Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 17.
Published in final edited form as: Cell Mol Gastroenterol Hepatol. 2016 Nov;2(6):733–749. doi: 10.1016/j.jcmgh.2016.09.002

The Hepatic Lymphatic Vascular System: Structure, Function, Markers, and Lymphangiogenesis

Masatake Tanaka 1, Yasuko Iwakiri 1
PMCID: PMC5240041  NIHMSID: NIHMS816847  PMID: 28105461

Abstract

The lymphatic vascular system has been little explored in the liver despite its indispensable properties including maintenance of tissue fluid homeostasis. The discovery of specific markers for lymphatic endothelial cells has advanced various experimental methods including imaging, cell isolation and transgenic animal models, and has resulted in the rapid progress of the lymphatic vascular research in other tissues and organs during the last decade. These studies have also brought concrete evidence that lymphatic vessel dysfunction plays an important role in the pathogeneses of various diseases. This review will provide current knowledge of structure, function and markers of the hepatic lymphatic vascular system as well as factors associated with hepatic lymphangiogenesis in reference to those of other tissues.

Keywords: VEGF, inflammation, cirrhosis, portal hypertension

Introduction

The lymphatic vascular system constitutes the major circulatory system together with the blood vascular system and is engaged in indispensable physiological activities. The lymphatic vascular system maintains tissue fluid homeostasis by collecting excess tissue fluid and returning it to the venous circulation. It also plays an essential role in absorption and transport of dietary fat. Further, lymphatic vessels are crucial for immune surveillance and acquired immunity by serving as the main conduits of antigens and antigen-presenting cells from peripheries to lymph nodes.14

The lymphatic vascular research had been delayed partly because of lack of knowledge about markers and signaling pathways specific to the lymphatic vasculature. In 1995 to 1997, however, the important findings were brought, which showed that vascular endothelial growth factor receptor (VEGFR)-3 is expressed in the lymphatic endothelium and that its ligand vascular endothelial growth factor (VEGF)-C promotes lymphangiogenesis.5, 6 These findings on signaling pathways specific to the lymphatic vasculature and the subsequent discoveries of other specific markers for lymphatic endothelial cells (LyECs), such as lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1)7, prospero homeobox protein 1 (Prox1)8 and podoplanin9, have advanced the lymphatic vascular research. In consequence, it has been recognized that lymphatic vessel dysfunction plays an important role in the pathogeneses of various diseases.

In the liver, however, the study of the lymphatic vascular system has been little explored. This review will provide an overview of structure, function and markers of the lymphatic vascular system as well as factors associated with lymphangiogenesis in the liver along with knowledge gained in other tissues.

1. Structure of the hepatic lymphatic vascular system

This section will address structure of the lymphatic vascular system in general, followed by that of the liver. Detailed anatomical structure of the lymphatic and hepatic lymphatic vascular systems is available in other review articles.3, 1012

1) Anatomy of the Lymphatic Vascular System

Lymphatic capillaries

Lymphatic fluid originates from plasma components leaked from capillary vessels into the interstitium. Lymphatic fluid then enters into lymphatic capillaries, which are blind-ended, thin-walled vessels consisting of a single layer of LyECs. Lymphatic capillaries are not covered by pericytes or smooth muscle cells and lack basement membranes.13, 14 They are highly permeable with discontinuous “button-like” junctions, through which interstitial fluid, macromolecules and immune cells can be transported.15 In addition, LyECs have anchoring filaments that are mainly composed of emilin-1 and fibrillin and bind LyECs to the surrounding extracellular matrix.14, 16, 17 These filaments keep lymphatic vessel lumens open to facilitate tissue fluid intake in conditions of tissue swelling.

Collecting vessels

Lymphatic capillaries coalesce into collecting vessels, which are covered with smooth muscle cells and basement membranes.14 Collecting vessels are much less permeable than lymphatic capillaries because LyECs form “zipper-like” junctions. Collecting vessels can be divided into smaller functional units called lymphangions that have unidirectional bicuspid valves at each end.18 The phasic contraction of smooth muscle cells covering lymphangions has collecting vessels act as a pump to drive lymphatic flow. Stimulation of smooth muscle cells causes depolarization of cell membrane and opens Ca2+ channels, resulting in Ca2+ influx and smooth muscle cell contraction. Smooth muscle cells also have stretch-activated Ca2+ channels, facilitating phasic contraction.19, 20 On the other hand, LyECs produce nitric oxide (NO), a vasodilator, in response to shear stress by fluid flow, which counteracts the Ca2+-dependent contraction.21, 22 Therefore, spatiotemporal alterations of Ca2+ and NO levels are thought to modulate the phasic contraction of lymphangions.23

Lymph nodes and lymph trunks

Collecting vessels are connected to one or more lymph nodes. Antigen presenting cells including dendritic cells and macrophages in lymphatic fluid interact with lymphocytes in lymph nodes, facilitating adaptive immune responses. Then, lymphatic fluid flows to secondary central lymph nodes, tertiary central lymph nodes and lymph trunks finally.24 Lymphatic fluid from the left side of the body, abdomen and lower limb is ultimately drained into the thoracic duct, the largest lymphatic vessel, which is connected to the left subclavian vein (Figure 1). Lymphatic fluid from the other parts of the body is drained into the right lymph trunk connected to the right subclavian vein.25 Lymphatic fluid entering into the subclavian veins returns to the systemic blood circulation.

Figure 1. The schematic diagram of the macro-anatomy of the hepatic lymphatic vascular system.

Figure 1

(➀): Lymphatic capillaries in the portal tract coalesce into collecting vessels, which drain to lymph nodes at the hepatic hilum and the lesser omentum. Efferent lymphatic vessels (LV) from these lymph nodes connect to celiac lymph nodes, which drain to the cisterna chyli, the enlarged origin of the thoracic duct. Lymphatic fluid through the thoracic duct drains to the left subclavicular vein and returns to the systemic blood circulation. (➁): Lymphatic vessels along the central vein (CV) converge into large lymphatic vessels along the hepatic vein (HV), which traverse along the inferior vena cava (IVC) through the diaphragm toward mediastinal lymph nodes. (➂): Lymphatic fluid running underneath the capsule of the liver-convex (3i) drains to mediastinal lymph nodes through the coronary ligament, while that of the liver-concave surface (3ii) drains to lymph nodes of the hepatic hilum and regional lymph nodes. HA, hepatic artery; PV, portal vein; BD, bile duct; LN, lymph node.

2) Anatomy of the Hepatic Lymphatic Vascular System

The schematic diagram of the hepatic lymphatic vascular system is shown in Figures 1 and 2. The liver has sinusoids instead of capillaries.26 Sinusoids are distinct from capillaries as they consist of one layer of liver sinusoidal endothelial cells (LSECs) and lack pericytes and basement membranes, similar to lymphatic capillaries. The origin of hepatic lymphatic fluid is thought to be plasma components filtered through fenestrae of LSECs into the space of Disse, the interstitial space between LSECs and hepatocytes.10, 11 Lymphatic fluid in the space of Disse mostly flows through the space of Mall, a space between the stroma of the portal tract and the outermost hepatocytes27, into the interstitium of the portal tract and then into lymphatic capillaries. Some portion of the lymphatic fluid in the space of Disse flows into the interstitium around the central vein, which is located in the center of the liver acinus and connected to the hepatic vein28, or underneath the hepatic capsule (Figure 2).

Figure 2. The schematic diagram of the micro-anatomy of the hepatic lymphatic vascular system.

Figure 2

Blood flow (red arrows) from the portal vein (PV) and the hepatic artery (HA) enters to the liver. Plasma components filtered through liver sinusoidal endothelial cells (LSECs) into the space of Disse, the interstitial space between LSECs and hepatocytes, are regarded as the origin of lymphatic fluid. Lymphatic fluid in the space of Disse mostly flows through the space of Mall, the space between the stroma of the portal tract and the outermost hepatocytes, into the interstitium of the portal tract and then into lymphatic capillaries (➀). Some portion of the lymphatic fluid in the space of Disse flows into the interstitium around the central vein (➁) or underneath the hepatic capsule (➂).

Lymphatic capillaries in the portal tract coalesce into collecting vessels and drain to lymph nodes at the hepatic hilum. Lymphatic vessels along the central vein converge into 5–6 large lymphatic vessels, which traverse along the inferior vena cava through the diaphragm toward posterior mediastinal lymph nodes. Lymphatic fluid running underneath the capsule of the liver-convex drains to mediastinal lymph nodes through the coronary ligament, while that of the liver-concave surface drains to lymph nodes of the hepatic hilum and regional lymph nodes (Figure 1).1012, 29 Based on their locations, lymphatic vessels along the portal tract and the central vein are called the deep lymphatic system and those along the hepatic capsule are called the superficial lymphatic system.1012, 29

2. Markers of lymphatic vessels

Lymphatic vessel markers generally refer to those of LyECs, reflecting their structural significance. Among those markers, LYVE-17, 30, 31, podoplanin9, 32, Prox18, 3335 and VEGFR-35 are most commonly used for microscopic imaging of lymphatic vessels.36 Finding more specific markers for the liver may be needed, because most well known LyEC markers, such as LYVE-1 and Prox1, are also expressed in liver sinusoidal endothelial cells (LSECs) and hepatocytes, respectively. Table 1 summarizes LyEC markers whose expressions were histologically examined in the liver. Some of them are also briefly discussed below.

Table 1.

Lymphatic markers

Marker Postnatal expression EXCEPT for lymphatic vessels Hepatic expression in pathological conditions Ref.
Liver Other organs/cells
LYVE-1 Sinusoidal endothelial cells A portion of macrophages, pulmonary capillaries, epididymal adipose tissue, mesentery, eye (cornea, sclera, choroid, iris, and retina), wounded skin, and malignant tumors (melanoma and insulinoma) In chronic hepatitis and liver cirrhosis in humans, LYVE-1(+) lymphatic vessels increase but LYVE-1(+) sinusoidal endothelial cells decrease. 38, 4043, 107, 161166
Prox1 Hepatocytes Adrenal medulla, megakaryocytes, and platelets Intra-hepatic CCC, ductular cells in cirrhotic livers, and HCC in humans. 8, 52, 59, 60
Podoplanin Cholangiocytes Inflammatory macrophages, mesothelial cells, cardiomyocytes, FRCs, follicular dendritic cells, TH17 cells, and osteoblasts Podoplanin(+) lymphatic vessels increase in decompensated cirrhosis in humans. Podoplanin(+) FRCs increase in the livers of primary biliary cirrhosis patients. EHE and angiomyolipoma in humans. 73, 74, 7781, 167, 168
VEGFR-3 Cholangiocytes A portion of macrophages, proliferating blood vessels, and fenestrated capillaries in endocrine glands, choroid plexus, kidney, and small intestine HBx Ag positive HCC and hepatic progenitor cells in primary biliary cirrhosis in humans. 82, 8587, 104, 169, 170
CCL21 Sinusoidal endothelial cells A portion of dendritic cells, HEVs of lymph nodes and Peyer’s patches, T-cell areas of spleen, lymph nodes, and Peyer’s patches Lymphoid tissue in primary biliary cirrhosis and primary sclerosing cholangitis in humans. 171173
MMR1 Sinusoidal endothelial cells and Kupffer cells A portion of macrophages, sinusoidal endothelial cells in bone marrow and spleen, perivascular microglia, and glomerular mesangial cells Unknown 174177
Desmoplakin Basolateral plasma membrane of hepatocytes and cholangiocytes Esophagus, intestine, colon, salivary gland, mammary gland, sweat gland, thymus, and endocervix Entire plasma membrane of HCC cells 176, 178181
Integrin α9 Hepatocytes Airway epithelial cells, keratinocytes, muscle cells (smooth/skeletal/cardiac), neutrophils, osteoclasts, and oocytes Unknown 176, 182

[Abbreviations] LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1; Prox1, prospero homeobox protein 1; CCC, cholangiocellular carcinoma; HCC, hepatocellular carcinoma; FRC, fibroblastic reticular cell; EHE, epithelioid hemangioendothelioma; VEGFR, vascular endothelial growth factor receptor; HBx Ag, Hepatitis B x antigen; CCL21, C-C motif chemokine ligand 21; HEV, high endothelial venules; MMR, macrophage mannose receptor 1

LYVE-1

LYVE-1 is a lymphatic vessel endothelial hyaluronan (HA) receptor, which belongs to the Link protein superfamily that contains a conserved HA-binding domain, known as the Link module.37 LYVE-1 is a homolog of the CD44 HA receptor.7 With a Link module and homology shared with the CD44 HA receptor, LYVE-1 may be involved in the transport of HA across the lymphatic endothelium. LYVE-1 is strongly expressed on the entire luminal and abluminal surfaces of LyECs, even on fine filopodia of growing vessels during lymphangiogenesis.

