Open Access

Lymphangiogenesis: A new strategy for heart disease treatment (Review)

  • Authors:
    • Liding Bai
    • Yanyan Wang
    • Siqi Du
    • Yumeng Si
    • Lu Chen
    • Lin Li
    • Yuhong Li
  • View Affiliations

  • Published online on: February 22, 2024     https://doi.org/10.3892/ijmm.2024.5359
  • Article Number: 35
  • Copyright: © Bai et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Heart disease remains a global health challenge, contributing notably to morbidity and mortality. The lymphatic vasculature, an integral component of the cardiovascular system, plays a crucial role in regulating essential physiological processes, including fluid balance, transportation of extravasated proteins and immune cell trafficking, all of which are important for heart function. Through thorough scientometric analysis and extensive research, the present review identified lymphangiogenesis as a hotspot in cardiovascular disease research, and the mechanisms underlying impaired cardiac lymphangiogenesis and inadequate lymph drainage in various cardiovascular diseases are discussed. Furthermore, the way used to improve lymphangiogenesis to effectively regulate a variety of heart diseases and associated signaling pathways was investigated. Notably, the current review also highlights the impact of Traditional Chinese Medicine (TCM) on lymphangiogenesis, aiming to establish a clinical basis for the potential of TCM to improve cardiovascular diseases by promoting lymphangiogenesis.

1. Introduction

A number of diseases such as atherosclerosis (AS), myocardial infarction (MI), heart failure (HF), hypertensive heart disease (HHD) and congenital heart disease (CHD), profoundly impact cardiac health (1). Despite the numerous treatment options available, heart disease remains the leading global cause of mortality, requiring innovative therapeutic approaches. The data show that there were 4.58 million estimated heart disease deaths in 2020; the age-standardized mortality rate (ASMR) was 245.39 per 100,000 in 2020 (2,3). The discovery of lymphatic marker genes, such as acid receptor 1 (LYVE-1) and vascular endothelial growth factor (VEGF) receptor (VEGFR)-3, alongside advancements in lymphatic function imaging and quantification techniques, has brought attention to the role of lymphatic vessels in the progression of heart disease (4). The lymphatic system is an open, low-pressure, one-way transmission network between the extracellular space and the veins that differs from closed, hypertensive circulatory vascular networks. This extensive lymphatic network plays a key role in maintaining tissue fluid homeostasis and monitoring cardiac immunity (5,6). Obstruction of cardiac lymph flow leads to edema and cardiac dysfunction (7), underscoring the clinical significance of elucidating the role of the lymphatic system in cardiovascular diseases. In the field of information science, scientometrics, a highly specialized branch of bibliometrics, offers a quantitative method leveraging mathematical and statistical approaches to visualize emerging research trends and hotspots in scientific literature (8). This method aids in identifying publications, researchers, journals and research institutions that significantly contribute to a field, and enhances our understanding of scientific citations (9). In the present review, scientometrics was used to analyze research hotspots of lymphatic vessels in heart disease and evaluate the impact of the relevant literature. Additionally, a detailed review of molecular mechanisms surrounding these hotspots is presented to advance the development of drugs targeting lymphatic vessels.

2. Scientometric study

Data and methods

A search in the Web of Science (https://webofscience.clarivate.cn/wos/woscc/basic-search) core collection database for documents published from Jan 1, 2012 to August 1, 2023, was conducted using the key words 'heart disease' and 'lymphatic system'. A total of 197 documents were included, articles and reviews were selected, and other types of documents such as proceeding paper, editorial material, book chapters and others were excluded, and finally a total of 189 documents were used. Subsequently, a scientometric analysis was conducted on both articles and reviews. Raw data, encompassing comprehensive records and cited references, were downloaded from the Web of Science. Using CiteSpace (version 6.1.R2) (10), primary information such as publication numbers per year, countries, journals, key words and literature titles were extracted from the raw data. The data were visualized using Microsoft® Excel® 2021MSO and Hiplot (https://hiplot.com.cn/home/index.html) (Fig. 1).

General analysis

The search in the Web of Science core collection database revealed 189 publications related to the therapeutic effects of the lymphatic system on heart disease before August 1, 2023. On average, these documents were cited 28.06 times. In 2021, the highest number of articles was published, while the maximum number of citations was observed in 2013 (Fig. 2A). The observed trend indicates a growing body of research on the therapeutic effects of the lymphatic system on heart disease.

Country/region analysis

A total of 35 countries were represented in the publications included in the present review. The USA published the highest number of published articles, followed by United Kingdom, Germany, Italy, China, Japan, Finland, France, Brazil and Canada (Fig. 2B). Collaborative efforts were evident among several countries (Fig. 2C).

Publisher analysis

Publications originated from 53 different types of publishers, with the top 10 shown in Fig. 2D. Notably, Elsevier emerged as the most influential publisher, followed by Wiley, Springer Nature, Frontiers Media SA, MDPI, Lippincott Williams & Wilkins, American Physiological Society, BMJ Publishing Group, Public Library Science and International Society of Lymphology and the University of Arizona Libraries.

Key word analysis

Research key words reflect current trends, hotspots and the essence of a research document (11). The present review presents a co-occurrence network of key words (Fig. 3; Table I). In these clusters, 'lymphangiogenesis' emerged as a cross-key word, suggesting its potential impact on heart disease. The scientometric results further indicate that the lymphatic vessel is a research hotspot and potentially a promising treatment avenue for heart diseases, including MI, HF and CHD, with a focus on the cardiac lymphatic system.

Table I

Top 10 keyword co-occurrence networks.

Table I

Top 10 keyword co-occurrence networks.

RankingKey wordsNo. of occurrencesTotal link strength
1 Lymphangiogenesis31332
2Heart26217
3Congenital heart disease14157
4Lymphatic system13108
5Myocardial infarction678
6Heart failure675
7 Atherosclerosis569
8Cardiac lymphatics552
9Myocardial edema651
10Cardiac hypertrophy230
Literature and cited reference analysis

Citations play a crucial role in acknowledging sources during the writing or editing of a manuscript, including both the cited literature and the specific content of the citation (12). In response to the analysis of the key words, current research interests and topic hotspots were explored through literature analysis and references. Table II presents the top 10 most-cited articles on the therapeutic effect of lymphangiogenesis on heart diseases. The most influential article highlights the critical role of the lymphatic vascular system in maintaining interstitial fluid homeostasis, immune cell transport, nutrient lipid uptake and transport, and reverse cholesterol transportation (13). The second most-cited article underscores that selective stimulation of cardiac lymphangiogenesis post-MI markedly improves myocardial edema and restores cardiac function (14). Additionally, Table III outlines the top 10 cited references on the therapeutic roles of lymphangiogenesis in heart diseases. These articles offer a comprehensive overview of the most influential studies in the field for further research into the therapeutic effect of lymphangiogenesis on heart diseases.

Table II

Top 10 most-cited articles about the therapeutic effect of lymphangiogenesis on heart disease.

Table II

Top 10 most-cited articles about the therapeutic effect of lymphangiogenesis on heart disease.

Author(s), yearRankingCitedJournal
2022 Impact factor(Refs.)
NameCountry
Lohrke et al, 20161199Advances in TherapyUSA3.8(133)
Henri et al, 20162172CirculationUSA37.8(14)
Quintanilla et al, 2019391International Journal of Molecular SciencesSwitzerland5.6(134)
Aldea et al, 2019481Frontiers in Aging NeuroscienceSwitzerland4.8(135)
Brakenhielm and Alitalo, 2019574Nature Reviews CardiologyUK49.6(136)
Gordon-Walker et al, 2019670Journal of CardiologyJapan2.5(137)
Savla et al, 2017768Journal of the American College of CardiologyNetherlands24(138)
Goumans et al, 2018860Cold Spring Harbor Perspectives in BiologyUSA7.2(139)
Nash et al, 2016954Current Molecular MedicineUnited Arab Emirates2.5(140)
Melenotte et al, 20161052JAMA Network OpenUSA13.8(141)

Table III

Top 10 cited references about the therapeutic effect of lymphangiogenesis on heart disease.

Table III

Top 10 cited references about the therapeutic effect of lymphangiogenesis on heart disease.

Author(s), yearRankingCitedJournal
2022 Impact factor(Refs.)
NameCountry
Henri et al, 2016116CirculationUSA37.8(14)
Klotz et al, 2015216NatureUK64.8(120)
Dori et al, 2016313CirculationUSA37.8(13)
Aspelund et al, 2016411Circ ResUSA20.1(7)
Vieira et al, 2018511J Clin InvestUSA15.9(142)
Savla et al, 2017611J Am Coll CardiolNetherlands24(138)
Brakenhielm and Alitalo, 201978Nat Rev CardiolUK49.6(136)
Chavhan et al, 201788Nat Rev CardiolUSA5.5(143)
Oliver et al, 202098CellUSA64.5(17)
Trincot et al, 2019107Circ ResUSA20.1(44)

The clinical applications of lymphangiogenesis have been used to improve cardiac function (15). However, The clinical trial have yet to elucidate the specific signaling pathways regulating lymphangiogenesis for improved cardiac function. Therefore, animal experiments would be useful in establishing whether and how lymphangiogenesis improves cardiac function. The present review focused extensively on lymphangiogenesis in heart diseases. Furthermore, despite scientometric analysis indicating that lymphangiogenesis is a research hotspot and a promising treatment option for heart disease, a comprehensive and systematic exploration of relevant mechanisms is lacking. Therefore, in the current review, existing research at the mechanistic level on lymphangiogenesis and cardiovascular disease was examined.

