1
|
Xiang Q, Ni L, Xu Z, Ma X, Wang Y, Xuan Z, Xu F. Exploring the therapeutic potential of Danggui Shaoyao San in nephrotic syndrome: Impact on skin sodium content and renal function. Heliyon 2025; 11:e42577. [PMID: 40051859 PMCID: PMC11883388 DOI: 10.1016/j.heliyon.2025.e42577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
The purpose of this study was to explore the mechanism of sodium transport in the skin of rats with nephrotic syndrome (NS) and the intervention effect of Danggui Shaoyao San (DSS). NS model was established by tail vein injection of adriamycin (ADR), and different doses of DSS, vascular endothelial growth factor receptor-3 (VEGFR-3) inhibitor Levatinib and diuretic amiloride were given for intervention. We analysed serum biochemical parameters, urine sodium, skin sodium and glycosaminoglycan (GAG), lymphatic vessel density, and the expression and mRNA levels of key proteins involved in lymphangiogenesis. The results showed that DSS treatment not only significantly reduced the content of sodium ions in the skin of NS rats, but also effectively alleviated the pathological damage of the kidney, improved proteinuria and promoted the excretion of sodium in urine. At the same time, Levatinib can reduce the content of sodium ions in the skin of NS rats by inhibiting lymphangiogenesis, while amiloride can improve the state of sodium retention in the body and reduce the level of sodium ions in the skin by promoting the excretion of sodium ions in the urine of NS rats. Especially important, DSS can effectively inhibit the proliferation of renal cortical lymphatic vessels, down-regulate the expression of lymphangiogenesis related proteins and mRNA, promote urinary sodium excretion, and inhibit the transport of sodium ions from kidney to skin. In summary, this study reveals that during sodium retention in NS rats, sodium ions can be further transported to the skin for storage through increased lymph in the kidney, and DSS can inhibit renal lymphangiogenesis, promote urinary sodium excretion, and reduce the content of sodium ions in the skin, which provides a novel and potential strategy for the treatment of NS edema.
Collapse
Affiliation(s)
- Qingzhen Xiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lianghou Ni
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zaiping Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xiaowen Ma
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yunlai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Zihua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Fan Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
- Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| |
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
|
Wu Q, Fu J, Zhu B, Meng W, Ma J, Lv Y, Zhao W, Wang F, Liu J, Wang Y, Peng C, Zhang S. VEGFR3 mitigates hypertensive nephropathy by enhancing mitophagy via regulating crotonylation of HSPA1L. Cell Commun Signal 2025; 23:52. [PMID: 39875989 PMCID: PMC11773936 DOI: 10.1186/s12964-025-02045-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Oxidative stress-associated proximal tubular cells (PTCs) damage is an important pathogenesis of hypertensive renal injury. We previously reported the protective effect of VEGFR3 in salt-sensitive hypertension. However, the specific mechanism underlying the role of VEGFR3 in kidney during the overactivation of the renin-angiotensin-aldosterone system remains unclear. In the present study, hypertensive nephropathy was established by angiotensin II (Ang II). We found that VEGFR3 was highly increased in PTCs of Ang II-infused mice. Activation of VEGFR3 mitigated renal dysfunction, pathological damage, and oxidative stress in Ang II-induced hypertensive mice. Moreover, we found that VEGFR3 restored mitophagy deficiency induced by Ang II both in vivo and in vitro to alleviate oxidative stress injury in PTCs. Furthermore, in vitro experiment demonstrated that VEGFR3 improved abnormal mitophagy by enhancing PARKIN mitochondrial translocation. LC-MS/MS and Co-IP assays identified HSPA1L as the interacted protein of VEGFR3, which promoted the mitochondrial translocation of PARKIN. Mechanistically, VEGFR3 disorder domain bound to HSPA1L, and crotonylation modification of HSPA1L at K130 by VEGFR3 was required for mitophagy regulation in the context of Ang II-induced PTCs. Finally, the protective effect of VEGFR3 on mitophagy and oxidative stress were attenuated by transfection K130 (HSPA1L-K130R) mutant plasmid in vivo and in vitro. These findings indicated that VEGFR3 alleviated oxidative stress by promoting PARKIN-dependent mitophagy pathway via regulating HSPA1L crotonylation at K130 site in Ang II-induced PTCs, which provided a mechanistic basis for the therapeutic target in hypertensive renal injury.