No definite alterations in lymphatic vessel structure and function were reported for LYVE-1−/− mice.38 However, diphtheria toxin-induced LYVE-1 depletion in mice caused acute loss of lymphatic lacteals in intestinal villi and lymphatic vessels in systemic lymph nodes. These changes resulted in the structural distortion of blood capillaries and whole architecture of the villi, leading to death due to sepsis within 60 hours after LYVE-1 depletion.39 These observations indicate that LYVE-1 plays an important role in the maintenance of the lymphatic vascular system, especially lacteals in intestinal villi and lymph nodes. Therefore, some compensatory mechanisms might work in the setting of congenital loss of LYVE-1.

In the liver, LYVE-1 is expressed not only in LyECs but also in LSECs in mice40 and humans.4043 However, LYVE-1 positivity in LSECs was reported to diminish in inflamed human livers, such as those of chronic hepatitis and cirrhosis.40, 42 Expression levels of LYVE-1 in human hepatocellular carcinoma (HCC) negatively correlated with the overall survival of patients.44

Prox1

Prox1, a homolog of the Drosophila melanogaster homeobox gene prospero, is a transcriptional factor and regulates the genes related to lymphatic endothelial cells, such as VEGFR-3 and podoplanin.45, 46 Prox1 is essential for the development of the lymphatic vascular system,8 while it also plays a role in the development of other tissues, including the lens34, 47, retina48, heart49, central nervous system50, pancreas51 and liver52, 53. Prox1 is positive in the nucleus in contrast to other lymphatic markers that are expressed in the cytoplasm and/or the plasma membrane.

Prox1−/− mice are devoid of the lymphatic vascular system and die at approximately E14.5 because Prox1 is essential for budding of lymphatic endothelial sacs.8 Prox1+/− mice die in a few days after their birth and present dysfunction of lymphatic vessels with chylous ascites.8, 47, 54 Several lines of Prox1-promoter directed reporter mice have recently been established as effective research tools (GFP55, mOrange56 and tdTomato5759).

In early endoderm, Prox1 expression is restricted to the primordia of the liver and the pancreas.51 Prox1 regulates hepatocyte migration during liver morphogenesis51 and is expressed in postnatal hepatocytes, but not in postnatal pancreas.52 In humans, cholangiocytes of normal livers were negative for Prox1, but intra-hepatic cholangiocarcinoma and ductular cells in fibrotic septa of cirrhotic livers and HCC were positive.60 In addition, expression levels of Prox1 in human HCC negatively correlated with the overall survival of patients.61 Prox1 acts with nuclear receptors, such as hepatocyte nuclear factor (HNF) 4α62, estrogen-related receptor (ERR) α63, 64, liver receptor homolog (LRH)-165 and retinoic acid-related orphan receptors (ROR) α/γ66, and regulates bile acid synthesis65 and circadian metabolism in the liver.64, 66

Podoplanin

Podoplanin is a type I transmembrane glycoprotein and is known to be essential for the development of the heart6770, lung71, spleen and lymph nodes72. Its expression is regulated by Prox1.45 Podoplanin is also a ligand of C-type lectin receptor CLEC-2, which is highly expressed in platelets and immune cells and promotes platelet aggregation and activation.73

Podoplanin−/− mice die at their birth due to respiratory failure. These mice present congenital lymphedema due to lymphatic vessel defect although blood vessel formation is normal.74 Podoplanin+/− mice are healthy and fertile only with partial incompleteness of the lymphatic vessel network.74 Recently, keratinocyte-specific podoplanin deficient mice75 and a tamoxifen-inducible podoplanin depletion mouse model (Pdpnf/f; CagCre)76 have been developed.

Histological analysis of normal mouse livers showed expression of podoplanin in cholangiocytes besides LyECs.77 Podoplanin-positive lymphatic vessels increased in human livers of decompensated cirrhosis.78 It was also reported that podoplanin-positive fibroblastic reticular cells increased in human livers of primary biliary cirrhosis.79 Podoplanin is a useful histological marker for diagnosing patients with vascular tumors with lymphatic differentiation, such as epithelioid hemangioendothelioma (EHE)80 and angiomyolipoma.81

VEGFR-3

VEGFR-3 is a membrane-anchored tyrosine kinase and the receptor of VEGF-C and VEGF-D. VEGFR-3 plays a crucial role in lymphangiogenesis. In early embryogenesis prior to LyEC differentiation, VEGFR-3 is expressed in most endothelial cells, but in the later stages of development, its expression becomes mostly restricted to the lymphatic endothelium.5

VEGFR-3−/− mice present lymphatic vessel defect and die at approximately E10.5.82 VEGFR-3+/− mice present leaky lymphatic vessels and transient chylous ascites.82, 83 A mouse line (Vegfr3EGFPLuc), in which a dual reporter for fluorescence and luminescence is expressed under VEGFR-3-promoter, was established recently and luminescence imaging of tumor-induced lymphangiogenesis became available.84

VEGFR-3 expression was seen in cholangiocytes of normal rat livers and increased in those of cholestatic rat livers induced by bile duct ligation.85 Hepatic progenitor cells were also found to express VEGFR-3 in patients with primary biliary cirrhosis.86 Hepatitis B x antigen (HBx Ag) is one of the antigens of hepatitis B virus (HBV) and promotes hepatocarcinogenesis by upregulating expression of genes associated with proliferation of hepatocytes. Upregulation of VEGFR-3 expression was observed in HBx Ag-positive human HCC, and the prognosis of patients with VEGFR-3 positive HCC was worse than that with VEGFR-3 negative HCC.87

3. Lymphangiogenesis

This section addresses the mechanism of lymphangiogenesis in the postnatal stage and factors that affect lymphangiogenesis, including inflammatory cells, in the lymphatic system in general. Then, implications of lymphangiogenesis in the pathophysiology of the liver are summarized.

1) Factors associated with lymphangiogenesis

In the postnatal stage, lymphatic vessels are mostly quiescent. Therefore, lymphangiogenesis generally occurs in pathological conditions such as tissue repair, inflammation and tumor-related conditions.88 Many cytokines/growth factors have been reported to promote lymphangiogenesis (lymphangiogenic) or inhibit lymphangiogenesis (anti-lymphangiogenic), which are summarized in Table 2. The extent and duration of lymphangiogenesis are determined by balances between lymphangiogenic factors and antilymphangiogenic factors.89, 90

Table 2.

Lymphangiogenic and Anti-lymphangiogenic Factors

Lymphangiogenic factors Experimental model Remarks Ref.
VEGF-A Mouse corneal lymphangiogenesis VEGF-A recruits macrophages, which promote lymphangiogenesis by secreting VEGF-C/VEGF-D. 105
Mouse subcutaneous immunization model VEGF-A expression is upregulated concomitantly with lymphangiogenesis in LNs of immunized mice. 119
Oxazolone sensitized delayed-type hypersensitivity in mouse ear Systemic blockade of VEGF-A attenuates lymphangiogenesis in draining LNs. 183
HSV-1 infection of cornea HSV-1 causes lymphangiogenesis by promoting infected cells to secrete VEGF-A. 184
VEGF-C, VEGF-D VEGF-C transgenic mouse VEGF-C promotes LyEC proliferation and LV enlargement in the skin. 6
Isolated LyEC VEGF-C stimulates survival, growth, and migration of LyEC. 93
FGF-2-induced corneal lymphangiogenesis VEGFR-3 blockade cancels lymphangiogenesis. 185
Chronic airway inflammation VEGFR-3 blockade cancels lymphangiogenesis. 186
LPS-induced peritonitis VEGF-C and VEGF-D promote lymphangiogenesis in diaphragm. 187
Ang 2 Mouse corneal lymphangiogenesis Ang 2 is upregulated in inflamed cornea and Ang2 blockade inhibits inflammatory lymphangiogenesis. 188
Mouse corneal lymphangiogenesis Ang 2 is expressed in lymphatic vessels and macrophages in inflamed cornea. Inflammatory lymphangiogenesis of cornea is suppressed in Ang2 knockout mice. Ang2 blockade inhibits LyEC proliferation and capillary tube formation. 189
HGF Canine primary LyEC, rat tail lymphedema HGF promotes proliferation and migration of LyEC. Weekly HGF gene transfer improves lymphedema in vivo. 190
LT CCL21 transgenic mouse, RAG knockout mouse defective in T and B cell LT overexpression by CCL21 transgene promotes lymphangiogenesis in thyroid. T cell depletion cancels this phenomenon. 191
LTα knockout mouse, LTα transgenic mouse LTα gene deletion decreases LV. Ectopic LTα expression causes lymphangiogenesis in tertiary lymphoid organs. 192
IL-1β Mouse corneal lymphangiogenesis IL-1β promotes lymphangiogenesis by upregulating expression of VEGF-A, VEGFC, and VEGF-D. 193
IL-7 Breast cancer cell lines, subcutaneous injection of MatrigelTM and/or IL-7 and/or breast cancer cell lines IL-7 promotes VEGF-D expression of cell lines in vitro and promotes lymphangiogenesis in vivo. 194
HECV cell line (originated from human umbilical cord), subcutaneous injection of MatrigelTM and/or IL-7 and/or HECV cell IL-7 promotes expression of Prox1, LYVE-1 and podoplanin and proliferation, migration and tubular formation of LyEC via upregulation of VEGF-D. 195
IL-8 Human primary LyEC, IL-8 transgenic mouse and Prox1-GFP mouse IL-8 promotes proliferation, migration and tube formation of LyEC. IL-8 overexpression promotes lymphangiogenesis in vivo. 196
IL-17 Cornea micro pocket assay, autoimmune ocular disease mouse IL-17 promotes proliferation of LyEC via upregulation of VEGF-D. Blockade of IL-17 decreases corneal lymphangiogenesis. 197
IL-20 Human telomerase-transfected dermal LyEC IL-20 promotes proliferation, migration and tubular formation of LyEC via PI3K and mTOR pathways. 198
Anti-lymphangiogenic factors Experimental model Remarks Ref.
TGF-β Human dermal lymphatic microvascular endothelial cells TGF-β inhibits LyEC proliferation, cord formation, migration, expression of lymphatic markers (LYVE-1, Prox1) and lymphangiogenesis by VEGF-A/C via TGF-β type I receptor. 199
Mouse tail skin excision and lymphatic vessel ligation TGF-β1 inhibition promotes lymphatic vessel regeneration. TGF-β1 inhibits LyEC proliferation and fibrosis. 200
Biopsy specimens from limbs of secondary lymphedema patients and mouse tail skin excision TGF-β1 positive cells increase threefold in human lymphedema specimens. TGFβ1 inhibition decreases fibrosis, increases lymphangiogenesis and lymphatic function. 201
BMP2 Zebrafish BMP2 transgenic model BMP2 inhibits LyEC differentiation from cardinal veins via inhibition of Prox1 expression. 202
IFN-α, IFN-γ LyEC isolated from pig thoracic duct IFN-α or IFN-γ decreases LyEC proliferation and migration. Treatment with both IFN-α and IFN-γ promotes LyEC apoptosis. 203
Cervical LNs of T-cell deprived mouse T cells inhibit lymphangiogenesis in LNs by secreting IFN-γ. 118
IL-4, IL-13 Mouse LyEC isolated from LNs, human dermal LyEC, mouse asthma model IL-4 and IL-13 inhibit expression of Prox1 and LYVE-1 and tube formation of LyEC. Blockade of IL-4 and/or IL-13 increases the density and function of lung LVs in asthma model. 204
IL-27 Human dermal lymphatic microvascular endothelial cells IL-27 inhibits LyEC proliferation and migration via STAT1/CXCL10, CXCL-11 axis. 205
Activin A Subcutaneous injection of melanoma cell line to mouse Activin A reduces lymphangiogenesis in melanoma model and inhibits sprouting of LyEC via phosphorylation of SMAD2. 206