3. Lymphangiogenesis and cardiovascular disease

Lymphangiogenesis and AS

AS is a chronic inflammatory disease characterized by the accumulation of cholesterol, lipoproteins and inflammatory cells in the arterial vessel wall (16). Lymphatics are present at atherosclerotic sites in the adventitia of the artery, adjacent to blood vessels and dilate within atherosclerotic plaques. With a deeper understanding of lymphatic vessel function, mounting evidence reveals the critical role of lymphangiogenesis in AS progression. Particularly, it is increasingly acknowledged that lymphatics are the main route for transporting high-density lipoprotein particles in cholesterol to the bloodstream (17). AS promotes lymphangiogenesis (18), which in turn, alleviates AS by facilitating reverse cholesterol transport and simultaneously draining local inflammation (19,20). The number and function of lymphatic vessels are limited. Consequently, promoting lymphangiogenesis holds great promise for the treatment of AS.

According to clinical and experimental studies, lymphangiogenesis in AS is regulated by multiple signaling pathways and molecules. Clinical evidence indicates that VEGF-C/D levels are notably and inversely associated with all-cause mortality among patients with suspected or known AS (21). Early treatment with VEGF-C inhibited plaque formation, promoting lymphatic molecular transport and triggering inflammatory cell migration (22). Besides VEGF-C/D, angiopoietin-2 (Angpt2) is associated with angiogenesis and inflammation. Clinical evidence suggests that the upregulation of Angpt2 may benefit patients with AS (23). This effect may be associated with the increase in VEGF-C/D secretion regulated by Angpt2, promoting lymphangiogenesis around atherosclerotic coronary arteries (23). R-spondin 2 (RSPO2), an important factor for regulating cell proliferation and differentiation, has previously shown potential in promoting lymphangiogenesis (24,25). A study indicated that the interaction between RSPO2 and G protein-coupled receptor 5 inhibits Wnt/β-catenin signaling (26), limiting lymphatic vessel formation, weakening lymphatic drainage of LDL cholesterol in the arterial wall and promoting AS development. A further study found that the phosphatidylinositol-3-hydroxykinase (PI3K)-protein kinase B (AKT) pathway is a downstream pathway of Wnt/β-catenin performs that promotes lymphangiogenesis (24). Additionally, the activation of the C-X-C chemokine receptor type 4 (CXCR4)/chemokine C-X-C motif ligand 12 (CXCL12) axis exacerbates inflammation by promoting leukocyte infiltration (27). A study suggested that the CXCL12/CXCR4 axis regulates cardiac adventitia lymphangiogenesis, reducing inflammation during AS by enhancing T-cell drainage (18). Details are provided in Table IV and Fig. 4.

Figure 4

Mechanisms and pathways in cardiovascular diseases and lymphangiogenesis (images partly modified from https://smart.servier.com). Lymphangiogenesis alleviates the pathological changes of the heart and blood vessels, including fibrosis, edema, and plaque formation, by reversing cholesterol transport, draining local inflammation, and eliminating excessive interstitial fluid, thereby playing a crucial role in delaying the progression of cardiovascular diseases. Numerous pathways and mechanisms are involved in the regulation of lymphangiogenesis. ERα, estrogen receptor α; JNK, c-Jun N-terminal kinase; AM, adrenomedullin; Cx43, connexin 43; Tβ4, thymosin β 4; EPDCs, epicardium-derived cells; APJ, Apelin receptor; Ang II, angiopoietin-2; SPNS2, spinster homolog 2; VEGF-C, vascular endothelial growth factor C; S1P, sphinogosine-1-phosphate; S1PR1, sphinogosine-1-phosphate receptor 1; ERK, extracellular regulated protein kinase; CXCL12, chemokine C-X-C motif ligand 12; AKT, protein kinase B; Nfatc1, nuclear factor of activated T-cells, cytoplasmic 1; FOXC2, fork head box C2; VCAM-1, vascular cell adhesion molecule 1; CFs, cardiac fibroblasts; IL-1β, interleukin-1β; COX-2, cyclooxygenase 2; PGE2, prostaglandin 2; AT1R, angiotensin II type1 receptor; MKP5, mitogen-activated protein kinase phosphatases 5; MAPK, mitogen-activated protein kinase; SIRT3, silencing regulatory protein 3; TonEBP, tonicity enhancer binding protein; CXCR4, C-X-C chemokine receptor type 4; Angpt2, angiotensin II; RSPO2, R-spondin 2; LGR4, leucine-rich repeat containing G protein-coupled receptor 4; PI3K, phosphatidylinositol-3-hydroxykinase; KLF2, Kruppel factor 2; PPAR-γ, peroxisome proliferator-activated receptor-γ; NO, nitric oxide; HIF-α: hypoxia-inducible factor 1-α.

Table IV

Signal pathways related to lymphangiogenesis in cardiovascular diseases.

Table IV

Signal pathways related to lymphangiogenesis in cardiovascular diseases.

Author(s), yearHeart diseaseModel
Lymphatic endothelial cell phenotypeMolecular targets/mechanisms(Refs.)
Animal strainsCell lines
Milasan et al, 2019 AtherosclerosisLdlr−/− female miceHMVEC-dLyAdDensity migration permeability contractilityVEGFR3-FOXC2(22)
Drosos et al, 2019Homo sapiensHDLECsSprouts ANGPT2/VEGF-C/VEGF-D/LEVY-1/podoplanin(23)
Singla et al, 2021C57BL/6 male ApoE−/−HDLECsSuppresses lymphangiogenesisRSPO2–LGR5-NO Wnt-β-catenin(24)
Rademakers et al, 2017Homo sapiens male ApoE−/−Unknown Lymphangiogenesis CXCL12/CXCR4-VEGFR3(18)
Li et al, 2022Myocardial infarction S1pr1flox/flox C57BL/6HDLECsMigration, proliferation S1P/S1pr1/ERK/CCL2(35)
Mahmoodzadeh et al, 2014ERα overexpressionVentricular cardiomyocytesSprout lymphangiogenesisER-alpha (ERα)-JNK-VEGF(38)
Tatin et al, 2017Apelin-KO C57BL/6HDLECsLymphatic barrier integrity apelin-S1P/SPNS2(41)
Wang et al, 2021 WT1CreERT2/+ Rosa26mTmG/+EPDCsPromote differentiationTβ4-EPDCs(43)
Trincot et al, 2016Adm-overexpression (Admhi/hi) C57BL/6HDLECsProliferation Adrenomedullin-Connexin 43(44)
Lin et al, 2021 Lyve-1EGFP/cre VEGFR-3f/f VEGFR-3f/−NRCMs, HUVECs mouse primary macrophages, mouse LECsProliferationVEGF-C-VEGFR-3 ATK/ERK、CaNA/NFATc1/FOXC2/CX43(45)
Al-Kofahi et al, 2018Heart failureC3H/HeN mice MaleRCLMCsContractility IL-1β/TNF-α-COX-2/PGE2(58)
Bai et al, 2022 Lyve-1Cre VEGFR-3f/−HDLECsPermeability, lymphangiogenesis MKP-5/p38MAPK/VE-cadherin(61)
Iwamiya et al, 2020F344/N Jcl-rnu/rnu ratsHUVECsTube formation, lymphangiogenesiscardiac fibroblasts-VCAM1(63)
Yang et al, 2014Hypertensive heart diseaseMale SHRs and WKYUnknown LymphangiogenesisVEGFC-VEGFR3(70)
Yang et al, 2017Male SHR and WKYHDLECs LymphangiogenesisTonEBP/VEGF-C(72)
Zhang et al, 2021SIRT3-KOMouse LECsMigration, proliferation SIRT3-VEGFC-VEGFR3(73)
Boehme et al, 2021Congenital heart diseaseOvine modelHDLECs LymphangiogenesisVEGFR2/VEGFR3/VE Cadherin(78)
Choi et al, 2017LambPrimary lymphatic endothelial cell linesLymph functionKLF2-PPAR-γ-Lymph flow(81)

[i] HMVEC-dLyAd, primary human dermal lymphatic microvascular endothelial cells adult; EPDCs, epicardium-derived cells; LECs, lymphatic endothelial cells; HDLECs, human dermal lymphatic endothelial cells; HUVECs, human umbilical vein endothelial cells; NRCMs, neonatal-rat-cardiomyocytes; RCLMCs, rat cardiac lymphatic muscle cells; KO, knockout; SHR, spontaneously hypertensive rats; WKY, Winstar-Kyoto.

Lymphangiogenesis and MI

The rupture of an unstable atherosclerotic plaque and subsequent coronary thrombosis often leads to MI (28). MI is often accompanied by myocardial edema, inflammation and fibrosis (29). Cardiac lymphatic flow begins from small endocardium lymphatic vessels, traverses through the myocardium and enters epicardial capillaries. These capillaries converge into large collecting lymphatic vessels, facilitating the transportation of excess water and immune cells, and inhibiting myocardial edema, inflammation and fibrosis (6). Lymphatic vessel formation is investigated as a potential target for MI in clinical practice (30). Lymphatic vessels play a crucial role in heart repair after MI, maintaining tissue fluid homeostasis and suppressing excessive immune responses (31). The VEGF family and their endothelial tyrosine kinase receptors induce lymphangiogenesis (32), but their efficiency is limited when injected intravenously or intracoronarily after MI due to short half-lives (33). Thus, researchers are exploring new pathways new pathways regulating lymphangiogenesis.