Collapse
Affiliation(s)
- Qiuwen Wu
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), 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
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Bin Zhu
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Wei Meng
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jingyi Ma
- Department of Endocrinology and Diabetes, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), 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
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Fan Wang
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jingjin Liu
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Yongshun Wang
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Cong Peng
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Shuo Zhang
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
| |
Collapse
|
4
|
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
|
5
|
Li Y, Hunter A, Wakeel MM, Sun G, Lau RWK, Broughton BRS, Pino IEO, Deng Z, Zhang T, Murthi P, Del Borgo MP, Widdop RE, Polo JM, Ricardo SD, Samuel CS. The renoprotective efficacy and safety of genetically-engineered human bone marrow-derived mesenchymal stromal cells expressing anti-fibrotic cargo. Stem Cell Res Ther 2024; 15:375. [PMID: 39443975 PMCID: PMC11515549 DOI: 10.1186/s13287-024-03992-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Kidney fibrosis is a hallmark of chronic kidney disease (CKD) and compromises the viability of transplanted human bone marrow-derived mesenchymal stromal cells (BM-MSCs). Hence, BM-MSCs were genetically-engineered to express the anti-fibrotic and renoprotective hormone, human relaxin-2 (RLX) and green fluorescent protein (BM-MSCs-eRLX + GFP), which enabled BM-MSCs-eRLX + GFP delivery via a single intravenous injection. METHODS BM-MSCs were lentiviral-transduced with human relaxin-2 cDNA and GFP, under a eukaryotic translation elongation factor-1α promoter (BM-MSCs-eRLX + GFP) or GFP alone (BM-MSCs-eGFP). The ability of BM-MSCs-eRLX + GFP to differentiate, proliferate, migrate, produce RLX and cytokines was evaluated in vitro, whilst BM-MSC-eRLX + GFP vs BM-MSCs-eGFP homing to the injured kidney and renoprotective effects were evaluated in preclinical models of ischemia reperfusion injury (IRI) and high salt (HS)-induced hypertensive CKD in vivo. The long-term safety of BM-MSCs-RLX + GFP was also determined 9-months after treatment cessation in vivo. RESULTS When cultured for 3- or 7-days in vitro, 1 × 106 BM-MSCs-eRLX + GFP produced therapeutic RLX levels, and secreted an enhanced but finely-tuned cytokine profile without compromising their proliferation or differentiation capacity compared to naïve BM-MSCs. BM-MSCs-eRLX + GFP were identified in the kidney 2-weeks post-administration and retained the therapeutic effects of RLX in vivo. 1-2 × 106 BM-MSCs-eRLX + GFP attenuated the IRI- or therapeutically abrogated the HS-induced tubular epithelial damage and interstitial fibrosis, and significantly reduced the HS-induced hypertension, glomerulosclerosis and proteinuria. This was to an equivalent extent as RLX and BM-MSCs administered separately but to a broader extent than BM-MSCs-eGFP or the angiotensin-converting enzyme inhibitor, perindopril. Additionally, these renoprotective effects of BM-MSCs-eRLX + GFP were maintained in the presence of perindopril co-treatment, highlighting their suitability as adjunct therapies to ACE inhibition. Importantly, no major long-term adverse effects of BM-MSCs-eRLX + GFP were observed. CONCLUSIONS BM-MSCs-eRLX + GFP produced greater renoprotective and therapeutic efficacy over that of BM-MSCs-eGFP or ACE inhibition, and may represent a novel and safe treatment option for acute kidney injury and hypertensive CKD.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Alex Hunter
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Miqdad M Wakeel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Guizhi Sun
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Ricky W K Lau
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Ivan E Oyarce Pino
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Zihao Deng
- Department of Medicine (Alfred Hospital), Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Tingfang Zhang
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Padma Murthi
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Mark P Del Borgo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Jose M Polo
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Adelaide Centre for Epigenetics, School of Biomedicine, The University of Adelaide, Adelaide, SA, 5005, Australia
- The South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Sharon D Ricardo
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia.
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
6
|
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
|
7
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
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
|
9
|
Chu W, Ma LL, Li BX, Li MC. Clinical significance of vascular endothelial growth factor and endothelin-1 in serum levels as novel indicators for predicting the progression of diabetic nephropathy. EUR J INFLAMM 2023. [DOI: 10.1177/1721727x231151526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Objective: Early diagnosis and intervention of diabetic nephropathy (DN) is necessary to optimize therapy in order to delay the progression of diabetes. This research aimed to reveal the change of vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1) in patients with DN, and to assess possible correlations with glycated hemoglobin (HbAlc) values. Methods: The present study was a retrospective, single-center study conducted at a teaching hospital in the northeast China. A total of 120 patients were divided into proteinuria-positive group ( n = 40), the microalbuminuria group ( n = 40), and the high proteinuria group ( n = 40) according to the urinary albumin excretion rate (UAER), and 40 healthy volunteers were selected as the control group. The levels of VEGF, ET-1 and HbA1c were measured in all subjects and principal component analysis (PCA) was performed to classify and reveal correlations between VEGF, ET-1 and HbA1c. Results: Compared to the control group, a significant difference in the increase of HbA1c was detected in group I, II and III. A significant increase in the concentrations of serum VEGF and ET-1 was also observed. HbA1c in DN patients had proven to be positively correlated with VEGF (r = 0.7941; p < 0. 0001) and ET-1 (r = 0.8504; p < 0.0001) respectively. Conclusion: The elevated levels of VEGF and ET-1 in serum have been proposed as being able to supplement the additional information about the progression of DN. These data suggest that the decrease in endothelial function may be related to poor glycemic control.