[Abbreviations] VEGF, vascular endothelial growth factor; LN, lymph node; HSV-1, Herpes simplex virus 1; LyEC, lymphatic endothelial cell; LV, lymphatic vessel; FGF-2, fibroblast growth factors-2; VEGFR, vascular endothelial growth factor receptor; LPS, lipopolysaccharide; HGF, hepatocyte growth factor; LT, lymphotoxin; IL, interleukin; Prox1, prospero homeobox protein 1; LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; mTOR, mammalian target of rapamycin; TGF, tumor growth factor; IFN, interferon; STAT, signal transducer and activator of transcription; Ref, references.

2) Intracellular signaling pathways in lymphangiogenesis

Various growth factors activate their receptors on the surface of LyECs and initiate diverse signaling cascades that lead to the growth of lymphatic vessels (Table 2). These signaling pathways have largely been determined in studies of developmental lymphangiogenesis. Signaling via VEGF-C/D and VEGFR-3 is the most well-known pathway for lymphangiogenesis (Figure 3).6 VEGF-C or VEGF-D binding to VEGFR-3 results in autophosphorylation of multiple C-terminal tyrosine residues in VEGFR-391, which transduces signaling through the Ras/Raf/MEK/ERK pathway.92 Signal transduction also occurs through the PI3K/Akt pathway93 which causes phosphorylation of Akt, thereby activating mTOR and Rac1.94 Activation of these signaling pathways facilitates LyEC proliferation and migration, i.e. lymphangiogenesis.93 As discussed later, chronic inflammation and malignant tumors in the liver are known to induce several pro-lymphangiogenic growth factors including VEGF-C/D. However, a direct link between these increased pro-lymphangiogenic growth factors and lymphangiogenesis in these pathological conditions remains to be demonstrated (Figure 3). Excellent review papers are available for detailed signaling pathways in lymphangiogenesis.9597

Figure 3. Intracellular signaling pathways in lymphangiogenesis.

Figure 3

Signaling via VEGF-C/D and VEGFR-3 is the most well-known pathway for lymphangiogenesis. VEGF-C or VEGF-D binds to its receptor VEGFR-3 in the plasma membrane of lymphatic endothelial cells (LyECs), which facilitates signal transduction through various intracellular signaling pathways, leading to lymphangiogenesis. In the liver, activated macrophages in chronic inflammatory conditions, such as chronic hepatitis and liver cirrhosis, secrete VEGF-C and/or VEGF-D. Hepatic malignant tumors, such as hepatocellular carcinoma and intrahepatic cholangiocarcinoma, also secrete VEGF-C and/or VEGF-D. Further, these malignant tumors activate tumor-associated macrophages, which also secrete VEGF-C and/or VEGF-D. These secreted VEGF-C and VEGF-D are likely related to lymphangiogenesis in liver diseases through the VEGFR-3 mediated pathways.

3) Role of immune cells

Adaptive immune responses are initiated by migration of immune cells to inflamed sites. These immune cells phagocytose pathogens and transmigrate through lymphatic vessels to lymph nodes to present antigens to T cells. However, immune cells not only migrate through lymphatic vessels but also interact with lymphatic vessels and promote lymphangiogenesis.98 An increase in lymphatic vessels helps infiltrating immune cells to evacuate from inflamed sites via lymphatic vessels and accelerates resolution of inflammation.99101

Macrophage

Among various immune cells, macrophages most highly interact with lymphatic vessels. LyECs secrete chemotactic factors, such as C10, monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1 (MIP-1), to attract macrophages.102 Macrophages secrete lymphangiogenic cytokines, such as VEGF-C, VEGF-D and VEGF-A103, and promote tumor-associated lymphangiogenesis104 and inflammation-induced lymphangiogenesis in the cornea105, skin100 and tail106. Macrophages were recently indicated to have the ability to transdifferentiate to LyECs107109. However, this phenomenon is still controversial and needs further investigations.

Dendritic cell

Upregulation of inflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β in inflamed tissues promotes expression of chemokines (e.g., CCL21/CCL19 and CXCL12) and their receptors (e.g., CCR7 and CXCR-4) in LyECs and dendritic cells110113, which enhances transmigration of dendritic cells through LyECs.114, 115 Inflammatory cytokines also increase expression of adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1) and E-selectin in LyECs and promote dendritic cell transmigration to lymphatic vessels.116 Dendritic cells were also reported to secrete VEGF-C and promote lymphangiogenesis.117

T cell

In a mouse model of tail lymphedema, nude mice exhibited less edema than wild-type mice concomitant with decreased lymphangiogenic cytokines and increased antilymphangiogenic cytokines. The balance of these cytokines was modulated by T-cell mediated inflammation.90 T cells negatively regulated lymph node lymphangiogenesis by secreting interferon (IFN)-γ in mice.118

B cell

B cells promoted lymphangiogenesis in inflamed lymph nodes by secreting a robust amount of VEGF-A in mice given keyhole limpet hemocyanin (KLH) emulsified in complete Freund’s adjuvant (CFA) (an experimental model of inflamed lymph nodes).119 Interestingly, VEGF-C was not detected in this study. Another study using transgenic mice overexpressing VEGF-A specifically in B cells showed increased lymphangiogenesis as well as angiogenesis.120

Neutrophil

Neutrophils are reported to contribute to lymphangiogenesis by modulating bioavailability and bioactivity of VEGF-A and secreting VEGF-D.121 Bioavailability of VEGF-A is increased by the secretion of matrix metalloproteinases 9 and heparanase. Depletion of neutrophils in mice developed skin inflammation in response to immunization or contact hypersensitization. Further, lymphangiogenesis was decreased in these mice with increased local inflammation, suggesting that neutrophils play a role in lymphangiogenesis and that lymphangiogenesis is helpful for reducing inflammation.

4) Lymphangiogenesis in the liver

Given that 25–50% of lymph passing through the thoracic duct originates from the liver1,122, the liver can be considered the most important organ for lymphatic fluid production. However, the lymphatic vascular system in the liver has not been much explored. Only several studies have reported the occurrence of hepatic lymphangiogenesis in pathological conditions such as chronic hepatitis, liver fibrosis/cirrhosis, portal hypertension, malignant tumors and post-transplantation. This section summarizes these studies.

Chronic hepatitis, liver fibrosis and cirrhosis

The resistance to the blood flow in sinusoids increases in cirrhotic livers due to deformation of architecture including portal and central venules. Consequently, the hydrostatic pressure of sinusoids is elevated and plasma components filtrated through sinusoids, i.e., lymphatic fluid, increase. In cirrhotic patients, lymphatic fluid produced in the liver increased up to 30-fold123127 and peritoneoscopic observation showed dilation of lymphatic vessels in the liver surface.128

Ascites formation in association with cirrhosis is one of the most recognized clinical manifestations of lymphatic vascular system disorders. How ascites is formed still remains to be elucidated. While several theories have developed129131, currently the most accepted one is “the peripheral arterial vasodilation theory or “the forward theory”.132134 According to this theory, splanchnic arterial vasodilation caused by portal hypertension results in underfilling of the splanchnic arterial circulation or hypovolemia. In moderate stages, the hypovolemia is compensated by renal retention of sodium and water. However, severe portal hypertension and splanchnic arterial vasodilation make sodium and water retention persistent and lead to leakage of fluid into the peritoneal cavity. When its amount exceeds the absorption capacity of lymphatic vessels, ascites results.131, 135

On a related note, impaired lymphatic drainage in the splanchnic and peripheral regions was reported in cirrhotic rats with ascites. This was accompanied with a significantly increased activity of eNOS and increased production of nitric oxide (NO) in LyECs of these regions.136 In addition, smooth muscle cell coverage of the lymphatic vessels in these regions was significantly decreased. Treatment of these cirrhotic rats with a NOS inhibitor significantly improved lymphatic drainage, decreased ascites volume and increased smooth muscle cell coverage. This study thus suggests the role of NO in impairment of lymphatic vessels in the splanchnic and peripheral regions and the development of ascites. It is not known whether lymphatic vessels in cirrhotic livers show similar pathological features.

The occurrence of hepatic lymphangiogenesis was reported for the first time in liver fibrosis/cirrhosis by Vollmar B. et al. in 1997.137 They found lymphatic vessels increased and enlarged in cirrhotic rat livers induced by carbon tetrachloride (CCl4). These observations of hepatic lymphatic vessels were confirmed in the following year in patients with chronic viral hepatitis/cirrhosis.138

Microarray analysis demonstrated a 4-fold increase in VEGF-D expression in endothelial cells from CCl4-induced cirrhotic rat livers, compared to control rat livers. Given that VEGF-D is a well-known lymphangiogenic factor with binding to VEGFR-3139, which was also highly expressed in LyECs of these cirrhotic rats5, increased VEGF-D could be associated with lymphangiogenesis observed in liver cirrhosis (Figure 3).

Lymphangiogenesis was also reported to occur in idiopathic portal hypertension in human patients.140 It was presumed that increased lymph production due to increased portal pressure caused lymphangiogenesis. In two rat models of portal hypertension (portacaval shunt and portal vein ligation), upregulation of Vegfr-3 expression was observed, leading us to speculate the occurrence of lymphangiogenesis.141 In any case including those of chronic hepatitis and liver fibrosis/cirrhosis mentioned above, the significance and the mechanism of hepatic lymphangiogenesis remain unknown.