VEGF-C-VEGFR3, sphinogosine-1-phosphate (S1P)/S1P receptor 1 (S1PR1) is another noteworthy receptor-ligand complex. S1PR1 is a G-protein-coupled receptor, expressed in lymphatic endothelial cells, and modulates immune cell transportation (34). In injured hearts, S1PR1/S1P activates downstream signals, including extracellular regulated protein kinases/chemokine ligand 2 (ERK), promoting lymphangiogenesis in the injured heart, driving macrophage trafficking, immune modulation, and cardiac repair after MI (35,36). Estrogen receptor α (ERα)/estrogen ligand plays a protective role in MI, associated with regulating lymphangiogenesis (37). Overexpression of cardiomyocyte-specific ERα promotes lymphangiogenesis via inducing c-Jun N-terminal kinase phosphorylation, alleviating ventricular fibrosis (38). Spinster homolog 2 (SPNS2) and thymosin beta 4 (Tβ4) drive lymphangiogenesis by controlling lymphatic endothelial cell proliferation and differentiation. SPNS2, an S1P transporter, contributes to S1P release into extracellular space and the development of lymphatic vessels (39,40). Apelin/Apelin receptor signaling targets SPNS2, promoting lymphatic endothelial cell proliferation and maintaining lymphatic endothelial cell integrity, limiting persistent edema and inflammation after MI (41). Tβ4, which is a 43-amino-acid-sequestering peptide in the embryonic heart, is important for epicardial development and coronary artery formation (42). Tβ4 promotes the differentiation of epicardium-derived cells towards lymphatic endothelial cells, attenuating adverse myocardial remodeling, enhancing myocardial regeneration, and improving cardiac function post-MI (43). Connexin43 (Cx43) may control the function of nascent lymphatic vessels by determining endothelial cell connections. Cx43 modulates gap connection coupling between lymphatic endothelial cells and has been implicated in the pathological process of MI. A previous study showed that adrenomedullin targeted Cx43 to increase gap connections between human lymphatic endothelial cells, promoting the proliferation of the cardiac lymphatic vascular system and relieving MI-induced edema (44). Additionally, VEGF-C-VEGFR-3 indirectly promotes lymphangiogenesis, polarizing macrophage from M1 to M2 in the MI area via activation of downstream AKT/ERK1/2 and calcineurin/nuclear factor of activated T cells 1, forkhead box C2 signaling pathways, accelerating post-MI repair (45). Furthermore, when dead cells are engulfed by macrophages, more VEGF-C is released, further promoting myocardial lymphangiogenesis through its receptor (46). Details are provided in Table IV and Fig. 4.

Lymphangiogenesis and HF

At the end stage of heart disease, the clinical manifestation is HF, not a single pathological diagnosis but a clinical syndrome characterized by breathlessness, ankle swelling, fatigue, pulmonary crackles, peripheral edema and elevated jugular venous pressure (47,48). Common pathological manifestations of HF include myocarditis, myocardial edema, and myocardial fibrosis (49-51). Excessive inflammatory infiltration forms the biological basis for these manifestations (52), and edema results as a major consequence of the inflammatory response in the myocardium (53). Lymphatics are crucial to the progression of HF. Cardiac lymphatics play a crucial role in the progression of HF by adapting their number and function to the increased demand for inflammatory factors and fluid drainage, thus alleviating inflammation and edema (54,55).

Most studies suggest that VEGF-C-VEGFR3 signaling, as the key pathway of lymphangiogenesis, may be a therapeutic target for HF (45). Additionally, interleukin-1β (IL-1β), an inflammatory cytokine associated with MI, has been reported to exert a profound depressant effect on the contractility of lymphatic muscles (56). Prostaglandin E2 (PGE2), the main downstream product of cyclooxygenase-2 activation, induces relaxation by binding to PGE2 to prostaglandin E receptor 2 or 4, elevating intracellular cyclic adenosine monophosphate accumulation (57). A recent study showed that IL-1β reduced the contractility of cardiac lymphatic muscle cells through COX-2/PGE2 signal transduction, weakening the transport of local inflammation in the heart and promoting the progression of acute myocarditis to HF (58). Furthermore, angiotensin-2 (Ang II), is an important factor in inducing inflammation in the vascular system (59). Angiotensin, serotonin, and endothelin are also known to regulate lymphatic vessels (60). Ang II can activate AT1R to promote the expression and activity of proteasome catalytic subunits β2i and β5i, affecting MKP5 and VE-cadherin degradation and p38MAPK activation, resulting in lymphatic endothelial hyperpermeability (61). Moreover, cardiac fibroblasts, principal contributors to cardiac fibrosis leading to cardiac remodeling and HF, express vascular cell adhesion molecule-1 and promote lymphangiogenesis (62). This mobilizes lymphatic endothelial cells into the infarct zone, restoring ventricular wall mechanical properties. The mechanism may involve the formation of lymphatic vessels, inhibition of interstitial edema, and consequently, reduced inflammation and myocardial fibrosis (63). Details are provided in Table IV and Fig. 4.

Lymphangiogenesis and HHD

HHD is an organic heart disease caused by long-term poorly controlled hypertension, characterized by cardiac hypertrophy due to progressively increasing ventricular load (64). Studies indicated that high sodium intake contributes to hypertension development (65). The large accumulation of sodium leads to electrolyte disorders and promotes edema formation (66). Lymphatics is a key route in draining interstitial fluid, and in HHD, lymphangiogenesis is an effective strategy for reducing cardiac interstitial edema. As a cytokine regulating lymphangiogenesis, VEGF-C relieves edema caused by electrolyte imbalances (67). VEGF-C treatment notably improves cardiac function, preserving cardiac function, reducing cardiac hypertrophy and fibrosis, accelerating edema clearance by inducing skin lymphangiogenesis and enhancing endothelial nitric oxide (NO) synthase expression in blood vessels to lower blood pressure (68).

Lymphangiogenesis plays an important role not only in maintaining interstitial fluid balance but also in facilitating immune cell transport in HHD. The activation of the VEGF-C-VEGFR3 pathway is activated in mononuclear phagocytes, triggered by sodium salt accumulation, and stimulates skin lymphangiogenesis (69). Systemic overexpression of VEGF-C enhances lymphangiogenesis within the myocardial interstitium, attenuating local infiltration of macrophages in the heart (70). This dual mechanism of eliminating excess interstitial fluid and transporting immune cells contributes to the reduction of cardiac decompensation-induced myocardial edema and inflammation. In HHD, tonicity enhancer binding protein (TonEBP) and silencing regulatory protein 3 (SIRT3) play key roles in the upstream regulation of VEGFC-VEGFR3 signaling. TonEBP, a key regulator of cellular responses to hypertonic stress, binds to the promoter region of VEGF-C, enhancing its secretion and promoting lymphangiogenesis (71). This, in turn, inhibits left ventricular remodeling induced by high salt intake (72). SIRT3 is a major mitochondrial deacetylase closely associated with angiogenesis. A recent study indicated that SIRT3 activation upregulates the VEGF-C-VEGFR3 signaling axis protein expression, thereby promoting migration and proliferation of lymphatic endothelial cells (LECs), and lymphangiogenesis in AngⅡ-induced hypertensive heart models (73,74). Details are provided in Table IV and Fig. 4.

Lymphangiogenesis in CHD

CHD, a common congenital cardiovascular anomaly (75), significantly affects the lymphatic system due to associated hemodynamic changes (76). The development of lymphatic structures relies on mechanical forces generated by fluid accumulation in the interstitial space. However, chronic elevation of lymph flow can lead to lymphatic system dysfunction under prolonged mechanical stimuli (77). In models of CHD with increased pulmonary blood flow, mechanical stimulation and shear stress upregulate HIF-1 α expression and induce structural disorders in LECs (78). NO plays a pivotal role in lymphangiogenesis and lymphatic function maintenance, with a notable reduction in NO bioavailability observed in congenital heart defect models (79). PPAR-γ, a ligand-activated transcription factor, increases NO bioavailable in LECs (80). Kruppel Factor 2 (KLF2) induces LEC proliferation and lymphatic sprouting under laminar flow conditions (81). The KLF2-mediated PPAR-γ signal, however, inhibits NO production, aggravating lymphatic vessel dysfunction (82). Understanding the role of lymphatic vessels in hemorheology provides innovative treatment strategies for congenital heart defects and other lymphatic disorders. Details are provided in Table IV and Fig. 4.

4. Traditional Chinese Medicine regulating lymphangiogenesis

The rapid advancements in modernization research within Traditional Chinese Medicine (TCM) have garnered international recognition for its efficacy in disease treatment. TCM is characterized by personalized treatment, fewer side effects and a focus on treating both symptoms and root causes and promoting self-healing ability of the body. A study reported the explicitly regulatory effects of TCM on lymphatic vessels, particularly in promoting lymphangiogenesis (83).

Due to its core role in tissue inflammation (20), lymphangiogenesis holds the potential to augment the excretion of extracellular fluid, thereby reducing edema during both acute and chronic inflammatory attacks (84). Han et al (85) and Chen et al (86) identified that Du-huo-ji-sheng-tang and Fang-Ji-Huang-Qi-Tang exhibit the capability to enhance lymphangiogenesis and promote lymphatic drainage function. This enhancement proves pivotal in alleviating local edema and tissue damage caused by inflammation. Additionally, a study demonstrated the efficacy of ginsenoside Rg1, a tetracyclic triterpenoid compound extracted from ginseng, in promoting the recovery of lymphatic drainage function, leading to a reduction in joint swelling (87).

Several studies highlight the efficacy of Chinese herbal extracts and monomers in alleviating lymphedema by promoting lymphangiogenesis. Ishii et al (88) reported that piper retrofractum extract and piperine, isolated from piper retrofractum, notably enhanced the proliferation, migration and tube formation in human dermal lymphatic microvascular endothelial cells, thereby promoting lymphangiogenesis through the AKT and ERK pathways and effectively alleviating lymphedema. T total saponin of Panax notoginseng (PNS) can markedly promote lymphangiogenesis, and its mechanism may be related to the activation of the ERK1/2, PI3K and P38-MAPK signaling pathways, which stimulate the release of VEGF-C. PNS is a promising therapeutic option for managing secondary lymphedema or other lymphatic system disorder (89).