Collapse
Affiliation(s)
- Wei Chu
- Department of Renal Medicine, People’s Hospital of Jilin, Jilin, China
| | - Lin-Lin Ma
- Department of Clinical Laboratory, Beihua University, Jilin, China
| | - Bin-Xian Li
- Department of Clinical Laboratory, Beihua University, Jilin, China
| | - Ming-Cheng Li
- Department of Molecular diagnosis, Beihua University, Jilin, China
| |
Collapse
|
10
|
Li J, Li XL, Li CQ. Immunoregulation mechanism of VEGF signaling pathway inhibitors and its efficacy on the kidney. Am J Med Sci 2023; 366:404-412. [PMID: 37699444 DOI: 10.1016/j.amjms.2023.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 05/10/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023]
Abstract
Angiogenesis and immunosuppression are closely related pathophysiologic processes. Widely prescribed in malignant tumor and proliferative retinal lesions, VEGF signaling pathway inhibitors may cause hypertension and renal injury in some patients, presenting with proteinuria, nephrotic syndrome, renal failure and thrombotic microangiopathy. VEGF signaling pathway inhibitors block the action of both VEGF-A and VEGF-C. However, VEGF-A and VEGF-C produced by podocytes are vital to maintain the physiological function of glomerular endothelial cells and podocytes. There is still no effective treatment for kidney disease associated with VEGF signaling pathway inhibitors and some patients have progressive renal failure even after withdrawal of the drug. Recent studies reveal that blocking of VEGF-A and VEGF-C can activate CD4 +and CD8+ T cells, augment antigen-presenting function of dendritic cells, enhance cytotoxicity of macrophages and initiate complement cascade activation. VEGF and VEGFR are expressed in immune cells, which are involved in the immunosuppression and cross-talk among immune cells. This review summarizes the expression and function of VEGF-A and VEGF-C in the kidney. The current immunoregulation mechanisms of VEGF signaling pathway inhibitors are reviewed. Finally, combinate strategies are summarized to highlight the proposal for VEGF signaling pathway inhibitors.
Collapse
Affiliation(s)
- Jun Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Jiangsu, China.
| | - Xiao-Lin Li
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Chun-Qing Li
- Department of Nephrology, Affiliated Hospital of Jiangnan University, Jiangsu, China
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Zhuang T, Lei Y, Chang JJ, Zhou YP, Li Y, Li YX, Yang YF, Chen MH, Meng T, Fu SM, Huang LH, Cheang WS, Cooke JP, Dong ZH, Bai YN, Ruan CC. A2AR-mediated lymphangiogenesis via VEGFR2 signaling prevents salt-sensitive hypertension. Eur Heart J 2023; 44:2730-2742. [PMID: 37377160 PMCID: PMC10393074 DOI: 10.1093/eurheartj/ehad377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 04/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS Excess dietary sodium intake and retention lead to hypertension. Impaired dermal lymphangiogenesis and lymphatic dysfunction-mediated sodium and fluid imbalance are pathological mechanisms. The adenosine A2A receptor (A2AR) is expressed in lymphatic endothelial cells (LECs), while the roles and mechanisms of LEC-A2AR in skin lymphangiogenesis during salt-induced hypertension are not clear. METHODS AND RESULTS The expression of LEC-A2AR correlated with lymphatic vessel density in both high-salt diet (HSD)-induced hypertensive mice and hypertensive patients. Lymphatic endothelial cell-specific A2AR knockout mice fed HSD exhibited 17 ± 2% increase in blood pressure and 17 ± 3% increase in Na+ content associated with decreased lymphatic density (-19 ± 2%) compared with HSD-WT mice. A2AR activation by agonist CGS21680 increased lymphatic capillary density and decreased blood pressure in HSD-WT mice. Furthermore, this A2AR agonist activated MSK1 directly to promote VEGFR2 activation and endocytosis independently of VEGF as assessed by phosphoprotein profiling and immunoprecipitation assays in LECs. VEGFR2 kinase activity inhibitor fruquintinib or VEGFR2 knockout in LECs but not VEGF-neutralizing antibody bevacizumab suppressed A2AR activation-mediated decrease in blood pressure. Immunostaining revealed phosphorylated VEGFR2 and MSK1 expression in the LECs were positively correlated with skin lymphatic vessel density and A2AR level in hypertensive patients. CONCLUSION The study highlights a novel A2AR-mediated VEGF-independent activation of VEGFR2 signaling in dermal lymphangiogenesis and sodium balance, which might be a potential therapeutic target in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Jin-Jia Chang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Yan-Ping Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pu-Jian Road, Shanghai 200032, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, 149 Chong-Qing-Nan Road, Shanghai 200032, China