Malignant tumors

Lymphatic vessels play a pivotal role in the pathogenesis of malignant tumors by serving as a pathway through which tumor cells spread from their original places to other places. The incidence of lymph node metastasis differs among tumors. For example, it is 5.1% in hepatocellular carcinoma and is 45.1% in intrahepatic cholangiocarcinoma. The prognosis of tumor-bearing patients with lymph node metastasis is worse than the cases without metastasis.142, 143 Many malignant tumors are known to secrete lymphangiogenic factors such as VEGF-C and VEGF-D and promote lymphangiogenesis in their adjacent tissues, which helps tumor cells to metastasize to lymph nodes.144 In particular, many studies have demonstrated that tumor-associated macrophages play a vital role in lymphangiogenesis in malignant tumors by secreting VEGF-C and VEGF-D.104, 145147 In intrahepatic cholangiocarcinoma, the lymphatic vessel density of surgically resected tumors was positively correlated with the incidence of lymphatic metastasis.148 In hepatocellular carcinoma, VEGF-C expression was positively correlated with the size of tumors and the number of extrahepatic metastasis, and was negatively correlated with disease-free survival time.149 Thus, blockade of VEGF-C may be a potential therapeutic strategy against malignant tumors. In fact, VEGF-C neutralizing antibody (VGX-100) is under a Phase I clinical trial for adult patients with advanced or metastatic solid tumors (NCT01514123).150

Post-transplant lymphangiogenesis

In solid organ transplantations, the connection of lymphatic vessels between the graft and the recipient is interrupted at the time of operation. Since lymphatic vessels are essential for adaptive immunity, the association between lymphangiogenesis and graft rejection has received attention. Post-transplant lymphangiogenesis in grafts was associated with acute cellular graft rejection in various human transplantations (kidney151153, heart154 and lung155). However, a question still remains in regard to the pathological role of post-transplant lymphangiogenesis in graft rejection.153 Post-transplant lymphangiogenesis could be detrimental if newly formed lymphatic vessels promote antigen presentation in draining lymph nodes and provoke alloimmune responses that result in graft rejection. However, it could be beneficial if these newly formed lymphatic vessels efficiently clear immune cells. In fact, in a rat model of liver transplantation, post-transplant lymphangiogenesis in grafts was associated with a long-term survival of recipients for more than 90 days.156 In addition, rats that failed grafting by 11 days with acute cellular rejection and antibody-mediated rejection showed disappearance of lymphatic vessels from severely rejected areas, suggesting that lymphatic vessels take an important part in mitigation of inflammation at least in the early stage of transplantation. Further investigations to determine the mechanism and the time course of clearance of infiltrating immune cells by lymphatic vessels, especially in the early period of post-transplantation, may help successful grafting.

Conclusion and perspective

The lymphatic vascular system has been poorly studied in the liver. To drive research in this area, it is essential to identify better LyEC markers that do not overlap with LSECs, hepatocytes and other liver cells. Further, the development of experimental models for studying the lymphatic vascular system in postnatal livers will help to examine its role and molecular mechanisms in physiological and pathophysiological conditions. While all subjects are virtually novel in this area, it may be helpful to raise some specific questions to initiate the study.

First, the mechanism of hepatic lymphangiogenesis is largely unknown. The VEGFC/VEGFR-3 axis is considered the most potent signaling pathway that regulates lymphangiogenesis.97 However, cellular sources of VEGF-C and VEGFR-3 have not been fully identified in the liver. Further, as shown in Table 2, many other molecules are reported to regulate lymphangiogenesis. These molecules are mostly observed in the liver in physiological and pathophysiological conditions. It is worth characterizing these molecules in relation to hepatic lymphangiogenesis.

Second, the relation between the lymphatic vascular system and metastasis is well known. The growth of lymphatic capillaries in liver tumors has been observed. However, the role of their growth in the development and the progression of liver tumors is largely undetermined. Like angiogenesis, it would be interesting to investigate lymphangiogenesis in liver cancer.

Third, inflammation is closely related to the development of many liver diseases. Infiltrating immune cells are drained to lymphatic vessels. Thus, it would be interesting to examine lymphangiogenesis in relation to inflammation. It is also unknown how immune cells can recognize lymphatic vessels to migrate. Elucidation of these mechanisms may help to develop anti-inflammatory strategies by facilitating immune cell clearance.

Fourth, on a biological matter, although LyECs are derived from cardinal veins8, 83 and LSECs are derived from the septum transversum157, LyECs and LSECs have many similarities. As described previously, both LyECs and LSECs express LYVE-1.4043 VAP-1, a type II transmembrane protein that supports leukocyte adhesion, and Reelin, a glycoprotein that is associated with embryonic development, are also expressed in LyECs and LSECs.158, 159 In normal conditions, both LyECs and LSECs do not have basement membranes in lymphatic capillaries and sinusoids, respectively. Examining similarities and differences of these two types of endothelial cells could help to understand endothelial cell-related liver function.

As such, the lymphatic vascular system in the liver is a large open area for investigation160, which will significantly advance our understanding of liver physiology and pathophysiology and in turn contribute to the development of new therapeutic strategies for many liver diseases.

Synopsis (Summary).

The research of the lymphatic vascular system has advanced rapidly during the last decade and has shown its dysfunction implicated in the pathogeneses of various diseases. This review article provides an overview of the lymphatic vascular system in the liver.

Acknowledgments

Grant support

This work was supported by NIH grants R01 DK082600, R21AA023599 and Connecticut DPH grant #2015-0901 (YI), and a research fellowship of The Uehara Memorial Foundation and a grants-in-aid of The International Research Fund for Subsidy of Kyushu University School of Medicine Alumni (MT).

The authors thank Dr. Teruo Utsumi for his careful review of the manuscript and helpful suggestions.

Abbreviation used in this paper

VEGFR

vascular endothelial growth factor receptor

VEGF

vascular endothelial growth factor

LyEC

lymphatic endothelial cell

LYVE-1

lymphatic vessel endothelial hyaluronan receptor 1

Prox1

prospero homeobox protein 1

ADH

antidiuretic hormone

NO

nitric oxide

LSEC

liver sinusoidal endothelial cell

HA

hyaluronan

HCC

hepatocellular carcinoma

HNF

hepatocyte nuclear factor

ERR

estrogen-related receptor

LRH

liver receptor homolog

ROR

retinoic acid-related orphan receptor

EHE

epithelioid hemangioendothelioma

HBxAg

hepatitis B x antigen

HBV

hepatitis B virus

BDL

bile duct ligation

HBx Ag

Hepatitis B x antigen

HBV

hepatitis virus B

MCP-1

monocyte chemoattractant protein-1

MIP-1

macrophage inflammatory protein-1

TNF

tumor necrosis factor

IL

interleukin

ICAM-1

intercellular adhesion molecule 1

VCAM-1

vascular cell adhesion molecule 1

IFN

interferon

KLH

keyhole limpet hemocyanin

CFA

complete Freund’s adjuvant

CCl4

carbon tetrachloride

CCC

cholangiocellular carcinoma

FRC

fibroblastic reticular cell

CCL21

C-C motif chemokine ligand 21

HEV

high endothelial venules

MMR

macrophage mannose receptor 1

HSV-1

Herpes simplex virus 1

LV

lymphatic vessel

FGF-2

fibroblast growth factors-2

LPS

lipopolysaccharide

HGF

hepatocyte growth factor

LT

lymphotoxin

PI3K

phosphatidylinositol-4,5-bisphosphate 3-kinase

mTOR

mammalian target of rapamycin

TGF

tumor growth factor

STAT

signal transducer and activator of transcription

Footnotes

Disclosure

The authors disclose no conflicts.