Notably, there is a limited number of TCM studies addressing the regulation of lymphangiogenesis in the treatment of cardiovascular diseases (90). Studies have indicated that salvianolic acid B (SAB), a water-soluble phenolic compound isolated from Danshen, exhibits therapeutic effects on dilated cardiomyopathy, which is associated with lymphangiogenesis deficiency (91). Mechanistically, SAB promotes the proliferation and migration of LECs by upregulating the TNF-α/NF-κB/VEGF-C signaling pathway, thus regulating the formation of lymphatic vessels and inhibiting the development of the pathological process of dilated heart disease (92).

The lymphatic vessel emerges as a key conduit for local inflammation, playing an important role in various diseases, including osteoarthritis, lymphedema and cardiovascular disease (31,93). Notably, a study also highlighted the rich distribution of lymphatic vessels in the cornea (94). Furthermore, the discovery of meningeal lymphatic vessels challenges the prevailing notion of the absence of lymphatic systems in the brain. This discovery opens up new avenues for efficiently eliminating metabolic products produced by brain parenchyma (95), offering potential treatment ideas for Alzheimer's disease, insomnia and meningitis (96-98).

A study indicated a growing interest in the regulation of lymphatic vessels by TCM (99). Although current research primarily focuses on arthritis and lymphedema, there are numerous promising signs that TCM can improve microcirculation in the eyes and brain, while also regulating local oxidative stress and immune responses (100,101). These findings prompt us to consider the potential role of TCM in regulating lymphatic vessels, providing a promising perspective for further exploration.

5. Lymphatic dysfunction as the biological basis of phlegm/phlegm dampness in TCM

According to TCM theory, all normal bodily fluids, including inter-tissue fluids and fluids within internal organs fluids such as gastric and intestinal fluids, are categorized as body fluids (102). The generation, distribution and excretion of body fluids are intricately balanced in a healthy body. An imbalance between body fluid generation and excretion can lead to insufficient body fluid and symptoms such as thirst and dry skin (103). TCM syndromes, including phlegm dampness or phlegm retention, stem from abnormal fluid distribution and slow flow within the body (104).

Understanding the biological basis of TCM syndrome has long presented a scientific challenge. Some authors propose that the biological basis of phlegm retention or phlegm dampness lies in the accumulation of tissue fluid (105), hyperlipidemia (106), aggregation of adhesion factors (107) and localized immune inflammation (108). Modern medicine recognizes lymphatic functions that involve the clearance of low-density lipoprotein in macrophages, transportation of immune cells and drainage of tissue fluid (109). Phlegm and dampness in TCM can also be understood as local inflammation, lipid accumulation and fluid retention resulting from the obstruction of lymphatic reflux (110). Through extensive study of phlegm and dampness, it is hypothesized that obstruction of lymphatic reflux might offer a more appropriate explanation.

6. Conclusion and future perspectives

Scientometrics is a branch of information science that uses quantitative analysis and statistics to study scholarly literature, including publications, citations and author collaboration. Researchers can use it to identify research trends in a specific area, assess the impact of related research, understand research progress and identify partners. In the present review, scientometric analysis was applied to identify new research hotspots in cardiovascular-related diseases, specifically analyzing the therapeutic role of lymphangiogenesis. The lymphatic system, as another important circulatory pathway alongside blood circulation, facilitates the circulation of body fluids together with the blood circulatory system. It achieves this by transporting lymphatic fluid containing water and macromolecules, contributing to overall body metabolism, while maintaining the stability of internal lymphangiogenesis (111). Currently, it is well established that the VEGF family is one of the potent lymphangiogenic factors (112). Additional factors, including IL-6 (113), platelet-derived growth factor (114), neuropilin-2 (115) and calcium-binding EGF domain-containing protein 1 (116) have also been identified for their role in stimulating lymphangiogenesis. Various indirect factors, such as activated macrophages secreting lymphatic vessel-promoting factors, also influence lymphangiogenesis (117). Considerable attention is currently directed towards the VEGF-C/D-VEGFR3 pathway as a potential therapeutic target. In relevant experiments, recombinant VEGF-C-C156S has demonstrated the ability to promote lymphangiogenesis and improve prognosis after MI. Furthermore, certain proteins, such as Polydom (118) and Notch (119) play an important role in lymphangiogenesis. Therefore, gaining a better understanding of the cellular and molecular mechanisms underlying specific lymphatic vessel diseases, coupled with the development of new drugs, holds the potential to limit the development and progression of heart diseases.

The therapeutic strategy aimed at specifically targeting lymphangiogenesis in treating heart disease has been thoroughly validated in animal models. This includes significant improvements in lipid transport, cardiac function, and immune response observed after heart transplantation through the administration of VEGF-C (120-122). However, clinical studies on the treatment of heart disease with lymphatic neovascularization, particularly involving VEGF, remain limited. An injection of VEGF-2 into the endocardium has shown notable improvement in angina pectoris symptoms and was proven to be relatively safe. Conducting a larger phase III trial is deemed necessary for further validation (123). AdVEGF-DΔNΔC gene therapy is safe, well tolerated, and notably improves myocardial perfusion reserve, enhancing the quality of life for patients with MI during year 1 of follow-up (124). Moreover, researchers are actively exploring changes in drug dosage forms to target lesion sites and enhance drug efficacy. This includes innovative approaches such as using nano and hyaluronic acid hydrogel-wrapped VEGF to achieve site-specific delivery. This method of drug delivery not only avoids the short half-life associated with intravenous VEGF injection, but also alleviates the disadvantages of easy degradation in vivo, making it suitable for clinical lymphangiogenesis treatment (125,126). In conclusion, the exploration of lymphatic neovascularization factors and related pathways, along with the development of corresponding therapeutic drugs and dosage forms, represents a promising therapeutic strategy for heart disease treatment.

TCM, as a natural medicine, holds a marked advantage in regulating lymphangiogenesis. Current research indicates that TCM exerts remarkable anti-inflammatory effects by regulating lymphatic neovascularization (99). A major effect of TCM lies in its ability to regulate lymphatic vessel function, including lipid transport, tissue fluid homeostasis and immunity (127). The structure of lymphatic vessels plays a crucial role in their function. The mature lymphatic network comprises initial lymphatics, precollectors and collecting ducts (128). Due to structural differences, these vessels serve distinct functions. Blind-ended initial lymphatics take up lymphatic fluid containing fluid, macromolecules and immune cells (129). Precollectors and collecting ducts possess valvular structures that effectively prevent lymph fluid reflux (130). At present, TCM research on lymphatic vessels mainly focuses on their quantity and function, with limited attention to their structures and mechanisms (99). Consequently, future research should focus on further exploring the structure of lymphatic vessels to enhance our understanding of the way TCM operates. Although lymphangiogenesis is gradually recognized as a novel target for intervention in cardiovascular diseases, relatively few studies have been reported on the TCM for the treatment of cardiovascular diseases focusing on lymphangiogenesis (90) By contrast, TCM has been reported to treat arthritis and lymphedema by promoting lymphangiogenesis (131,132). Since lymphangiogenesis helps to impede the pathological processes of cardiovascular diseases, including fibrosis, edema and plaque formation by reversing cholesterol transport, draining local inflammation and eliminating excessive interstitial fluid, the role of TCM in the regulation of lymphangiogenesis will be a promising research direction. While the majority of drugs are absorbed into the bloodstream after further breakdown in the intestines, lymphatic vessels are widely distributed throughout the intestinal tract, ultimately directing lymph into the veins. It would be of interest to investigate the presence of small molecules of TCM in lymphatic fluid, enabling direct administration of drugs through lymphatic vessels. This approach could notably reduce the first-pass effect of drugs, thereby enhancing their efficacy.

Availability of data and materials

Not applicable.

Authors' contributions

BL carried out investigation and visualization, and wrote the original draft of the manuscript. WY conceptualized the study, acquired funding, developed methodology and wrote the original draft of the manuscript. DS carried out investigation and data curation. SY carried out investigation and used software. CL acquired funding, reviewed the manuscript and completed language editing. LL developed methodology, supervised the project, and reviewed and edited the manuscript. LY conceptualized the study, supervised the project, and reviewed and edited the manuscript. All authors have read and approved the final version of the manuscript. Data authentication not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Abbreviations:

AS

atherosclerosis

Angpt2

angiopoietin-2

AKT

protein kinase B

CXCL12

chemokine C-X-C motif ligand 12

CXCR4

C-X-C chemokine receptor type 4

CHD

congenital heart disease

Cx43

connexin43

ERK

extracellular regulated protein kinase

ERα

estrogen receptor α

HHD

hypertensive heart disease

HF

heart failure

IL-1β

interleukin-1β

KLF2

Kruppel factor 2

LEC

lymphatic endothelial cell

MI

myocardial infarction

NO

nitric oxide

PGE2

prostaglandin E2

PI3K

phosphatidylinositol-3-hydroxykinase

RSPO2

R-spondin 2

S1P

sphinogosine-1-phosphate

SPNS2

spinster homolog 2

S1PR1

sphinogosine-1-phosphate receptor 1

SIRT3

silencing regulatory protein 3

Tβ4

thymosin β 4

TCM

Traditional Chinese medicine

TonEBP

tonicity enhancer binding protein

VEGF-C/D

vascular endothelial growth factor-C/D

Acknowledgments

The authors would like to thank Shanghai Tengyun Biotechnology Co., Ltd. for providing technical assistance and valuable tools for data analysis and visualization using the Hiplot Pro platform (https://hiplot.com.cn/).

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 82204867 and 82174205) and Tianjin Municipal Health Commission (grant no. 2023145).

References

1 

Wang J, Duan Y, Sluijter JP and Xiao J: Lymphocytic subsets play distinct roles in heart diseases. Theranostics. 9:4030–4046. 2019.