| | - Yan-Xiu Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guang-Zhou Road, Nanjing 210000, China
| | - Yong-Feng Yang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Mei-Hua Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Shi-Man Fu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Li-Hao Huang
- Department of Chemistry and Institute of Metabolism and Integrative Biology, Shanghai Key Laboratory of Metabolic Remodeling and Health, Fudan University, 38 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Wai-San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Long-Ma Road, Macau 999078, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhi-Hui Dong
- Department of Vascular Surgery, Zhongshan Hospital, and Center for Vascular Surgery and Wound Care, Jinshan Hospital, Fudan University, 180 Feng-Lin Road, Shanghai 200032, China
| | - Ying-Nan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Feng-Lin Road, Shanghai 200032, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| |
Collapse
|
14
|
Zhang X, Gao Y, Yang B, Ma S, Zuo W, Wei J. The mechanism and treatment of targeted anti-tumour drugs induced cardiotoxicity. Int Immunopharmacol 2023; 117:109895. [PMID: 36806040 DOI: 10.1016/j.intimp.2023.109895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/29/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
As the intensive anti-tumour therapy and combination of multiple anti-tumour drugs, cardiotoxicity events caused by anti-tumour drugs have also increased significantly, and the incidence of cardiotoxicity also increased with survival time. Different types of anti-tumour drugs could cause all kinds of cardiotoxicity which increase the difficulties in treatment and even live threatening. In this review, we concentrated in the targeted anti-tumour drugs such as human epidermal growth factor receptor-2 (HER2) inhibitors, tyrosine kinase inhibitors (TKIs), immune checkpoint inhibitors (ICIs), and proteasome inhibitors (Pls). The molecular mechanism of how these drugs induce cardiotoxicity is introduced which includes several signal pathways. These drugs induced cardiotoxicity involved heart failure, hypertension, atherosis and thrombosis, QT interval prolongation, and myocarditis. Some of the cardiotoxicity could be moderate and reversible but others could have happened severely.The aim of this review is to summarise the targeted anti-tumour drugs induced cardiotoxicity and treatment strategies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Yiqiang Gao
- Department of Pharmacy, State Key Laboratory of Complex Sever and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China; School of Pharmacy, Tianjin University of Traditional Chinese Medicine, China.
| | - Benyu Yang
- Department of Pharmacy, State Key Laboratory of Complex Sever and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China; School of Pharmacy, Tianjin University of Traditional Chinese Medicine, China
| | - Siqing Ma
- School of Mechanical and Materials Engineering, University College Dublin, Belfield, Dublin, Ireland
| | - Wei Zuo
- Department of Pharmacy, State Key Laboratory of Complex Sever and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Junji Wei
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
15
|
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
|
16
|
Heron C, Dumesnil A, Houssari M, Renet S, Lemarcis T, Lebon A, Godefroy D, Schapman D, Henri O, Riou G, Nicol L, Henry JP, Valet M, Pieronne-Deperrois M, Ouvrard-Pascaud A, Hägerling R, Chiavelli H, Michel JB, Mulder P, Fraineau S, Richard V, Tardif V, Brakenhielm E. Regulation and impact of cardiac lymphangiogenesis in pressure-overload-induced heart failure. Cardiovasc Res 2022; 119:492-505. [PMID: 35689481 PMCID: PMC10064842 DOI: 10.1093/cvr/cvac086] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 04/14/2022] [Accepted: 05/12/2022] [Indexed: 12/11/2022] Open
Abstract