Transcript Profiling

N/A

Writing Assistance

N/A

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Chung C, Iwakiri Y. The lymphatic vascular system in liver diseases: its role in ascites formation. Clin Mol Hepatol. 2013;19:99–104. doi: 10.3350/cmh.2013.19.2.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Tammela T, Alitalo K. Lymphangiogenesis: Molecular mechanisms and future promise. Cell. 2010;140:460–76. doi: 10.1016/j.cell.2010.01.045. [DOI] [PubMed] [Google Scholar]
  • 3.Schulte-Merker S, Sabine A, Petrova TV. Lymphatic vascular morphogenesis in development, physiology, and disease. J Cell Biol. 2011;193:607–18. doi: 10.1083/jcb.201012094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Koltowska K, Betterman KL, Harvey NL, Hogan BM. Getting out and about: the emergence and morphogenesis of the vertebrate lymphatic vasculature. Development. 2013;140:1857–70. doi: 10.1242/dev.089565. [DOI] [PubMed] [Google Scholar]
  • 5.Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VW, Fang GH, Dumont D, Breitman M, Alitalo K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc Natl Acad Sci U S A. 1995;92:3566–70. doi: 10.1073/pnas.92.8.3566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jeltsch M, Kaipainen a, Joukov V, Meng X, Lakso M, Rauvala H, Swartz M, Fukumura D, Jain RK, Alitalo K. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science (New York, NY) 1997;276:1423–25. doi: 10.1126/science.276.5317.1423. [DOI] [PubMed] [Google Scholar]
  • 7.Banerji S, Ni J, Wang SX, Clasper S, Su J, Tammi R, Jones M, Jackson DG. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. J Cell Biol. 1999;144:789–801. doi: 10.1083/jcb.144.4.789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wigle JT, Oliver G. Prox1 function is required for the development of the murine lymphatic system. Cell. 1999;98:769–78. doi: 10.1016/s0092-8674(00)81511-1. [DOI] [PubMed] [Google Scholar]
  • 9.Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K, Kerjaschki D. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 1999;154:385–94. doi: 10.1016/S0002-9440(10)65285-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Trutmann M, Sasse D. The lymphatics of the liver. Anat Embryol (Berl) 1994;190:201–9. doi: 10.1007/BF00234299. [DOI] [PubMed] [Google Scholar]
  • 11.Ohtani O, Ohtani Y. Lymph Circulation in the Liver. The Anatomical Record: Advances in Integrative Anatomy and Evolutionary Biology. 2008;291:643–52. doi: 10.1002/ar.20681. [DOI] [PubMed] [Google Scholar]
  • 12.Pupulim LF, Vilgrain V, Ronot M, Becker CD, Breguet R, Terraz S. Hepatic lymphatics: anatomy and related diseases. Abdom Imaging. 2015;40:1997–2011. doi: 10.1007/s00261-015-0350-y. [DOI] [PubMed] [Google Scholar]
  • 13.Alitalo K, Tammela T, Petrova TV. Lymphangiogenesis in development and human disease. Nature. 2005;438:946–53. doi: 10.1038/nature04480. [DOI] [PubMed] [Google Scholar]
  • 14.Maby-El Hajjami H, Petrova TV. Developmental and pathological lymphangiogenesis: from models to human disease. Histochem Cell Biol. 2008;130:1063–78. doi: 10.1007/s00418-008-0525-5. [DOI] [PubMed] [Google Scholar]
  • 15.Baluk P, Fuxe J, Hashizume H, Romano T, Lashnits E, Butz S, Vestweber D, Corada M, Molendini C, Dejana E, McDonald DM. Functionally specialized junctions between endothelial cells of lymphatic vessels. The Journal of experimental medicine. 2007;204:2349–62. doi: 10.1084/jem.20062596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Danussi C, Spessotto P, Petrucco A, Wassermann B, Sabatelli P, Montesi M, Doliana R, Bressan GM, Colombatti A. Emilin1 deficiency causes structural and functional defects of lymphatic vasculature. Mol Cell Biol. 2008;28:4026–39. doi: 10.1128/MCB.02062-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Solito R, Alessandrini C, Fruschelli M, Pucci AM, Gerli R. An immunological correlation between the anchoring filaments of initial lymph vessels and the neighboring elastic fibers: a unified morphofunctional concept. Lymphology. 1997;30:194–202. [PubMed] [Google Scholar]
  • 18.Breslin JW. Mechanical forces and lymphatic transport. Microvasc Res. 2014;96:46–54. doi: 10.1016/j.mvr.2014.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shirasawa Y, Benoit JN. Stretch-induced calcium sensitization of rat lymphatic smooth muscle. Am J Physiol Heart Circ Physiol. 2003;285:H2573–7. doi: 10.1152/ajpheart.00002.2003. [DOI] [PubMed] [Google Scholar]
  • 20.Davis MJ, Scallan JP, Wolpers JH, Muthuchamy M, Gashev AA, Zawieja DC. Intrinsic increase in lymphangion muscle contractility in response to elevated afterload. Am J Physiol Heart Circ Physiol. 2012;303:H795–808. doi: 10.1152/ajpheart.01097.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bohlen HG, Gasheva OY, Zawieja DC. Nitric oxide formation by lymphatic bulb and valves is a major regulatory component of lymphatic pumping. Am J Physiol Heart Circ Physiol. 2011;301:H1897–906. doi: 10.1152/ajpheart.00260.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shyy JY, Chien S. Role of integrins in endothelial mechanosensing of shear stress. Circ Res. 2002;91:769–75. doi: 10.1161/01.res.0000038487.19924.18. [DOI] [PubMed] [Google Scholar]
  • 23.Kunert C, Baish JW, Liao S, Padera TP, Munn LL. Mechanobiological oscillators control lymph flow. Proc Natl Acad Sci U S A. 2015;112:10938–43. doi: 10.1073/pnas.1508330112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Forster R, Braun A, Worbs T. Lymph node homing of T cells and dendritic cells via afferent lymphatics. Trends Immunol. 2012;33:271–80. doi: 10.1016/j.it.2012.02.007. [DOI] [PubMed] [Google Scholar]
  • 25.Jeltsch M, Tammela T, Alitalo K, Wilting J. Genesis and pathogenesis of lymphatic vessels. Cell Tissue Res. 2003;314:69–84. doi: 10.1007/s00441-003-0777-2. [DOI] [PubMed] [Google Scholar]
  • 26.Wake K, Sato T. “The sinusoid” in the liver: lessons learned from the original definition by Charles Sedgwick Minot (1900) Anat Rec (Hoboken) 2015;298:2071–80. doi: 10.1002/ar.23263. [DOI] [PubMed] [Google Scholar]
  • 27.Mall FP. A study of the structural unit of the liver. American Journal of Anatomy. 1906;5:227–308. [Google Scholar]
  • 28.Munoz SJ, Fenkel JM, Kiley K. Chapter 34: The Liver in Circulatory Failure. In: Schiff ER, Maddrey WC, Sorrell MF, editors. Schiff’s Diseases of the Liver. Wiley-Blackwell; Hoboken, GB: 2011. pp. 924–33. [Google Scholar]
  • 29.Ross MH. Histology: a text and atlas. 3. Lippincott Williams and Wilkins; 1995. [Google Scholar]
  • 30.Prevo R, Banerji S, Ferguson DJ, Clasper S, Jackson DG. Mouse LYVE-1 is an endocytic receptor for hyaluronan in lymphatic endothelium. J Biol Chem. 2001;276:19420–30. doi: 10.1074/jbc.M011004200. [DOI] [PubMed] [Google Scholar]
  • 31.Jackson DG. Biology of the lymphatic marker LYVE-1 and applications in research into lymphatic trafficking and lymphangiogenesis. APMIS. 2004;112:526–38. doi: 10.1111/j.1600-0463.2004.apm11207-0811.x. [DOI] [PubMed] [Google Scholar]
  • 32.Schacht V, Ramirez MI, Hong Y-K, Hirakawa S, Feng D, Harvey N, Williams M, Dvorak AM, Dvorak HF, Oliver G, Detmar M. T1α/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. The EMBO Journal. 2003;22:3546–56. doi: 10.1093/emboj/cdg342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wigle JT, Harvey N, Detmar M, Lagutina I, Grosveld G, Gunn MD, Jackson DG, Oliver G. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J. 2002;21:1505–13. doi: 10.1093/emboj/21.7.1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Duncan MK, Cui W, Oh D-J, Tomarev SI. Prox1 is differentially localized during lens development. Mechanisms of Development. 2002;112:195–98. doi: 10.1016/s0925-4773(01)00645-1. [DOI] [PubMed] [Google Scholar]
  • 35.Wilting J, Papoutsi M, Christ B, Nicolaides KH, von Kaisenberg CS, Borges J, Stark GB, Alitalo K, Tomarev SI, Niemeyer C, Rössler J. The transcription factor Prox1 is a marker for lymphatic endothelial cells in normal and diseased human tissues. The FASEB Journal. 2002 doi: 10.1096/fj.01-1010fje. [DOI] [PubMed] [Google Scholar]
  • 36.Baluk P, McDonald DM. Markers for microscopic imaging of lymphangiogenesis and angiogenesis. Ann N Y Acad Sci. 2008;1131:1–12. doi: 10.1196/annals.1413.001. [DOI] [PubMed] [Google Scholar]
  • 37.Neame PJ, Barry FP. The link proteins. Experientia. 1993;49:393–402. doi: 10.1007/BF01923584. [DOI] [PubMed] [Google Scholar]
  • 38.Gale NW, Prevo R, Espinosa J, Ferguson DJ, Dominguez MG, Yancopoulos GD, Thurston G, Jackson DG. Normal lymphatic development and function in mice deficient for the lymphatic hyaluronan receptor LYVE-1. Mol Cell Biol. 2007;27:595–604. doi: 10.1128/MCB.01503-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jang JY, Koh YJ, Lee SH, Lee J, Kim KH, Kim D, Koh GY, Yoo OJ. Conditional ablation of LYVE-1+ cells unveils defensive roles of lymphatic vessels in intestine and lymph nodes. Blood. 2013;122:2151–61. doi: 10.1182/blood-2013-01-478941. [DOI] [PubMed] [Google Scholar]
  • 40.Mouta Carreira C, Nasser SM, di Tomaso E, Padera TP, Boucher Y, Tomarev SI, Jain RK. LYVE-1 is not restricted to the lymph vessels: expression in normal liver blood sinusoids and down-regulation in human liver cancer and cirrhosis. Cancer Res. 2001;61:8079–84. [PubMed] [Google Scholar]
  • 41.Lalor PF, Lai WK, Curbishley SM, Shetty S, Adams DH. Human hepatic sinusoidal endothelial cells can be distinguished by expression of phenotypic markers related to their specialised functions in vivo. World J Gastroenterol. 2006;12:5429–39. doi: 10.3748/wjg.v12.i34.5429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Arimoto J, Ikura Y, Suekane T, Nakagawa M, Kitabayashi C, Iwasa Y, Sugioka K, Naruko T, Arakawa T, Ueda M. Expression of LYVE-1 in sinusoidal endothelium is reduced in chronically inflamed human livers. J Gastroenterol. 2010;45:317–25. doi: 10.1007/s00535-009-0152-5. [DOI] [PubMed] [Google Scholar]
  • 43.Nonaka H, Tanaka M, Suzuki K, Miyajima A. Development of murine hepatic sinusoidal endothelial cells characterized by the expression of hyaluronan receptors. Dev Dyn. 2007;236:2258–67. doi: 10.1002/dvdy.21227. [DOI] [PubMed] [Google Scholar]