2 

Brownrigg JR, Leo V, Rose J, Low E, Richards S, Carr-White G and Elliott PM: Epidemiology of cardiomyopathies and incident heart failure in a population-based cohort study. Heart. 108:1383–1391. 2022.

3 

Wang W, Liu Y, Liu J, Yin P, Wang L, Qi J, You J, Lin L, Meng S, Wang F and Zhou M: Mortality and years of life lost of cardiovascular diseases in China, 2005-2020: Empirical evidence from national mortality surveillance system. Int J Cardiol. 340:105–112. 2021.

4 

Harris NR, Bálint L, Dy DM, Nielsen NR, Méndez HG, Aghajanian A and Caron KM: The ebb and flow of cardiac lymphatics: A tidal wave of new discoveries. Physiol Rev. 103:391–432. 2023.

5 

Monaghan RM, Page DJ, Ostergaard P and Keavney BD: The physiological and pathological functions of VEGFR3 in cardiac and lymphatic development and related diseases. Cardiovasc Res. 117:1877–1890. 2021.

6 

Vuorio T, Ylä-Herttuala E, Laakkonen JP, Laidinen S, Liimatainen T and Ylä-Herttuala S: Downregulation of VEGFR3 signaling alters cardiac lymphatic vessel organization and leads to a higher mortality after acute myocardial infarction. Sci Rep. 8:167092018.

7 

Aspelund A, Robciuc MR, Karaman S, Makinen T and Alitalo K: Lymphatic system in cardiovascular medicine. Circ Res. 118:515–530. 2016.

8 

Nair AS: Scientometrics in medical journals: Indices, their pros and cons. Indian J Anaesth. 63:955–957. 2019.

9 

Martinez P, Al-Hussein M and Ahmad R: A scientometric analysis and critical review of computer vision applications for construction. Autom Constr. 107:1029472019.

10 

Chen C: CiteSpace: A practical guide for mapping scientific literature. New York: Nova Science Publishers; 2016

11 

Bilal, Hysa E, Akbar A, Yasmin F, Rahman AU and Li S: Virtual learning during the covid-19 pandemic: A bibliometric review and future research agenda. Risk Manag Healthc Policy. 15:1353–1368. 2022.

12 

Force MM and Robinson NJ: Encouraging data citation and discovery with the data citation index. J Comput Aided Mol Des. 28:1043–1048. 2014.

13 

Dori Y, Keller MS, Rome JJ, Gillespie MJ, Glatz AC, Dodds K, Goldberg DJ, Goldfarb S, Rychik J and Itkin M: Percutaneous lymphatic embolization of abnormal pulmonary lymphatic flow as treatment of plastic bronchitis in patients with congenital heart disease. Circulation. 133:1160–1670. 2016.

14 

Henri O, Pouehe C, Houssari M, Galas L, Nicol L, Edwards-Lévy F, Henry JP, Dumesnil A, Boukhalfa I, Banquet S, et al: Selective stimulation of cardiac lymphangiogenesis reduces myocardial edema and fibrosis leading to improved cardiac function following myocardial infarction. Circulation. 133:1484–1497. 2016.

15 

Fudim M, Salah HM, Sathananthan J, Bernier M, Pabon-Ramos W, Schwartz RS, Rodés-Cabau J, Côté F, Khalifa A, Virani SA and Patel MR: Lymphatic dysregulation in patients with heart failure: JACC review topic of the week. J Am Coll Cardiol. 78:66–76. 2021.

16 

Liu MM, Peng J, Guo YL, Zhu CG, Wu NQ, Xu RX, Dong Q, Cui CJ and Li JJ: SORBS2 as a molecular target for atherosclerosis in patients with familial hypercholesterolemia. J Transl Med. 20:2332022.

17 

Oliver G, Kipnis J, Randolph GJ and Harvey NL: The lymphatic vasculature in the 21st century: Novel functional roles in homeostasis and disease. Cell. 182:270–296. 2020.

18 

Rademakers T, van der Vorst EP, Daissormont IT, Otten JJ, Theodorou K, Theelen TL, Gijbels M, Anisimov A, Nurmi H, Lindeman JH, et al: Adventitial lymphatic capillary expansion impacts on plaque T cell accumulation in atherosclerosis. Sci Rep. 7:452632017.

19 

Nielsen NR, Rangarajan KV, Mao L, Rockman HA and Caron KM: A murine model of increased coronary sinus pressure induces myocardial edema with cardiac lymphatic dilation and fibrosis. Am J Physiol Heart Circ Physiol. 318:H895–H907. 2020.

20 

Chakraborty A, Scogin CK, Rizwan K, Morley TS and Rutkowski JM: Characterizing lymphangiogenesis and concurrent inflammation in adipose tissue in response to VEGF-D. Front Physiol. 11:3632020.

21 

Wada H, Suzuki M, Matsuda M, Ajiro Y, Shinozaki T, Sakagami S, Yonezawa K, Shimizu M, Funada J, Takenaka T, et al: VEGF-C and mortality in patients with suspected or known coronary artery disease. J Am Heart Assoc. 7:e103552018.

22 

Milasan A, Smaani A and Martel C: Early rescue of lymphatic function limits atherosclerosis progression in Ldlr−/− mice. Atherosclerosis. 283:106–119. 2019.

23 

Drosos I, Pavlaki M, Ortega Carrillo MDP, Kourkouli A, Buschmann K, Konstantinou F, Gogiraju R, Bochenek ML, Chalikias G, Tortopidis C, et al: Increased lymphangiogenesis and lymphangiogenic growth factor expression in perivascular adipose tissue of patients with coronary artery disease. J Clin Med. 8:10002019.

24 

Singla B, Lin HP, Chen A, Ahn W, Ghoshal P, Cherian-Shaw M, White J, Stansfield BK and Csányi G: Role of R-spondin 2 in arterial lymphangiogenesis and atherosclerosis. Cardiovasc Res. 117:1489–1509. 2021.

25 

Okura T, Ohkawara B, Takegami Y, Ito M, Masuda A, Seki T, Ishiguro N and Ohno K: Mianserin suppresses R-spondin 2-induced activation of Wnt/β-catenin signaling in chondrocytes and prevents cartilage degradation in a rat model of osteoarthritis. Sci Rep. 9:28082019.

26 

Wu C, Qiu S, Lu L, Zou J, Li WF, Wang O, Zhao H, Wang H, Tang J, Chen L, et al: RSPO2-LGR5 signaling has tumour-suppressive activity in colorectal cancer. Nat Commun. 5:31492014.

27 

Yu SJ, Wu KJ, Wang YS, Song JS, Wu CH, Jan JJ, Bae E, Chen H, Shia KS and Wang Y: Protective effect of CXCR4 antagonist CX807 in a rat model of hemorrhagic stroke. Int J Mol Sci. 21:70852020.

28 

Reinhardt SW, Lin CJ, Novak E and Brown DL: Noninvasive cardiac testing vs clinical evaluation alone in acute chest pain: A secondary analysis of the ROMICAT-II randomized clinical trial. JAMA Intern Med. 178:212–219. 2018.

29 

Ibanez B, Aletras AH, Arai AE, Arheden H, Bax J, Berry C, Bucciarelli-Ducci C, Croisille P, Dall'Armellina E, Dharmakumar R, et al: Cardiac MRI endpoints in myocardial infarction experimental and clinical trials: JACC scientific expert panel. J Am Coll Cardiol. 74:238–256. 2019.

30 

Klaourakis K, Vieira JM and Riley PR: The evolving cardiac lymphatic vasculature in development, repair and regeneration. Nat Rev Cardiol. 18:368–379. 2021.

31 

Liu X, De la Cruz E, Gu X, Balint L, Oxendine-Burns M, Terrones T, Ma W, Kuo HH, Lantz C, Bansal T, et al: Lymphoangiocrine signals promote cardiac growth and repair. Nature. 588:705–711. 2020.

32 

Lohela M, Bry M, Tammela T and Alitalo K: VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol. 21:154–165. 2009.

33 

Hendel RC, Henry TD, Rocha-Singh K, Isner JM, Kereiakes DJ, Giordano FJ, Simons M and Bonow RO: Effect of intracoronary recombinant human vascular endothelial growth factor on myocardial perfusion: Evidence for a dose-dependent effect. Circulation. 101:118–121. 2000.

34 

Eken A, Yetkin MF, Vural A, Okus FZ, Erdem S, Azizoglu ZB, Haliloglu Y, Cakir M, Turkoglu EM, Kilic O, et al: Fingolimod alters tissue distribution and cytokine production of human and murine innate lymphoid cells. Front Immunol. 10:2172019.

35 

Li Q, Zhou C, Zhao K, Duan Y, Yue J, Liu X, Wu J and Deng S: Lymphatic endothelial sphingosine 1-phosphate receptor 1 enhances macrophage clearance via lymphatic system following myocardial infarction. Front Cardiovasc Med. 9:8721022022.

36 

B Gowda S, Gowda D, Kain V, Chiba H, Hui SP, Chalfant CE, Parcha V, Arora P and Halade GV: Sphingosine-1-phosphate interactions in the spleen and heart reflect extent of cardiac repair in mice and failing human hearts. Am J Physiol Heart Circ Physiol. 321:H599–H611. 2021.

37 

Du M, Shan J, Feng A, Schmull S, Gu J and Xue S: Oestrogen receptor β activation protects against myocardial infarction via Notch1 signalling. Cardiovasc Drugs Ther. 34:165–178. 2020.

38 

Mahmoodzadeh S, Leber J, Zhang X, Jaisser F, Messaoudi S, Morano I, Furth PA, Dworatzek E and Regitz-Zagrosek V: Cardiomyocyte-specific estrogen receptor alpha increases angiogenesis, lymphangiogenesis and reduces fibrosis in the female mouse heart post-myocardial infarction. J Cell Sci Ther. 5:1532014.