AIMS Lymphatics are essential for cardiac health, and insufficient lymphatic expansion (lymphangiogenesis) contributes to development of heart failure (HF) after myocardial infarction. However, the regulation and impact of lymphangiogenesis in non-ischemic cardiomyopathy following pressure-overload remains to be determined. Here, we investigated cardiac lymphangiogenesis following transversal aortic constriction (TAC) in C57Bl/6 and Balb/c mice, and in end-stage HF patients. METHODS & RESULTS Cardiac function was evaluated by echocardiography, and cardiac hypertrophy, lymphatics, inflammation, edema, and fibrosis by immunohistochemistry, flow cytometry, microgravimetry, and gene expression analysis. Treatment with neutralizing anti-VEGFR3 antibodies was applied to inhibit cardiac lymphangiogenesis in mice.We found that VEGFR3-signaling was essential to prevent cardiac lymphatic rarefaction after TAC in C57Bl/6 mice. While anti-VEGFR3-induced lymphatic rarefaction did not significantly aggravate myocardial edema post-TAC, cardiac immune cell levels were increased, notably myeloid cells at 3 weeks and T lymphocytes at 8 weeks. Moreover, whereas inhibition of lymphangiogenesis did not aggravate interstitial fibrosis, it increased perivascular fibrosis and accelerated development of left ventricular (LV) dilation and dysfunction. In clinical HF samples, cardiac lymphatic density tended to increased, although lymphatic sizes decreased, notably in patients with dilated cardiomyopathy. Similarly, comparing C57Bl/6 and Balb/c mice, lymphatic remodeling post-TAC was linked to LV dilation rather than to hypertrophy. The striking lymphangiogenesis in Balb/c was associated with reduced cardiac levels of macrophages, B cells, and perivascular fibrosis at 8 weeks post-TAC, as compared with C57Bl/6 mice that displayed weak lymphangiogenesis. Surprisingly, however, it did not suffice to resolve myocardial edema, nor prevent HF development.
Collapse
Affiliation(s)
- C Heron
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - A Dumesnil
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - M Houssari
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - S Renet
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - T Lemarcis
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - A Lebon
- Normandy University, UniRouen, PRIMACEN, Mont Saint Aignan, France
| | - D Godefroy
- Normandy University, UniRouen, Inserm UMR1239 (DC2N Laboratory), Mont Saint Aignan, France
| | - D Schapman
- Normandy University, UniRouen, PRIMACEN, Mont Saint Aignan, France
| | - O Henri
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - G Riou
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1234 (PANTHER Laboratory), Rouen, France
| | - L Nicol
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - J P Henry
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - M Valet
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - M Pieronne-Deperrois
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - A Ouvrard-Pascaud
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - R Hägerling
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical and Human Genetics, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Augustenburger Platz 1, 13353 Berlin, Germany
| | - H Chiavelli
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - J B Michel
- UMR 1148, Inserm-Paris University, X. Bichat Hospital, Paris, France
| | - P Mulder
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - S Fraineau
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - V Richard
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - V Tardif
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| | - E Brakenhielm
- Normandy University, UniRouen, Inserm (Institut National de la Santé et de la Recherche Médicale) UMR1096 (EnVI Laboratory), FHU CARNAVAL, Rouen, France
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Abstract
Cardiac lymphangiogenesis plays an important physiological role in the regulation of interstitial fluid homeostasis, inflammatory, and immune responses. Impaired or excessive cardiac lymphatic remodeling and insufficient lymph drainage have been implicated in several cardiovascular diseases including atherosclerosis and myocardial infarction (MI). Although the molecular mechanisms underlying the regulation of functional lymphatics are not fully understood, the interplay between lymphangiogenesis and immune regulation has recently been explored in relation to the initiation and development of these diseases. In this field, experimental therapeutic strategies targeting lymphangiogenesis have shown promise by reducing myocardial inflammation, edema and fibrosis, and improving cardiac function. On the other hand, however, whether lymphangiogenesis is beneficial or detrimental to cardiac transplant survival remains controversial. In the light of recent evidence, cardiac lymphangiogenesis, a thriving and challenging field has been summarized and discussed, which may improve our knowledge in the pathogenesis of cardiovascular diseases and transplant biology.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Faculty of Welfare and Health Science, Oita University, Oita, 870-1192, Japan.