  • 44.Kitagawa K, Nakajima G, Kuramochi H, Ariizumi SI, Yamamoto M. Lymphatic vessel endothelial hyaluronan receptor-1 is a novel prognostic indicator for human hepatocellular carcinoma. Mol Clin Oncol. 2013;1:1039–48. doi: 10.3892/mco.2013.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hong YK, Harvey N, Noh YH, Schacht V, Hirakawa S, Detmar M, Oliver G. Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate. Dev Dyn. 2002;225:351–7. doi: 10.1002/dvdy.10163. [DOI] [PubMed] [Google Scholar]
  • 46.Ordonez NG. Immunohistochemical endothelial markers: a review. Adv Anat Pathol. 2012;19:281–95. doi: 10.1097/PAP.0b013e3182691c2a. [DOI] [PubMed] [Google Scholar]
  • 47.Wigle JT, Chowdhury K, Gruss P, Oliver G. Prox1 function is crucial for mouse lens-fibre elongation. Nat Genet. 1999;21:318–22. doi: 10.1038/6844. [DOI] [PubMed] [Google Scholar]
  • 48.Dyer MA, Livesey FJ, Cepko CL, Oliver G. Prox1 function controls progenitor cell proliferation and horizontal cell genesis in the mammalian retina. Nat Genet. 2003;34:53–8. doi: 10.1038/ng1144. [DOI] [PubMed] [Google Scholar]
  • 49.Risebro CA, Searles RG, Melville AA, Ehler E, Jina N, Shah S, Pallas J, Hubank M, Dillard M, Harvey NL, Schwartz RJ, Chien KR, Oliver G, Riley PR. Prox1 maintains muscle structure and growth in the developing heart. Development. 2009;136:495–505. doi: 10.1242/dev.030007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lavado A, Oliver G. Prox1 expression patterns in the developing and adult murine brain. Dev Dyn. 2007;236:518–24. doi: 10.1002/dvdy.21024. [DOI] [PubMed] [Google Scholar]
  • 51.Burke Z, Oliver G. Prox1 is an early specific marker for the developing liver and pancreas in the mammalian foregut endoderm. Mech Dev. 2002;118:147–55. doi: 10.1016/s0925-4773(02)00240-x. [DOI] [PubMed] [Google Scholar]
  • 52.Dudas J, Elmaouhoub A, Mansuroglu T, Batusic D, Tron K, Saile B, Papoutsi M, Pieler T, Wilting J, Ramadori G. Prospero-related homeobox 1 (Prox1) is a stable hepatocyte marker during liver development, injury and regeneration, and is absent from “oval cells”. Histochem Cell Biol. 2006;126:549–62. doi: 10.1007/s00418-006-0191-4. [DOI] [PubMed] [Google Scholar]
  • 53.Sosa-Pineda B, Wigle JT, Oliver G. Hepatocyte migration during liver development requires Prox1. Nat Genet. 2000;25:254–5. doi: 10.1038/76996. [DOI] [PubMed] [Google Scholar]
  • 54.Wigle JT, Harvey N, Detmar M, Lagutina I, Grosveld G, Gunn MD, Jackson DG, Oliver G. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. The EMBO Journal. 2002;21:1505–13. doi: 10.1093/emboj/21.7.1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Choi I, Chung HK, Ramu S, Lee HN, Kim KE, Lee S, Yoo J, Choi D, Lee YS, Aguilar B, Hong YK. Visualization of lymphatic vessels by Prox1-promoter directed GFP reporter in a bacterial artificial chromosome-based transgenic mouse. Blood. 2011;117:362–5. doi: 10.1182/blood-2010-07-298562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hagerling R, Pollmann C, Kremer L, Andresen V, Kiefer F. Intravital two-photon microscopy of lymphatic vessel development and function using a transgenic Prox1 promoter-directed mOrange2 reporter mouse. Biochem Soc Trans. 2011;39:1674–81. doi: 10.1042/BST20110722. [DOI] [PubMed] [Google Scholar]
  • 57.Bianchi R, Teijeira A, Proulx ST, Christiansen AJ, Seidel CD, Rulicke T, Makinen T, Hagerling R, Halin C, Detmar M. A transgenic Prox1-Cre-tdTomato reporter mouse for lymphatic vessel research. PLoS One. 2015;10:e0122976. doi: 10.1371/journal.pone.0122976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Truman LA, NAG, Bentley KL, Ruddle NH. Lymphatic vessel function in head and neck inflammation. Lymphat Res Biol. 2013;11:187–92. doi: 10.1089/lrb.2013.0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Truman LA, Bentley KL, Smith EC, Massaro SA, Gonzalez DG, Haberman AM, Hill M, Jones D, Min W, Krause DS, Ruddle NH. ProxTom lymphatic vessel reporter mice reveal Prox1 expression in the adrenal medulla, megakaryocytes, and platelets. Am J Pathol. 2012;180:1715–25. doi: 10.1016/j.ajpath.2011.12.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dudas J, Mansuroglu T, Moriconi F, Haller F, Wilting J, Lorf T, Füzesi L, Ramadori G. Altered regulation of Prox1-gene-expression in liver tumors. BMC Cancer. 2008;8:1–15. doi: 10.1186/1471-2407-8-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shimoda M, Takahashi M, Yoshimoto T, Kono T, Ikai I, Kubo H. A homeobox protein, prox1, is involved in the differentiation, proliferation, and prognosis in hepatocellular carcinoma. Clin Cancer Res. 2006;12:6005–11. doi: 10.1158/1078-0432.CCR-06-0712. [DOI] [PubMed] [Google Scholar]
  • 62.Song KH, Li T, Chiang JY. A Prospero-related homeodomain protein is a novel co-regulator of hepatocyte nuclear factor 4alpha that regulates the cholesterol 7alpha-hydroxylase gene. J Biol Chem. 2006;281:10081–8. doi: 10.1074/jbc.M513420200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Charest-Marcotte A, Dufour CR, Wilson BJ, Tremblay AM, Eichner LJ, Arlow DH, Mootha VK, Giguere V. The homeobox protein Prox1 is a negative modulator of ERR{alpha}/PGC-1{alpha} bioenergetic functions. Genes Dev. 2010;24:537–42. doi: 10.1101/gad.1871610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Dufour CR, Levasseur MP, Pham NH, Eichner LJ, Wilson BJ, Charest-Marcotte A, Duguay D, Poirier-Heon JF, Cermakian N, Giguere V. Genomic convergence among ERRalpha, PROX1, and BMAL1 in the control of metabolic clock outputs. PLoS Genet. 2011;7:e1002143. doi: 10.1371/journal.pgen.1002143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Qin J, Gao DM, Jiang QF, Zhou Q, Kong YY, Wang Y, Xie YH. Prospero-related homeobox (Prox1) is a corepressor of human liver receptor homolog-1 and suppresses the transcription of the cholesterol 7-alpha-hydroxylase gene. Mol Endocrinol. 2004;18:2424–39. doi: 10.1210/me.2004-0009. [DOI] [PubMed] [Google Scholar]
  • 66.Takeda Y, Jetten AM. Prospero-related homeobox 1 (Prox1) functions as a novel modulator of retinoic acid-related orphan receptors alpha- and gamma-mediated transactivation. Nucleic Acids Res. 2013;41:6992–7008. doi: 10.1093/nar/gkt447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Martin-Villar E, Scholl FG, Gamallo C, Yurrita MM, Munoz-Guerra M, Cruces J, Quintanilla M. Characterization of human PA2. 26 antigen (T1alpha-2, podoplanin), a small membrane mucin induced in oral squamous cell carcinomas. Int J Cancer. 2005;113:899–910. doi: 10.1002/ijc.20656. [DOI] [PubMed] [Google Scholar]
  • 68.Mahtab EA, Wijffels MC, Van Den Akker NM, Hahurij ND, Lie-Venema H, Wisse LJ, Deruiter MC, Uhrin P, Zaujec J, Binder BR, Schalij MJ, Poelmann RE, Gittenberger-De Groot AC. Cardiac malformations and myocardial abnormalities in podoplanin knockout mouse embryos: Correlation with abnormal epicardial development. Dev Dyn. 2008;237:847–57. doi: 10.1002/dvdy.21463. [DOI] [PubMed] [Google Scholar]
  • 69.Mahtab EA, Vicente-Steijn R, Hahurij ND, Jongbloed MR, Wisse LJ, DeRuiter MC, Uhrin P, Zaujec J, Binder BR, Schalij MJ, Poelmann RE, Gittenberger-de Groot AC. Podoplanin deficient mice show a RhoA-related hypoplasia of the sinus venosus myocardium including the sinoatrial node. Dev Dyn. 2009;238:183–93. doi: 10.1002/dvdy.21819. [DOI] [PubMed] [Google Scholar]
  • 70.Douglas YL, Mahtab EA, Jongbloed MR, Uhrin P, Zaujec J, Binder BR, Schalij MJ, Poelmann RE, Deruiter MC, Gittenberger-de Groot AC. Pulmonary vein, dorsal atrial wall and atrial septum abnormalities in podoplanin knockout mice with disturbed posterior heart field contribution. Pediatr Res. 2009;65:27–32. doi: 10.1203/PDR.0b013e31818bc11a. [DOI] [PubMed] [Google Scholar]
  • 71.Ramirez MI, Millien G, Hinds A, Cao Y, Seldin DC, Williams MC. T1alpha, a lung type I cell differentiation gene, is required for normal lung cell proliferation and alveolus formation at birth. Dev Biol. 2003;256:61–72. doi: 10.1016/s0012-1606(02)00098-2. [DOI] [PubMed] [Google Scholar]
  • 72.Bekiaris V, Withers D, Glanville SH, McConnell FM, Parnell SM, Kim MY, Gaspal FM, Jenkinson E, Sweet C, Anderson G, Lane PJ. Role of CD30 in B/T segregation in the spleen. J Immunol. 2007;179:7535–43. doi: 10.4049/jimmunol.179.11.7535. [DOI] [PubMed] [Google Scholar]
  • 73.Astarita JL, Acton SE, Turley SJ. Podoplanin: emerging functions in development, the immune system, and cancer. Front Immunol. 2012;3:283. doi: 10.3389/fimmu.2012.00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schacht V, Ramirez MI, Hong YK, Hirakawa S, Feng D, Harvey N, Williams M, Dvorak AM, Dvorak HF, Oliver G, Detmar M. T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J. 2003;22:3546–56. doi: 10.1093/emboj/cdg342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Baars S, Bauer C, Szabowski S, Hartenstein B, Angel P. Epithelial deletion of podoplanin is dispensable for re-epithelialization of skin wounds. Exp Dermatol. 2015;24:785–7. doi: 10.1111/exd.12781. [DOI] [PubMed] [Google Scholar]
  • 76.Herzog BH, Fu J, Wilson SJ, Hess PR, Sen A, McDaniel JM, Pan Y, Sheng M, Yago T, Silasi-Mansat R, McGee S, May F, Nieswandt B, Morris AJ, Lupu F, Coughlin SR, McEver RP, Chen H, Kahn ML, Xia L. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature. 2013;502:105–9. doi: 10.1038/nature12501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li Y, Wang J, Asahina K. Mesothelial cells give rise to hepatic stellate cells and myofibroblasts via mesothelial-mesenchymal transition in liver injury. Proc Natl Acad Sci U S A. 2013;110:2324–9. doi: 10.1073/pnas.1214136110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yokomori H, Oda M, Kaneko F, Kawachi S, Tanabe M, Yoshimura K, Kitagawa Y, Hibi T. Lymphatic marker podoplanin/D2-40 in human advanced cirrhotic liver--re-evaluations of microlymphatic abnormalities. BMC Gastroenterol. 2010;10:131. doi: 10.1186/1471-230X-10-131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Link A, Hardie DL, Favre S, Britschgi MR, Adams DH, Sixt M, Cyster JG, Buckley CD, Luther SA. Association of T-zone reticular networks and conduits with ectopic lymphoid tissues in mice and humans. Am J Pathol. 2011;178:1662–75. doi: 10.1016/j.ajpath.2010.12.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fujii T, Zen Y, Sato Y, Sasaki M, Enomae M, Minato H, Masuda S, Uehara T, Katsuyama T, Nakanuma Y. Podoplanin is a useful diagnostic marker for epithelioid hemangioendothelioma of the liver. Mod Pathol. 2008;21:125–30. doi: 10.1038/modpathol.3800986. [DOI] [PubMed] [Google Scholar]