39 

Jeya PJ, Weigel C, Müller T, Heller R, Spiegel S and Gräler MH: Inflammatory conditions disrupt constitutive endothelial cell barrier stabilization by alleviating autonomous secretion of sphingosine 1-phosphate. Cells. 9:9282020.

40 

Nagahashi M, Abe M, Sakimura K, Takabe K and Wakai T: The role of sphingosine-1-phosphate in inflammation and cancer progression. Cancer Sci. 109:3671–3678. 2018.

41 

Tatin F, Renaud-Gabardos E, Godet AC, Hantelys F, Pujol F, Morfoisse F, Calise D, Viars F, Valet P, Masri B, et al: Apelin modulates pathological remodeling of lymphatic endothelium after myocardial infarction. Jci Insight. 2:e938872017.

42 

Poh KK, Lee PSS, Djohan AH, Galupo MJ, Songco GG, Yeo TC, Tan HC, Richards AM and Ye L: Transplantation of endothelial progenitor cells in obese diabetic rats following myocardial infarction: Role of thymosin beta-4. Cells. 9:9492020.

43 

Wang YL, Yu SN, Shen HR, Wang HJ, Wu XP, Wang QL, Zhou B and Tan YZ: Thymosin β4 released from functionalized self-assembling peptide activates epicardium and enhances repair of infarcted myocardium. Theranostics. 11:4262–4280. 2021.

44 

Trincot CE, Xu W, Zhang H, Kulikauskas MR, Caranasos TG, Jensen BC, Sabine A, Petrova TV and Caron KM: Adrenomedullin induces cardiac lymphangiogenesis after myocardial infarction and regulates cardiac edema via connexin 43. Circ Res. 124:101–113. 2019.

45 

Lin QY, Zhang YL, Bai J, Liu JQ and Li HH: VEGF-C/VEGFR-3 axis protects against pressure-overload induced cardiac dysfunction through regulation of lymphangiogenesis. Clin Transl Med. 11:e3742021.

46 

Glinton KE, Ma W, Lantz C, Grigoryeva LS, DeBerge M, Liu X, Febbraio M, Kahn M, Oliver G and Thorp EB: Macrophage-produced VEGFC is induced by efferocytosis to ameliorate cardiac injury and inflammation. J Clin Invest. 132:e1406852022.

47 

McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Bohm M, Burri H, Butler J, Čelutkienė J, Chioncel O, et al: 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J. 42:3599–3726. 2021.

48 

Schwinger RHG: Pathophysiology of heart failure. Cardiovasc Diagn Ther. 11:263–276. 2021.

49 

Weckbach LT, Uhl A, Boehm F, Seitelberger V, Huber BC, Kania G, Brunner S and Grabmaier U: Blocking LFA-1 aggravates cardiac inflammation in experimental autoimmune myocarditis. Cells. 8:12672019.

50 

Tkacz K, Rolski F, Czepiel M, Działo E, Siedlar M, Eriksson U, Kania G and Błyszczuk P: Haploinsufficient Rock1+/− and Rock2+/− mice are not protected from cardiac inflammation and postinflammatory fibrosis in experimental autoimmune myocarditis. Cells. 9:7002020.

51 

Zheng H, Liu X, Katsurada K and Patel KP: Renal denervation improves sodium excretion in rats with chronic heart failure: Effects on expression of renal ENaC and AQP2. Am J Physiol Heart Circ Physiol. 317:H958–H968. 2019.

52 

Maier A, Braig M, Jakob K, Bienert T, Schäper M, Merkle A, Wadle C, Menza M, Neudorfer I, Bojti I, et al: Molecular magnetic resonance imaging of activated platelets allows noninvasive detection of early myocarditis in mice. Sci Rep. 10:132112020.

53 

Huang B, Miao H, Yuan Y, Qiu F, Liu X, Liu Z, Zhang H, Zhao Q, Wang M, Dong H and Zhang Z: PEDF decreases cardiomyocyte edema during oxygen-glucose deprivation and recovery via inhibiting lactate accumulation and expression of AQP1. Int J Mol Med. 43:1979–1990. 2019.

54 

Card CM, Yu SS and Swartz MA: Emerging roles of lymphatic endothelium in regulating adaptive immunity. J Clin Invest. 124:943–952. 2014.

55 

Kajiya K and Detmar M: An important role of lymphatic vessels in the control of uvb-induced edema formation and inflammation. J Invest Dermatol. 126:919–921. 2006.

56 

Li N, Zhu L, Sun L and Shao G: The effects of novel coronavirus (SARS-CoV-2) infection on cardiovascular diseases and cardiopulmonary injuries. Stem Cell Res. 51:1021682021.

57 

Hata AN and Breyer RM: Pharmacology and signaling of prostaglandin receptors: Multiple roles in inflammation and immune modulation. Pharmacol Ther. 103:147–166. 2004.

58 

Al-Kofahi M, Omura S, Tsunoda I, Sato F, Becker F, Gavins FNE, Woolard MD, Pattillo C, Zawieja D, Muthuchamy M, et al: IL-1β reduces cardiac lymphatic muscle contraction via COX-2 and PGE2 induction: Potential role in myocarditis. Biomed Pharmacother. 107:1591–1600. 2018.

59 

Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R and Eguchi S: Angiotensin II signal transduction: An update on mechanisms of physiology and pathophysiology. Physiol Rev. 98:1627–1738. 2018.

60 

Doan TN, Bernard FC, McKinney JM, Dixon JB and Willett NJ: Endothelin-1 inhibits size dependent lymphatic clearance of PEG-based conjugates after intra-articular injection into the rat knee. Acta Biomater. 93:270–281. 2019.

61 

Bai J, Yin L, Yu WJ, Zhang YL, Lin QY and Li HH: Angiotensin II induces cardiac edema and hypertrophic remodeling through lymphatic-dependent mechanisms. Oxid Med Cell Longev. 2022:50440462022.

62 

Qu X, Song X, Yuan W, Shu Y, Wang Y, Zhao X, Gao M, Lu R, Luo S, Zhao W, et al: Expression signature of lncRNAs and their potential roles in cardiac fibrosis of post-infarct mice. Biosci Rep. 36:e003372016.

63 

Iwamiya T, Segard BD, Matsuoka Y and Imamura T: Human cardiac fibroblasts expressing VCAM1 improve heart function in postinfarct heart failure rat models by stimulating lymphangiogenesis. PLoS One. 15:e02378102020.

64 

Simko F, Pechanova O, Repova K, Aziriova S, Krajcirovicova K, Celec P, Tothova L, Vrankova S, Balazova L, Zorad S and Adamcova M: Lactacystin-induced model of hypertension in rats: Effects of melatonin and captopril. Int J Mol Sci. 18:16122017.

65 

Pan L, Sun X, Che H and Li M: CTRP9 mitigates vascular endothelial cell injury in patients with hypertensive heart disease by inhibiting PI3K/Akt/mTOR axis. Am J Transl Res. 14:6596–6603. 2022.

66 

Sterns RH: Disorders of plasma sodium-causes, consequences, and correction. N Engl J Med. 372:55–65. 2015.

67 

Balasubbramanian D, Baranwal G, Clark MCC, Goodlett BL, Mitchell BM and Rutkowski JM: Kidney-specific lymphangiogenesis increases sodium excretion and lowers blood pressure in mice. J Hypertens. 38:874–885. 2020.

68 

Wiig H, Schröder A, Neuhofer W, Jantsch J, Kopp C, Karlsen TV, Boschmann M, Goss J, Bry M, Rakova N, et al: Immune cells control skin lymphatic electrolyte homeostasis and blood pressure. J Clin Invest. 123:2803–2815. 2013.

69 

Machnik A, Dahlmann A, Kopp C, Goss J, Wagner H, van Rooijen N, Eckardt KU, Müller DN, Park JK, Luft FC, et al: Mononuclear phagocyte system depletion blocks interstitial tonicity-responsive enhancer binding protein/vascular endothelial growth factor C expression and induces salt-sensitive hypertension in rats. Hypertension. 55:755–761. 2010.

70 

Yang GH, Zhou X, Ji WJ, Zeng S, Dong Y, Tian L, Bi Y, Guo ZZ, Gao F, Chen H, et al: Overexpression of VEGF-C attenuates chronic high salt intake-induced left ventricular maladaptive remodeling in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol. 306:H598–H609. 2014.

71 

Ye BJ, Lee HH, Yoo EJ, Lee CY, Lee JH, Kang HJ, Jeong GW, Park H, Lee-Kwon W, Choi SY and Kwon HM: TonEBP in dendritic cells mediates pro-inflammatory maturation and Th1/Th17 responses. Cell Death Dis. 11:4212020.

72 

Yang GH, Zhou X, Ji WJ, Liu JX, Sun J, Dong Y, Jiang TM and Li YM: VEGF-C-mediated cardiac lymphangiogenesis in high salt intake accelerated progression of left ventricular remodeling in spontaneously hypertensive rats. Clin Exp Hypertens. 39:740–747. 2017.

73 

Zhang C, Li N, Suo M, Zhang C, Liu J, Liu L, Qi Y, Zheng X, Xie L, Hu Y and Bu P: Sirtuin 3 deficiency aggravates angiotensin II-induced hypertensive cardiac injury by the impairment of lymphangiogenesis. J Cell Mol Med. 25:7760–7771. 2021.

74 

Hosios AM and Vander Heiden MG: Endothelial cells Get β-ox-ed in to support lymphangiogenesis. Dev Cell. 40:118–119. 2017.

75 

Li X, Qiu J, Pan M, Zheng D, Su Y, Wei M, Kong X, Sun W and Zhu J: Correlation between congenital heart disease complicated with pulmonary artery hypertension and circulating endothelial cells as well as endothelin-1. Int J Clin Exp Pathol. 8:10743–10751. 2015.