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Zhang J, Cui J, Li X, Hao X, Guo L, Wang H, Liu H. Increased secretion of VEGF-C from SiO 2-induced pulmonary macrophages promotes lymphangiogenesis through the Src/eNOS pathway in silicosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 218:112257. [PMID: 33933809 DOI: 10.1016/j.ecoenv.2021.112257] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/20/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
Silicosis, a type of lung inflammation and fibrosis caused by long-term inhalation of SiO2 particles, lacks effective treatment currently. Based on the results of our previous animal experiments, in lungs of SiO2-induced silicosis rats, a large number of lymphatic vessels are generated in the early stage of inflammation, which is of great significance for the removal of dust and inflammatory mediators. Here, the molecular mechanism of lymphangiogenesis is further studied. Vascular endothelial growth factor (VEGF-C) is a key pro-lymphangiogenic factor, and its elevated expression is closely related to lymphangiogenesis. In this investigation, we demonstrated that the protein level of VEGF-C was differentially expressed in bronchoalveolar lavage fluid (BALF) and alveolar macrophages (AM) in silicosis patients and healthy controls. We further stimulated human monocyte-macrophage line U937 with SiO2, collected the culture supernatants as conditioned medium (CM) for culturing lymphatic endothelial cells (LECs) in vitro, and observed the expression of VEGF-C in the supernatant and its effect on LEC tube formation. The results showed that both CM and single VEGF-C recombinant protein stimulation significantly enhanced LEC proliferation [(1.80 ± 0.18), (1.73 ± 0.16)], chemotaxis [chemotactic cell number (101.40 ± 13.83), (93.40 ± 9.61)], and tube formation [tube number (32.20 ± 7.26), (25.00 ± 6.25); branch number (77.20 ± 6.80), (84.60 ± 7.90)], whereas CM treated with VEGF-CmAb inhibited the proliferation (1.37 ± 0.17), chemotaxis [chemotactic cell number (57.40 ± 8.62)], and tube formation [tube number (7.40 ± 1.85); branch number (47.20 ± 13.44)] of LECs. In addition, CM and VEGF-C can promote the expression of vascular endothelial growth factor receptor 3 (VEGFR-3) and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) in LECs, which may further mediate lymphangiogenesis by up-regulating the Src/eNOS downstream signaling molecular pathway. This study is the first to clarify the molecular mechanism of pulmonary lymphangiogenesis in silicosis and may point in the direction of eventual treatments, surveillance, and regulation at a molecular level.
Collapse
Affiliation(s)
- Jinsong Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Xinying Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Lingli Guo
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hongli Wang
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| |
Collapse
|
23
|
Wu Q, Bai B, Tian C, Li D, Yu H, Song B, Li B, Chu X. The Molecular Mechanisms of Cardiotoxicity Induced by HER2, VEGF, and Tyrosine Kinase Inhibitors: an Updated Review. Cardiovasc Drugs Ther 2021; 36:511-524. [PMID: 33847848 DOI: 10.1007/s10557-021-07181-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
AIM In recent decades, there has been a revolutionary decrease in cancer-related mortality and an increase in survival due to the introduction of novel targeted drugs. Nevertheless, drugs targeting human epidermal growth factor receptor 2 (HER-2), angiogenesis, and other tyrosine kinases also come with unexpected cardiac side effects, including heart failure, hypertension, arterial thrombosis, and arrhythmias, and have mechanisms that are unlike those of classic chemotherapeutic agents. In addition, it is challenging to address some problems, as the existing guidelines need to be more specific, and further large-scale clinical trials and experimental studies are required to confirm the benefit of administering cardioprotective agents to patients treated with targeted therapies. Therefore, an improved understanding of cardiotoxicity becomes increasingly important to minimize the pernicious effects and maximize the beneficial effects of targeted agents. METHODS "Cardiotoxicity", "targeted drugs", "HER2", "trastuzumab", "angiogenesis inhibitor", "VEGF inhibitor" and "tyrosine kinase inhibitors" are used as keywords for article searches. RESULTS In this article, we report several targeted therapies that induce cardiotoxicity and update knowledge of the clinical evidence, molecular mechanisms, and management measures.
Collapse
Affiliation(s)
- Qinchao Wu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Bingxue Song
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Bing Li
- Department of Hematology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong, China.
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, Shandong, China.
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
24
|
Brakenhielm E, González A, Díez J. Role of Cardiac Lymphatics in Myocardial Edema and Fibrosis: JACC Review Topic of the Week. J Am Coll Cardiol 2021; 76:735-744. [PMID: 32762908 DOI: 10.1016/j.jacc.2020.05.076] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/23/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022]
Abstract
The cardiac lymphatic network plays a key role in regulation of myocardial extracellular volume and immune cell homeostasis. In different pathological conditions cardiac lymphatics undergo significant remodeling, with insufficient lymphatic function and/or lymphangiogenesis leading to fluid accumulation and development of edema. Additionally, by modulating the reuptake of tissue-infiltrating immune cells, lymphatics regulate immune responses. Available evidence suggests that both edema and inadequate immune response resolution may contribute to extracellular matrix remodeling and interstitial myocardial fibrosis. Interestingly, stimulation of lymphangiogenesis has been shown to improve cardiac function and reduce the progression of myocardial fibrosis during heart failure development after myocardial infarction. This review goes through the available clinical and experimental data supporting a role for cardiac lymphatics in cardiac disease, focusing on the current evidence linking poor cardiac lymphatic transport to the fibrogenic process and discussing potential avenues for novel biomarkers and therapeutic targets to limit cardiac fibrosis and dysfunction.