  • 81.Xian ZH, Cong WM, Lu XY, Yu H, Wu MC. Angiogenesis and lymphangiogenesis in sporadic hepatic angiomyolipoma. Pathol Res Pract. 2011;207:403–9. doi: 10.1016/j.prp.2011.04.008. [DOI] [PubMed] [Google Scholar]
  • 82.Dumont DJ, Jussila L, Taipale J, Lymboussaki A, Mustonen T, Pajusola K, Breitman M, Alitalo K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science. 1998;282:946–9. doi: 10.1126/science.282.5390.946. [DOI] [PubMed] [Google Scholar]
  • 83.Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, Jeltsch M, Jackson DG, Talikka M, Rauvala H, Betsholtz C, Alitalo K. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol. 2004;5:74–80. doi: 10.1038/ni1013. [DOI] [PubMed] [Google Scholar]
  • 84.Martinez-Corral I, Olmeda D, Dieguez-Hurtado R, Tammela T, Alitalo K, Ortega S. In vivo imaging of lymphatic vessels in development, wound healing, inflammation, and tumor metastasis. Proc Natl Acad Sci U S A. 2012;109:6223–8. doi: 10.1073/pnas.1115542109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Gaudio E, Barbaro B, Alvaro D, Glaser S, Francis H, Ueno Y, Meininger CJ, Franchitto A, Onori P, Marzioni M, Taffetani S, Fava G, Stoica G, Venter J, Reichenbach R, De Morrow S, Summers R, Alpini G. Vascular endothelial growth factor stimulates rat cholangiocyte proliferation via an autocrine mechanism. Gastroenterology. 2006;130:1270–82. doi: 10.1053/j.gastro.2005.12.034. [DOI] [PubMed] [Google Scholar]
  • 86.Franchitto A, Onori P, Renzi A, Carpino G, Mancinelli R, Alvaro D, Gaudio E. Expression of vascular endothelial growth factors and their receptors by hepatic progenitor cells in human liver diseases. Hepatobiliary Surg Nutr. 2013;2:68–77. doi: 10.3978/j.issn.2304-3881.2012.10.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lian Z, Liu J, Wu M, Wang HY, Arbuthnot P, Kew M, Feitelson MA. Hepatitis B x antigen upregulates vascular endothelial growth factor receptor 3 in hepatocarcinogenesis. Hepatology. 2007;45:1390–9. doi: 10.1002/hep.21610. [DOI] [PubMed] [Google Scholar]
  • 88.Paupert J, Sounni NE, Noel A. Lymphangiogenesis in post-natal tissue remodeling: lymphatic endothelial cell connection with its environment. Mol Aspects Med. 2011;32:146–58. doi: 10.1016/j.mam.2011.04.002. [DOI] [PubMed] [Google Scholar]
  • 89.Kelley PM, Steele MM, Tempero RM. Regressed lymphatic vessels develop during corneal repair. Lab Invest. 2011;91:1643–51. doi: 10.1038/labinvest.2011.121. [DOI] [PubMed] [Google Scholar]
  • 90.Zampell JC, Avraham T, Yoder N, Fort N, Yan A, Weitman ES, Mehrara BJ. Lymphatic function is regulated by a coordinated expression of lymphangiogenic and anti-lymphangiogenic cytokines. Am J Physiol Cell Physiol. 2012;302:C392–404. doi: 10.1152/ajpcell.00306.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dixelius J, Makinen T, Wirzenius M, Karkkainen MJ, Wernstedt C, Alitalo K, Claesson-Welsh L. Ligand-induced vascular endothelial growth factor receptor-3 (VEGFR-3) heterodimerization with VEGFR-2 in primary lymphatic endothelial cells regulates tyrosine phosphorylation sites. J Biol Chem. 2003;278:40973–9. doi: 10.1074/jbc.M304499200. [DOI] [PubMed] [Google Scholar]
  • 92.Ichise T, Yoshida N, Ichise H. H-, N- and Kras cooperatively regulate lymphatic vessel growth by modulating VEGFR3 expression in lymphatic endothelial cells in mice. Development. 2010;137:1003–13. doi: 10.1242/dev.043489. [DOI] [PubMed] [Google Scholar]
  • 93.Makinen T, Veikkola T, Mustjoki S, Karpanen T, Catimel B, Nice EC, Wise L, Mercer A, Kowalski H, Kerjaschki D, Stacker SA, Achen MG, Alitalo K. Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3. EMBO J. 2001;20:4762–73. doi: 10.1093/emboj/20.17.4762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Vanhaesebroeck B, Stephens L, Hawkins P. PI3K signalling: the path to discovery and understanding. Nat Rev Mol Cell Biol. 2012;13:195–203. doi: 10.1038/nrm3290. [DOI] [PubMed] [Google Scholar]
  • 95.Zheng W, Aspelund A, Alitalo K. Lymphangiogenic factors, mechanisms, and applications. J Clin Invest. 2014;124:878–87. doi: 10.1172/JCI71603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Coso S, Bovay E, Petrova TV. Pressing the right buttons: signaling in lymphangiogenesis. Blood. 2014;123:2614–24. doi: 10.1182/blood-2013-12-297317. [DOI] [PubMed] [Google Scholar]
  • 97.Secker GA, Harvey NL. VEGFR signaling during lymphatic vascular development: From progenitor cells to functional vessels. Dev Dyn. 2015;244:323–31. doi: 10.1002/dvdy.24227. [DOI] [PubMed] [Google Scholar]
  • 98.Kim H, Kataru RP, Koh GY. Inflammation-associated lymphangiogenesis: a double-edged sword? J Clin Invest. 2014;124:936–42. doi: 10.1172/JCI71607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6:1191–7. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 100.Kataru RP, Jung K, Jang C, Yang H, Schwendener RA, Baik JE, Han SH, Alitalo K, Koh GY. Critical role of CD11b+ macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution. Blood. 2009;113:5650–9. doi: 10.1182/blood-2008-09-176776. [DOI] [PubMed] [Google Scholar]
  • 101.Kataru RP, Lee YG, Koh GY. Interactions of immune cells and lymphatic vessels. Adv Anat Embryol Cell Biol. 2014;214:107–18. doi: 10.1007/978-3-7091-1646-3_9. [DOI] [PubMed] [Google Scholar]
  • 102.Mancardi S, Vecile E, Dusetti N, Calvo E, Stanta G, Burrone OR, Dobrina A. Evidence of CXC, CC and C chemokine production by lymphatic endothelial cells. Immunology. 2003;108:523–30. doi: 10.1046/j.1365-2567.2003.01613.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ji RC. Macrophages are important mediators of either tumor- or inflammation-induced lymphangiogenesis. Cell Mol Life Sci. 2012;69:897–914. doi: 10.1007/s00018-011-0848-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Schoppmann SF, Birner P, Stockl J, Kalt R, Ullrich R, Caucig C, Kriehuber E, Nagy K, Alitalo K, Kerjaschki D. Tumor-associated macrophages express lymphatic endothelial growth factors and are related to peritumoral lymphangiogenesis. Am J Pathol. 2002;161:947–56. doi: 10.1016/S0002-9440(10)64255-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C, D’Amore PA, Dana MR, Wiegand SJ, Streilein JW. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest. 2004;113:1040–50. doi: 10.1172/JCI20465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yan A, Avraham T, Zampell JC, Aschen SZ, Mehrara BJ. Mechanisms of lymphatic regeneration after tissue transfer. PLoS One. 2011;6:e17201. doi: 10.1371/journal.pone.0017201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Maruyama K, Ii M, Cursiefen C, Jackson DG, Keino H, Tomita M, Van Rooijen N, Takenaka H, D’Amore PA, Stein-Streilein J, Losordo DW, Streilein JW. Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages. J Clin Invest. 2005;115:2363–72. doi: 10.1172/JCI23874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kerjaschki D, Huttary N, Raab I, Regele H, Bojarski-Nagy K, Bartel G, Kröber SM, Greinix H, Rosenmaier A, Karlhofer F, Wick N, Mazal PR. Lymphatic endothelial progenitor cells contribute to de novo lymphangiogenesis in human renal transplants. Nature medicine. 2006;12:230–34. doi: 10.1038/nm1340. [DOI] [PubMed] [Google Scholar]
  • 109.Lee S, Choi I, Hong YK. Heterogeneity and plasticity of lymphatic endothelial cells. Semin Thromb Hemost. 2010;36:352–61. doi: 10.1055/s-0030-1253457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Vigl B, Aebischer D, Nitschke M, Iolyeva M, Rothlin T, Antsiferova O, Halin C. Tissue inflammation modulates gene expression of lymphatic endothelial cells and dendritic cell migration in a stimulus-dependent manner. Blood. 2011;118:205–15. doi: 10.1182/blood-2010-12-326447. [DOI] [PubMed] [Google Scholar]
  • 111.Kabashima K, Shiraishi N, Sugita K, Mori T, Onoue A, Kobayashi M, Sakabe J, Yoshiki R, Tamamura H, Fujii N, Inaba K, Tokura Y. CXCL12-CXCR4 engagement is required for migration of cutaneous dendritic cells. Am J Pathol. 2007;171:1249–57. doi: 10.2353/ajpath.2007.070225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Teijeira A, Rouzaut A, Melero I. Initial afferent lymphatic vessels controlling outbound leukocyte traffic from skin to lymph nodes. Front Immunol. 2013;4:433. doi: 10.3389/fimmu.2013.00433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Johnson LA, Jackson DG. Control of dendritic cell trafficking in lymphatics by chemokines. Angiogenesis. 2014;17:335–45. doi: 10.1007/s10456-013-9407-0. [DOI] [PubMed] [Google Scholar]
  • 114.Saeki H, Moore AM, Brown MJ, Hwang ST. Cutting edge: secondary lymphoid-tissue chemokine (SLC) and CC chemokine receptor 7 (CCR7) participate in the emigration pathway of mature dendritic cells from the skin to regional lymph nodes. J Immunol. 1999;162:2472–5. [PubMed] [Google Scholar]
  • 115.Tal O, Lim HY, Gurevich I, Milo I, Shipony Z, Ng LG, Angeli V, Shakhar G. DC mobilization from the skin requires docking to immobilized CCL21 on lymphatic endothelium and intralymphatic crawling. J Exp Med. 2011;208:2141–53. doi: 10.1084/jem.20102392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Johnson LA, Clasper S, Holt AP, Lalor PF, Baban D, Jackson DG. An inflammation-induced mechanism for leukocyte transmigration across lymphatic vessel endothelium. J Exp Med. 2006;203:2763–77. doi: 10.1084/jem.20051759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gagliostro V, Seeger P, Garrafa E, Salvi V, Bresciani R, Bosisio D, Sozzani S. Pro-lymphangiogenic properties of IFN-gamma-activated human dendritic cells. Immunol Lett. 2016;173:26–35. doi: 10.1016/j.imlet.2016.03.008. [DOI] [PubMed] [Google Scholar]
  • 118.Kataru RP, Kim H, Jang C, Choi DK, Koh BI, Kim M, Gollamudi S, Kim YK, Lee SH, Koh GY. T lymphocytes negatively regulate lymph node lymphatic vessel formation. Immunity. 2011;34:96–107. doi: 10.1016/j.immuni.2010.12.016. [DOI] [PubMed] [Google Scholar]
  • 119.Angeli V, Ginhoux F, Llodra J, Quemeneur L, Frenette PS, Skobe M, Jessberger R, Merad M, Randolph GJ. B cell-driven lymphangiogenesis in inflamed lymph nodes enhances dendritic cell mobilization. Immunity. 2006;24:203–15. doi: 10.1016/j.immuni.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 120.Shrestha B, Hashiguchi T, Ito T, Miura N, Takenouchi K, Oyama Y, Kawahara K, Tancharoen S, Ki IY, Arimura N, Yoshinaga N, Noma S, Shrestha C, Nitanda T, Kitajima S, Arimura K, Sato M, Sakamoto T, Maruyama I. B cell-derived vascular endothelial growth factor A promotes lymphangiogenesis and high endothelial venule expansion in lymph nodes. J Immunol. 2010;184:4819–26. doi: 10.4049/jimmunol.0903063. [DOI] [PubMed] [Google Scholar]