76 

Kelly B, Mohanakumar S and Hjortdal VE: Diagnosis and management of lymphatic disorders in congenital heart disease. Curr Cardiol Rep. 22:1642020.

77 

Li D, Sheppard SE, March ME, Battig MR, Surrey LF, Srinivasan AS, Matsuoka LS, Tian L, Wang F, Seiler C, et al: Genomic profiling informs diagnoses and treatment in vascular anomalies. Nat Med. 29:1530–1539. 2023.

78 

Boehme JT, Morris CJ, Chiacchia SR, Gong W, Wu KY, Kameny RJ, Raff GW, Fineman JR, Maltepe E and Datar SA: HIF-1α promotes cellular growth in lymphatic endothelial cells exposed to chronically elevated pulmonary lymph flow. Sci Rep. 11:14682021.

79 

Scallan JP, Hill MA and Davis MJ: Lymphatic vascular integrity is disrupted in type 2 diabetes due to impaired nitric oxide signalling. Cardiovasc Res. 107:89–97. 2015.

80 

Hwang J, Kleinhenz DJ, Lassegue B, Griendling KK, Dikalov S and Hart CM: Peroxisome proliferator-activated receptor-gamma ligands regulate endothelial membrane superoxide production. Am J Physiol Cell Physiol. 288:C899–C905. 2005.

81 

Choi D, Park E, Jung E, Seong YJ, Hong M, Lee S, Burford J, Gyarmati G, Peti-Peterdi J, Srikanth S, et al: ORAI1 activates proliferation of lymphatic endothelial cells in response to laminar flow through Krüppel-like factors 2 and 4. Circ Res. 120:1426–1439. 2017.

82 

Morris CJ, Kameny RJ, Boehme J, Gong W, He Y, Zhu T, Maltepe E, Raff GW, Fineman JR and Datar SA: KLF2-mediated disruption of PPAR-γ signaling in lymphatic endothelial cells exposed to chronically increased pulmonary lymph flow. Am J Physiol Heart Circ Physiol. 315:H173–H181. 2018.

83 

Yu J, Mao L, Guan L, Zhang Y and Zhao J: Ginsenoside Rg1 enhances lymphatic transport of intrapulmonary silica via VEGF-C/VEGFR-3 signaling in silicotic rats. Biochem Biophys Res Commun. 472:182–188. 2016.

84 

Allard B, Cousineau I, Allard D, Buisseret L, Pommey S, Chrobak P and Stagg J: Adenosine A2a receptor promotes lymphangiogenesis and lymph node metastasis. Oncoimmunology. 8:16014812019.

85 

Han H, Ma Y, Wang X, Yun R, Lu S, Xia M, Wang Y, Shi Q, Zhai W, Liang Q and Xu H: Fang-Ji-Huang-Qi-Tang attenuates degeneration of early-stage KOA mice related to promoting joint lymphatic drainage function. Evid Based Complement Alternat Med. 2020:34716812020.

86 

Chen Y, Li J, Li Q, Wang T, Xing L, Xu H, Wang Y, Shi Q, Zhou Q and Liang Q: Du-Huo-Ji-Sheng-Tang attenuates inflammation of TNF-Tg mice related to promoting lymphatic drainage function. Evid Based Complement Alternat Med. 2016:70676912016.

87 

Hou T, Liu Y, Wang X, Jiao D, Xu H, Shi Q, Wang Y, Li W, Wu T and Liang Q: Ginsenoside Rg1 promotes lymphatic drainage and improves chronic inflammatory arthritis. J Musculoskelet Neuronal Interact. 20:526–534. 2020.

88 

Ishii M, Miyata H, Ikeda N, Tagawa T and Nishimura M: Piper retrofractum extract and its component piperine promote lymphangiogenesis via an AKT- and ERK-dependent mechanism. J Food Biochem. 46:e142332022.

89 

Li J, Chen Y, Zhang L, Xing L, Xu H, Wang Y, Shi Q and Liang Q: Total saponins of panaxnotoginseng promotes lymphangiogenesis by activation vegf-c expression of lymphatic endothelial cells. J Ethnopharmacol. 193:293–302. 2016.

90 

Peng L, Dong Y, Fan H, Cao M, Wu Q and Wang Y, Zhou C, Li S, Zhao C and Wang Y: Traditional Chinese medicine regulating lymphangiogenesis: A literature review. Front Pharmacol. 11:12592020.

91 

Benvenuti LA, da Silva AMG and Aiello VD: Quantification of lymphatic vessels in dilated and chronic chagasic cardiomyopathy. Arq Bras Cardiol. 94:564–567. 2010.In Portuguese.

92 

Peng L, Ma M, Dong Y, Wu Q, An S, Cao M, Wang Y, Zhou C, Zhou M, Wang X, et al: Kuoxin Decoction promotes lymphangiogenesis in zebrafish and in vitro based on network analysis. Front Pharmacol. 13:9151612022.

93 

Shi J, Liang Q, Zuscik M, Shen J, Chen D, Xu H, Wang YJ, Chen Y, Wood RW, Li J, et al: Distribution and alteration of lymphatic vessels in knee joints of normal and osteoarthritic mice. Arthritis Rheumatol. 66:657–666. 2014.

94 

Lou B, Wu W, Zeng L, Zhou W, Zhang X, Zhou X, Liu Z, Liu K, Gu X, Chen X, et al: Alleviating experimental allergic eye disease by inhibiting pro-lymphangiogenic VEGFR3 signal. Ocul Surf. 26:1–12. 2022.

95 

Ahn JH, Cho H, Kim JH, Kim SH, Ham JS, Park I, Suh SH, Hong SP, Song JH, Hong YK, et al: Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature. 572:62–66. 2019.

96 

Da Mesquita S, Papadopoulos Z, Dykstra T, Brase L, Farias FG, Wall M, Jiang H, Kodira CD, de Lima KA, Herz J, et al: Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature. 593:255–260. 2021.

97 

Nedergaard M and Goldman SA: Glymphatic failure as a final common pathway to dementia. Science. 370:50–56. 2020.

98 

Li X, Qi L, Yang D, Hao S, Zhang F, Zhu X, Sun Y, Chen C, Ye J, Yang J, et al: Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat Neurosci. 25:577–587. 2022.

99 

Zhou M and Wang Y, Zhou C, Peng L, Liang Q and Wang Y: A research progress of Traditional Chinese medicine acting on the lymphangiogenesis and lymphatic drainage function. Mod Tradit Chin Med Materia Medica-World Sci Tech. 24:802–809. 2022.In Chinese.

100 

Qi SM, Zhang JT, Zhu HY, Wang Z and Li W: Review on potential effects of traditional Chinese medicine on glaucoma. J Ethnopharmacol. 304:1160632023.

101 

Zhang J, Li Q and Zhou YY: Research progress on mechanism of traditional Chinese medicine polysaccharides in preventing and treating Alzheimer's disease. Chin Tradit Herb Drugs. 53:7553–7565. 2022.In Chinese.

102 

Zhang QM, Wang YG, Zhang JX, Han XY and Tian X: Functional properties and biological basis of essence, qi, blood and body fluid. Glob Tradit Chin Med. 14:841–847. 2021.

103 

Cui JK, Chen X and Jiang Q: Discussion on the treatment of Sjogren's syndrome from the metabolism of body fluid. J Basic Chin Med. 25:1662–1663. 2019.In Chinese.

104 

Jiang L, Zhao JX, Zhang YF, Meng FZ, Jian J and Liu JT: Discussion on the treatment of diabetic kidney disease edema in traditional Chinese medicine based on the theory of qi, blood and water. China J Tradit Chin Med Pharm. 36:887–890. 2021.In Chinese.

105 

Fang Y, Huang K and Li X: Preliminary study on the blood circulation characteristics of phlegm syndrome. Hubei J Tradit Chin Med. 1:33–34. 1992.In Chinese.

106 

Wei D, Liu M and Shi X: A study on the distribution of traditional Chinese medicine syndromes of elevated blood glucose in the acute stage of stroke. J Emerg Tradit Chin Med. 10:91–92. 2001.In Chinese.

107 

Wang J, Yan C, Deng Z and Pan Y: Exploring the mechanism of phlegm syndrome from the abnormal metabolism of adhesion molecules. Chin J Integr Tradit West Med. 20:296–297. 2000.In Chinese.

108 

Yu S, Liu C, Zhou L, Zhong Y, Xiong N, Xu J, Liu C, Cheng S and Wang P: Wendantang treats inflammation in obesity (Syndrome of Phlegm-dampness) by regulatingPI3K/Akt/mTOR pathway-mediated adipocyte autophagy. Chin J Exp Tradit Med Formulae. 29:1–10. 2023.In Chinese.

109 

Yi GH: Lymphatic vessels: A potential path to intervene the development of atherosclerosis. Chin J Arterioscler. 26:973–979. 2018.In Chinese.

110 

Mikami T, Koyama A, Hashimoto K, Maegawa J, Yabuki Y, Kagimoto S, Kitayama S, Kaneta T, Yasumura K, Matsubara S and Iwai T: Pathological changes in the lymphatic system of patients with secondary upper limb lymphoedema. Sci Rep. 9:84992019.

111 

Mukherjee A, Hooks J, Nepiyushchikh Z and Dixon JB: Entrainment of lymphatic contraction to oscillatory flow. Sci Rep. 9:58402019.

112 

Huang SW, Yang HY, Huang WJ, Chen WC, Yu MC, Wang SW, Hsu YF and Hsu MJ: WMJ-S-001, a novel aliphatic hydroxamate-based compound, suppresses lymphangiogenesis through p38mapk-p53-survivin signaling cascade. Front Oncol. 9:11882019.