Collapse
Affiliation(s)
- Ebba Brakenhielm
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1096, Faculty of Medicine and Pharmacy, Rouen, France
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain; Departments of Nephrology and Cardiology, University of Navarra Clinic, Pamplona, Spain.
| |
Collapse
|
25
|
Song L, Chen X, Swanson TA, LaViolette B, Pang J, Cunio T, Nagle MW, Asano S, Hales K, Shipstone A, Sobon H, Al-Harthy SD, Ahn Y, Kreuser S, Robertson A, Ritenour C, Voigt F, Boucher M, Sun F, Sessa WC, Roth Flach RJ. Lymphangiogenic therapy prevents cardiac dysfunction by ameliorating inflammation and hypertension. eLife 2020; 9:e58376. [PMID: 33200983 PMCID: PMC7695461 DOI: 10.7554/elife.58376] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
The lymphatic vasculature is involved in the pathogenesis of acute cardiac injuries, but little is known about its role in chronic cardiac dysfunction. Here, we demonstrate that angiotensin II infusion induced cardiac inflammation and fibrosis at 1 week and caused cardiac dysfunction and impaired lymphatic transport at 6 weeks in mice, while co-administration of VEGFCc156s improved these parameters. To identify novel mechanisms underlying this protection, RNA sequencing analysis in distinct cell populations revealed that VEGFCc156s specifically modulated angiotensin II-induced inflammatory responses in cardiac and peripheral lymphatic endothelial cells. Furthermore, telemetry studies showed that while angiotensin II increased blood pressure acutely in all animals, VEGFCc156s-treated animals displayed a delayed systemic reduction in blood pressure independent of alterations in angiotensin II-mediated aortic stiffness. Overall, these results demonstrate that VEGFCc156s had a multifaceted therapeutic effect to prevent angiotensin II-induced cardiac dysfunction by improving cardiac lymphatic function, alleviating fibrosis and inflammation, and ameliorating hypertension.
Collapse
Affiliation(s)
- LouJin Song
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Xian Chen
- Comparative Medicine, Pfizer IncCambridgeUnited States
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Terri A Swanson
- Early Clinical Development, Pfizer IncCambridgeUnited States
| | | | - Jincheng Pang
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Teresa Cunio
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
- Acceleron PharmaCambridgeUnited States
| | - Michael W Nagle
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
- Eisai IncCambridgeUnited States
| | - Shoh Asano
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Arun Shipstone
- Inflammation and Immunology Research Unit, Pfizer IncCambridgeUnited States
| | - Hanna Sobon
- Inflammation and Immunology Research Unit, Pfizer IncCambridgeUnited States
| | - Sabra D Al-Harthy
- Comparative Medicine, Pfizer IncCambridgeUnited States
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Youngwook Ahn
- Target Sciences, Emerging Science and Innovation, Pfizer IncCambridgeUnited States
| | | | - Andrew Robertson
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Casey Ritenour
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Frank Voigt
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Furong Sun
- Early Clinical Development, Pfizer IncCambridgeUnited States
| | - William C Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of MedicineNew HavenUnited States
| | | |
Collapse
|
26
|
Zhou Y, Huang C, Hu Y, Xu Q, Hu X. Lymphatics in Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2020; 40:e275-e283. [PMID: 33085520 DOI: 10.1161/atvbaha.120.314735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Yijiang Zhou
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Chengchen Huang
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanhua Hu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Qingbo Xu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaosheng Hu
- From the Department of Cardiology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Abstract
OBJECTIVE Hypertension is associated with renal immune cell accumulation and sodium retention. Lymphatic vessels provide a route for immune cell trafficking and fluid clearance. Whether specifically increasing renal lymphatic density can treat established hypertension, and whether renal lymphatics are involved in mechanisms of blood pressure regulation remain undetermined. Here, we tested the hypothesis that augmenting renal lymphatic density can attenuate blood pressure in established hypertension. METHODS Transgenic mice with inducible kidney-specific overexpression of VEGF-D ('KidVD+' mice) and KidVD- controls were administered a nitric oxide synthase inhibitor, L-NAME, for 4 weeks, with doxycycline administration beginning at the end of week 1. To identify mechanisms by which renal lymphatics alter renal Na handling, Na excretion was examined in KidVD+ mice during acute and chronic salt loading conditions. RESULTS Renal VEGF-D induction for 3 weeks enhanced lymphatic density and significantly attenuated blood pressure in KidVD+ mice whereas KidVD- mice remained hypertensive. No differences were identified in renal immune cells, however, the urinary Na excretion was increased significantly in KidVD+ mice. KidVD+ mice demonstrated normal basal sodium handling, but following chronic high salt loading, KidVD+ mice had a significantly lower blood pressure along with increased urinary fractional excretion of Na. Mechanistically, KidVD+ mice demonstrated decreased renal abundance of total NCC and cleaved ENaCα Na transporters, increased renal tissue fluid volume, and increased plasma ANP. CONCLUSION Our findings demonstrate that therapeutically augmenting renal lymphatics increases natriuresis and reduces blood pressure under sodium retention conditions.
