the blood vasculature, lymphatic vessels within the healthy kidney perform an important role in maintaining homeostasis. Additionally, in response to kidney injury, lymphatic vessels undergo substantial expansion, termed lymphangiogenesis, which shows a direct correlation to the extent of tubulointerstitial fibrosis. Kidney lymphatics expand through both the proliferation of lymphatic endothelial cells from existing lymphatic vessels, as well as from direct contribution by other cell types of nonvenous origin. The primary driver of lymphatic growth is vascular endothelial growth factor C, both in development and in response to injury. The clinical implications of lymphangiogenesis in the setting of kidney diseases remains debated, however growing evidence suggests lymphatic vessels may perform a protective role in clearing away accumulating interstitial fluid, inflammatory cytokines, and cellular infiltrates that occur with injury. SUMMARY There is increasing evidence the kidney lymphatics perform an active role in the response to kidney injury and the development of fibrosis. Recent advances in our understanding of these vessels raise the possibility of targeting kidney lymphatics for the treatment of kidney disease.
Collapse
Affiliation(s)
- Michael D Donnan
- Feinberg Cardiovascular & Renal Research Institute.,Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
33
|
Creed HA, Rutkowski JM. Emerging roles for lymphatics in acute kidney injury: Beneficial or maleficent? Exp Biol Med (Maywood) 2021; 246:845-850. [PMID: 33467886 DOI: 10.1177/1535370220983235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury, a sudden decline in renal filtration, is a surprisingly common pathology resulting from ischemic events, local or systemic infection, or drug-induced toxicity in the kidney. Unchecked, acute kidney injury can progress to renal failure and even recovered acute kidney injury patients are at an increased risk for developing future chronic kidney disease. The initial extent of inflammation, the specific immune response, and how well inflammation resolves are likely determinants in acute kidney injury-to-chronic kidney disease progression. Lymphatic vessels and their roles in fluid, solute, antigen, and immune cell transport make them likely to have a role in the acute kidney injury response. Lymphatics have proven to be an attractive target in regulating inflammation and immunomodulation in other pathologies: might these strategies be employed in acute kidney injury? Acute kidney injury studies have identified elevated levels of lymphangiogenic ligands following acute kidney injury, with an expansion of the lymphatics in several models post-injury. Manipulating the lymphatics in acute kidney injury, by augmenting or inhibiting their growth or through targeting lymphatic-immune interactions, has met with a range of positive, negative, and sometimes inconclusive results. This minireview briefly summarizes the findings of lymphatic changes and lymphatic roles in the inflammatory response in the kidney following acute kidney injury to discuss whether renal lymphatics are a beneficial, maleficent, or a passive contributor to acute kidney injury recovery.
Collapse
Affiliation(s)
- Heidi A Creed
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| | - Joseph M Rutkowski
- Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA
| |
Collapse
|