113 

Shinriki S, Jono H, Ueda M, Ota K, Ota T, Sueyoshi T, Oike Y, Ibusuki M, Hiraki A, Nakayama H, et al: Interleukin-6 signalling regulates vascular endothelial growth factor-C synthesis and lymphangiogenesis in human oral squamous cell carcinoma. J Pathol. 225:142–150. 2011.

114 

Zhang N, Hu H, Fu Y, He F, Wang L, Zhuang S and Ding M: The overexpression of PDGF-BB and its receptor is correlated with lymphatic metastasis in patients with non-small cell lung cancer. Int J Clin Exp Pathol. 11:6010–6017. 2018.

115 

Caunt M, Mak J, Liang WC, Stawicki S, Pan Q, Tong RK, Kowalski J, Ho C, Reslan HB, Ross J, et al: Blocking neuropilin-2 function inhibits tumor cell metastasis. Cancer Cell. 13:331–342. 2008.

116 

Song J, Chen W, Cui X, Huang Z, Wen D, Yang Y, Yu W, Cui L and Liu CY: CCBE1 promotes tumor lymphangiogenesis and is negatively regulated by TGFβ signaling in colorectal cancer. Theranostics. 10:2327–2341. 2020.

117 

Haemmerle M, Keller T, Egger G, Schachner H, Steiner CW, Stokic D, Neumayer C, Brown MK, Kerjaschki D and Hantusch B: Enhanced lymph vessel density, remodeling, and inflammation are reflected by gene expression signatures in dermal lymphatic endothelial cells in type 2 diabetes. Diabetes. 62:2509–2529. 2013.

118 

Morooka N, Futaki S, Sato-Nishiuchi R, Nishino M, Totani Y, Shimono C, Nakano I, Nakajima H, Mochizuki N and Sekiguchi K: Polydom is an extracellular matrix protein involved in lymphatic vessel remodeling. Circ Res. 120:1276–1288. 2017.

119 

Choi D, Park E, Yu RP, Cooper MN, Cho IT, Choi J, Yu J, Zhao L, Yum JI, Yu JS, et al: Piezo1-regulated mechanotransduction controls flow-activated lymphatic expansion. Circ Res. 131:e2–e21. 2022.

120 

Klotz L, Norman S, Vieira JM, Masters M, Rohling M, Dubé KN, Bollini S, Matsuzaki F, Carr CA and Riley PR: Cardiac lymphatics are heterogeneous in origin and respond to injury. Nature. 522:62–67. 2015.

121 

Lim HY, Thiam CH, Yeo KP, Bisoendial R, Hii CS, McGrath KC, Tan KW, Heather A, Alexander JS and Angeli V: Lymphatic vessels are essential for the removal of cholesterol from peripheral tissues by SR-BI-mediated transport of HDL. Cell Metab. 17:671–684. 2013.

122 

Dashkevich A, Raissadati A, Syrjälä SO, Zarkada G, Keränen MA, Tuuminen R, Krebs R, Anisimov A, Jeltsch M, Leppanen VM, et al: Ischemia-reperfusion injury enhances lymphatic endothelial VEGFR3 and rejection in cardiac allografts. Am J Transplant. 16:1160–1172. 2016.

123 

Losordo DW, Vale PR, Hendel RC, Milliken CE, Fortuin FD, Cummings N, Schatz RA, Asahara T, Isner JM and Kuntz RE: Phase 1/2 placebo-controlled, double-blind, dose-escalating trial of myocardial vascular endothelial growth factor 2 gene transfer by catheter delivery in patients with chronic myocardial ischemia. Circulation. 105:2012–2018. 2002.

124 

Hartikainen J, Hassinen I, Hedman A, Kivelä A, Saraste A, Knuuti J, Husso M, Mussalo H, Hedman M, Rissanen TT, et al: Adenoviral intramyocardial VEGF-DΔNΔC gene transfer increases myocardial perfusion reserve in refractory angina patients: A phase I/IIa study with 1-year follow-up. Eur Heart J. 38:2547–2555. 2017.

125 

O'Dwyer J, Murphy R, Dolan EB, Kovarova L, Pravda M, Velebny V, Heise A, Duffy GP and Cryan SA: Development of a nanomedicine-loaded hydrogel for sustained delivery of an angiogenic growth factor to the ischaemic myocardium. Drug Deliv Transl Res. 10:440–454. 2020.

126 

O'Dwyer J, Murphy R, González-Vázquez A, Kovarova L, Pravda M, Velebny V, Heise A, Duffy GP and Cryan SA: Translational studies on the potential of a VEGF nanoparticle-loaded hyaluronic acid hydrogel. Pharmaceutics. 13:7792021.

127 

Zheng W, Aspelund A and Alitalo K: Lymphangiogenic factors, mechanisms, and applications. J Clin Invest. 124:878–887. 2014.

128 

Hu D, Li L, Li S, Wu M, Ge N, Cui Y, Lian Z, Song J and Chen H: Lymphatic system identification, pathophysiology and therapy in the cardiovascular diseases. J Mol Cell Cardiol. 133:99–111. 2019.

129 

Hwang SD, Song JH, Kim Y, Lim JH, Kim MY, Kim EN, Hong YA, Chung S, Choi BS, Kim YS and Park CW: Inhibition of lymphatic proliferation by the selective VEGFR-3 inhibitor SAR131675 ameliorates diabetic nephropathy in db/db mice. Cell Death Dis. 10:2192019.

130 

Rahbar E, Weimer J, Gibbs H, Yeh AT, Bertram CD, Davis MJ, Hill MA, Zawieja DC and Moore JE Jr: Passive pressure-diameter relationship and structural composition of rat mesenteric lymphangions. Lymphat Res Biol. 10:152–163. 2012.

131 

Fang Y, Wang XY, You JF, Liang QQ and Zheng WC: Jia-Wei-Niu-Bang-Zi-Tang promotes lymphangiogenesis in TNF-α transgenic mice and alleviates rheumatoid arthritis. Chin Pharmacol Bulletin. 37:1469–1474. 2021.In Chinese.

132 

Feng X, Sun H, Gao F and Ma W: Observation of curative effect of the treatment of lymphedema of upper limb after breast cancer operation with fumigation and washing of traditional Chinese medicine combined with acupoint application of Jiawei Jinhuang ointment. J Mod Oncol. 31:1252–1256. 2023.In Chinese.

133 

Lohrke J, Frenzel T, Endrikat J, Alves FC, Grist TM, Law M, Lee JM, Leiner T, Li KC, Nikolaou K, et al: 25 years of contrast-enhanced MRI: Developments, current challenges and future perspectives. Adv Ther. 33:1–28. 2016.

134 

Quintanilla M, Montero-Montero L, Renart J and Martin-Villar E: Podoplanin in inflammation and cancer. Int J Mol Sci. 20:7072019.

135 

Aldea R, Weller RO, Wilcock DM, Carare RO and Richardson G: Cerebrovascular smooth muscle cells as the drivers of intramural periarterial drainage of the brain. Front Aging Neurosci. 11:12019.

136 

Brakenhielm E and Alitalo K: Cardiac lymphatics in health and disease. Nat Rev Cardiol. 16:56–68. 2019.

137 

Gordon-Walker TT, Bove K and Veldtman G: Fontan-associated liver disease: A review. J Cardiol. 74:223–232. 2019.

138 

Savla JJ, Itkin M, Rossano JW and Dori Y: Post-operative chylothorax in patients with congenital heart disease. J Am Coll Cardiol. 69:2410–2422. 2017.

139 

Goumans MJ, Zwijsen A, Ten Dijke P and Bailly S: Bone morphogenetic proteins in vascular homeostasis and disease. Cold Spring Harb Perspect Biol. 10:a0319892018.

140 

Nash LA, Burns JK, Chardon JW, Kothary R and Parks RJ: Spinal muscular atrophy: More than a disease of motor neurons? Curr Mol Med. 16:779–792. 2016.

141 

Melenotte C, Protopopescu C, Million M, Edouard S, Carrieri MP, Eldin C, Angelakis E, Djossou F, Bardin N, Fournier PE, et al: Clinical features and complications of coxiella burnetii infections from the french national reference center for Q fever. JAMA Netw Open. 1:e1815802018.

142 

Vieira JM, Norman S, Villa Del Campo C, Cahill TJ, Barnette DN, Gunadasa-Rohling M, Johnson LA, Greaves DR, Carr CA, Jackson DG and Riley PR: The cardiac lymphatic system stimulates resolution of inflammation following myocardial infarction. J Clin Invest. 128:3402–3412. 2018.

143 

Chavhan GB, Amaral JG, Temple M and Itkin M: MR lymphangiography in children: Technique and potential applications. Radiographics. 37:1775–1790. 2017.

Related Articles

Journal Cover

April-2024
Volume 53 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bai L, Wang Y, Du S, Si Y, Chen L, Li L and Li Y: Lymphangiogenesis: A new strategy for heart disease treatment (Review). Int J Mol Med 53: 35, 2024
APA
Bai, L., Wang, Y., Du, S., Si, Y., Chen, L., Li, L., & Li, Y. (2024). Lymphangiogenesis: A new strategy for heart disease treatment (Review). International Journal of Molecular Medicine, 53, 35. https://doi.org/10.3892/ijmm.2024.5359
MLA
Bai, L., Wang, Y., Du, S., Si, Y., Chen, L., Li, L., Li, Y."Lymphangiogenesis: A new strategy for heart disease treatment (Review)". International Journal of Molecular Medicine 53.4 (2024): 35.
Chicago
Bai, L., Wang, Y., Du, S., Si, Y., Chen, L., Li, L., Li, Y."Lymphangiogenesis: A new strategy for heart disease treatment (Review)". International Journal of Molecular Medicine 53, no. 4 (2024): 35. https://doi.org/10.3892/ijmm.2024.5359