Collapse
|
28
|
Fehrenbach DJ, Mattson DL. Inflammatory macrophages in the kidney contribute to salt-sensitive hypertension. Am J Physiol Renal Physiol 2020; 318:F544-F548. [PMID: 31984790 DOI: 10.1152/ajprenal.00454.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review will highlight recent studies that have investigated the relationship between Na+, renal macrophage polarization, and renal damage. A hyperosmotic environment drives the macrophage toward a proinflammatory phenotype and away from an anti-inflammatory phenotype. Animal models of salt-sensitive hypertension demonstrate a characteristic infiltration of macrophages into the kidney that is greatly reduced when blood pressure is lowered. Because general immunosuppression or macrophage depletion leads to a host of adverse side effects, more recent studies have modulated the interaction of specific signaling molecules, including NOD-like receptor family pyrin domain-containing 3, chemokine (C-X-C motif) ligand 16, and VEGF, to prevent the end-organ renal damage that accumulates in salt-sensitive disease.
Collapse
Affiliation(s)
- Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, Georgia
| | - David L Mattson
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, Georgia
| |
Collapse
|
29
|
Oppelaar JJ, Vogt L. Body Fluid-Independent Effects of Dietary Salt Consumption in Chronic Kidney Disease. Nutrients 2019; 11:E2779. [PMID: 31731658 PMCID: PMC6893804 DOI: 10.3390/nu11112779] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
The average dietary salt (i.e., sodium chloride) intake in Western society is about 10 g per day. This greatly exceeds the lifestyle recommendations by the WHO to limit dietary salt intake to 5 g. There is robust evidence that excess salt intake is associated with deleterious effects including hypertension, kidney damage and adverse cardiovascular health. In patients with chronic kidney disease, moderate reduction of dietary salt intake has important renoprotective effects and positively influences the efficacy of common pharmacological treatment regimens. During the past several years, it has become clear that besides influencing body fluid volume high salt also induces tissue remodelling and activates immune cell homeostasis. The exact pathophysiological pathway in which these salt-induced fluid-independent effects contribute to CKD is not fully elucidated, nonetheless it is clear that inflammation and the development of fibrosis play a major role in the pathogenic mechanisms of renal diseases. This review focuses on body fluid-independent effects of salt contributing to CKD pathogenesis and cardiovascular health. Additionally, the question whether better understanding of these pathophysiological pathways, related to high salt consumption, might identify new potential treatment options will be discussed.
Collapse
Affiliation(s)
| | - Liffert Vogt
- Section of Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
30
|
Trincot C, Caron KM. Lymphatic Function and Dysfunction in the Context of Sex Differences. ACS Pharmacol Transl Sci 2019; 2:311-324. [PMID: 32259065 PMCID: PMC7089000 DOI: 10.1021/acsptsci.9b00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Indexed: 02/08/2023]
Abstract
Endothelial cells are the building blocks of the blood vascular system and exhibit well-characterized sexually dimorphic phenotypes with regard to chromosomal and hormonal sex, imparting innate genetic and physiological differences between male and female vascular systems and cardiovascular disease. However, even though females are predominantly affected by disorders of lymphatic vascular function, we lack a comprehensive understanding of the effects of sex and sex hormones on lymphatic growth, function, and dysfunction. Here, we attempt to comprehensively evaluate the current understanding of sex as a biological variable influencing lymphatic biology. We first focus on elucidating innate and fundamental differences between the sexes in lymphatic function and development. Next, we delve into lymphatic disease and explore the potential underpinnings toward bias prevalence in the female population. Lastly, we incorporate more broadly the role of the lymphatic system in sex-biased diseases such as cancer, cardiovascular disease, reproductive disorders, and autoimmune diseases to explore whether and how sex differences may influence lymphatic function in the context of these pathologies.
Collapse
Affiliation(s)
- Claire
E. Trincot
- Department of Cell Biology
and Physiology, University of North Carolina
Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building,
CB#7545, Chapel Hill, North
Carolina 27599-7545, United States
| | - Kathleen M. Caron
- Department of Cell Biology
and Physiology, University of North Carolina
Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building,
CB#7545, Chapel Hill, North
Carolina 27599-7545, United States
| |
Collapse
|