  • 121.Tan KW, Chong SZ, Wong FH, Evrard M, Tan SM, Keeble J, Kemeny DM, Ng LG, Abastado JP, Angeli V. Neutrophils contribute to inflammatory lymphangiogenesis by increasing VEGF-A bioavailability and secreting VEGF-D. Blood. 2013;122:3666–77. doi: 10.1182/blood-2012-11-466532. [DOI] [PubMed] [Google Scholar]
  • 122.Cain JC, Grindlay JH. Lymph from liver and thoracic duct; an experimental study. Surg Gynecol Obstet. 1947;85:558–62. [PubMed] [Google Scholar]
  • 123.Nix JT, Flock EV, Bollman JL. Influence of cirrhosis on proteins of cisternal lymph. Am J Physiol. 1951;164:117–8. doi: 10.1152/ajplegacy.1950.164.1.117. [DOI] [PubMed] [Google Scholar]
  • 124.Dumont AE, Mulholland JH. Flow rate and composition of thoracic-duct lymph in patients with cirrhosis. N Engl J Med. 1960;263:471–4. doi: 10.1056/NEJM196009082631001. [DOI] [PubMed] [Google Scholar]
  • 125.Dumont AE, Mulholland JH. Alterations in Thoracic Duct Lymph Flow in Hepatic Cirrhosis: Significance in Portal Hypertension. Ann Surg. 1962;156:668–75. doi: 10.1097/00000658-196210000-00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Witte CL, Witte MH, Dumont AE. Lymph imbalance in the genesis and perpetuation of the ascites syndrome in hepatic cirrhosis. Gastroenterology. 1980;78:1059–68. [PubMed] [Google Scholar]
  • 127.Barrowman JA, Granger DN. Effects of experimental cirrhosis on splanchnic microvascular fluid and solute exchange in the rat. Gastroenterology. 1984;87:165–72. [PubMed] [Google Scholar]
  • 128.Shimada Y. Observations on hepatic superficial lymph flow. Lymphology. 1979;12:11–3. [PubMed] [Google Scholar]
  • 129.Atkinson M, Losowsky MS. The mechanism of ascites formation in chronic liver disease. Q J Med. 1961;30:153–66. [PubMed] [Google Scholar]
  • 130.Lieberman FL, Ito S, Reynolds TB. Effective plasma volume in cirrhosis with ascites. Evidence that a decreased value does not account for renal sodium retention, a sponteneous reduction in glomerular filtration rate (GFR), and a fall in GFR during drug-induced diuresis. J Clin Invest. 1969;48:975–81. doi: 10.1172/JCI106078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Schrier RW, Arroyo V, Bernardi M, Epstein M, Henriksen JH, Rodes J. Peripheral arterial vasodilation hypothesis: a proposal for the initiation of renal sodium and water retention in cirrhosis. Hepatology. 1988;8:1151–7. doi: 10.1002/hep.1840080532. [DOI] [PubMed] [Google Scholar]
  • 132.De Franchis R, Salerno F. Pathogenesis of ascites and predictors of resistance to therapy. J Gastroenterol Hepatol. 2002;17(Suppl 3):S242–7. doi: 10.1046/j.1440-1746.17.s3.7.x. [DOI] [PubMed] [Google Scholar]
  • 133.Arroyo V, Colmenero J. Ascites and hepatorenal syndrome in cirrhosis: pathophysiological basis of therapy and current management. J Hepatol. 2003;38(Suppl 1):S69–89. doi: 10.1016/s0168-8278(03)00007-2. [DOI] [PubMed] [Google Scholar]
  • 134.Gordon FD. Ascites. Clin Liver Dis. 2012;16:285–99. doi: 10.1016/j.cld.2012.03.004. [DOI] [PubMed] [Google Scholar]
  • 135.Sanyal AJ, Bosch J, Blei A, Arroyo V. Portal hypertension and its complications. Gastroenterology. 2008;134:1715–28. doi: 10.1053/j.gastro.2008.03.007. [DOI] [PubMed] [Google Scholar]
  • 136.Ribera J, Pauta M, Melgar-Lesmes P, Tugues S, Fernandez-Varo G, Held KF, Soria G, Tudela R, Planas AM, Fernandez-Hernando C, Arroyo V, Jimenez W, Morales-Ruiz M. Increased nitric oxide production in lymphatic endothelial cells causes impairment of lymphatic drainage in cirrhotic rats. Gut. 2013;62:138–45. doi: 10.1136/gutjnl-2011-300703. [DOI] [PubMed] [Google Scholar]
  • 137.Vollmar B, Wolf B, Siegmund S, Katsen AD, Menger MD. Lymph vessel expansion and function in the development of hepatic fibrosis and cirrhosis. Am J Pathol. 1997;151:169–75. [PMC free article] [PubMed] [Google Scholar]
  • 138.Yamauchi Y, Michitaka K, Onji M. Morphometric analysis of lymphatic and blood vessels in human chronic viral liver diseases. Am J Pathol. 1998;153:1131–7. doi: 10.1016/S0002-9440(10)65657-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Achen MG, Jeltsch M, Kukk E, Makinen T, Vitali A, Wilks AF, Alitalo K, Stacker SA. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4) Proc Natl Acad Sci U S A. 1998;95:548–53. doi: 10.1073/pnas.95.2.548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Oikawa H, Masuda T, Sato S, Yashima A, Suzuki K, Sato S, Satodate R. Changes in lymph vessels and portal veins in the portal tract of patients with idiopathic portal hypertension: a morphometric study. Hepatology. 1998;27:1607–10. doi: 10.1002/hep.510270621. [DOI] [PubMed] [Google Scholar]
  • 141.Guerin F, Wagner M, Line A, Zappa M, Fasseu M, Paradis V, Vilgrain V, Van Beers BE, Legagneux J, Moreau R, Letteron P. Hepatic proliferation and angiogenesis markers are increased after portal deprivation in rats: a study of molecular, histological and radiological changes. PLoS One. 2015;10:e0125493. doi: 10.1371/journal.pone.0125493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sun HC, Zhuang PY, Qin LX, Ye QH, Wang L, Ren N, Zhang JB, Qian YB, Lu L, Fan J, Tang ZY. Incidence and prognostic values of lymph node metastasis in operable hepatocellular carcinoma and evaluation of routine complete lymphadenectomy. J Surg Oncol. 2007;96:37–45. doi: 10.1002/jso.20772. [DOI] [PubMed] [Google Scholar]
  • 143.Yamamoto M, Takasaki K, Yoshikawa T. Lymph node metastasis in intrahepatic cholangiocarcinoma. Jpn J Clin Oncol. 1999;29:147–50. doi: 10.1093/jjco/29.3.147. [DOI] [PubMed] [Google Scholar]
  • 144.Das S, Skobe M. Lymphatic vessel activation in cancer. Ann N Y Acad Sci. 2008;1131:235–41. doi: 10.1196/annals.1413.021. [DOI] [PubMed] [Google Scholar]
  • 145.Skobe M, Hamberg LM, Hawighorst T, Schirner M, Wolf GL, Alitalo K, Detmar M. Concurrent induction of lymphangiogenesis, angiogenesis, and macrophage recruitment by vascular endothelial growth factor-C in melanoma. Am J Pathol. 2001;159:893–903. doi: 10.1016/S0002-9440(10)61765-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Iwata C, Kano MR, Komuro A, Oka M, Kiyono K, Johansson E, Morishita Y, Yashiro M, Hirakawa K, Kaminishi M, Miyazono K. Inhibition of cyclooxygenase-2 suppresses lymph node metastasis via reduction of lymphangiogenesis. Cancer Res. 2007;67:10181–9. doi: 10.1158/0008-5472.CAN-07-2366. [DOI] [PubMed] [Google Scholar]
  • 147.Schoppmann SF, Fenzl A, Nagy K, Unger S, Bayer G, Geleff S, Gnant M, Horvat R, Jakesz R, Birner P. VEGF-C expressing tumor-associated macrophages in lymph node positive breast cancer: impact on lymphangiogenesis and survival. Surgery. 2006;139:839–46. doi: 10.1016/j.surg.2005.12.008. [DOI] [PubMed] [Google Scholar]
  • 148.Thelen A, Scholz A, Weichert W, Wiedenmann B, Neuhaus P, Gessner R, Benckert C, Jonas S. Tumor-associated angiogenesis and lymphangiogenesis correlate with progression of intrahepatic cholangiocarcinoma. Am J Gastroenterol. 2010;105:1123–32. doi: 10.1038/ajg.2009.674. [DOI] [PubMed] [Google Scholar]
  • 149.Yamaguchi R, Yano H, Nakashima O, Akiba J, Nishida N, Kurogi M, Kojiro M. Expression of vascular endothelial growth factor-C in human hepatocellular carcinoma. J Gastroenterol Hepatol. 2006;21:152–60. doi: 10.1111/j.1440-1746.2005.04217.x. [DOI] [PubMed] [Google Scholar]
  • 150.Tampellini M, Sonetto C, Scagliotti GV. Novel anti-angiogenic therapeutic strategies in colorectal cancer. Expert Opin Investig Drugs. 2016:1–14. doi: 10.1517/13543784.2016.1161754. [DOI] [PubMed] [Google Scholar]
  • 151.Kerjaschki D, Regele HM, Moosberger I, Nagy-Bojarski K, Watschinger B, Soleiman A, Birner P, Krieger S, Hovorka A, Silberhumer G, Laakkonen P, Petrova T, Langer B, Raab I. Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates. J Am Soc Nephrol. 2004;15:603–12. doi: 10.1097/01.asn.0000113316.52371.2e. [DOI] [PubMed] [Google Scholar]
  • 152.Stuht S, Gwinner W, Franz I, Schwarz A, Jonigk D, Kreipe H, Kerjaschki D, Haller H, Mengel M. Lymphatic neoangiogenesis in human renal allografts: results from sequential protocol biopsies. Am J Transplant. 2007;7:377–84. doi: 10.1111/j.1600-6143.2006.01638.x. [DOI] [PubMed] [Google Scholar]
  • 153.Vass DG, Hughes J, Marson LP. Restorative and rejection-associated lymphangiogenesis after renal transplantation: friend or foe? Transplantation. 2009;88:1237–9. doi: 10.1097/TP.0b013e3181c1afa7. [DOI] [PubMed] [Google Scholar]
  • 154.Geissler HJ, Dashkevich A, Fischer UM, Fries JW, Kuhn-Regnier F, Addicks K, Mehlhorn U, Bloch W. First year changes of myocardial lymphatic endothelial markers in heart transplant recipients. Eur J Cardiothorac Surg. 2006;29:767–71. doi: 10.1016/j.ejcts.2005.12.024. [DOI] [PubMed] [Google Scholar]
  • 155.Dashkevich A, Heilmann C, Kayser G, Germann M, Beyersdorf F, Passlick B, Geissler HJ. Lymph angiogenesis after lung transplantation and relation to acute organ rejection in humans. Ann Thorac Surg. 2010;90:406–11. doi: 10.1016/j.athoracsur.2010.03.013. [DOI] [PubMed] [Google Scholar]
  • 156.Ishii E, Shimizu A, Kuwahara N, Arai T, Kataoka M, Wakamatsu K, Ishikawa A, Nagasaka S, Fukuda Y. Lymphangiogenesis associated with acute cellular rejection in rat liver transplantation. Transplant Proc. 2010;42:4282–5. doi: 10.1016/j.transproceed.2010.09.081. [DOI] [PubMed] [Google Scholar]
  • 157.Enzan H, Himeno H, Hiroi M, Kiyoku H, Saibara T, Onishi S. Development of hepatic sinusoidal structure with special reference to the Ito cells. Microsc Res Tech. 1997;39:336–49. doi: 10.1002/(SICI)1097-0029(19971115)39:4<336::AID-JEMT4>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 158.Salmi M, Jalkanen S. Cell-surface enzymes in control of leukocyte trafficking. Nat Rev Immunol. 2005;5:760–71. doi: 10.1038/nri1705. [DOI] [PubMed] [Google Scholar]
  • 159.Ikeda Y, Terashima T. Expression of reelin, the gene responsible for the reeler mutation, in embryonic development and adulthood in the mouse. Dev Dyn. 1997;210:157–72. doi: 10.1002/(SICI)1097-0177(199710)210:2<157::AID-AJA8>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 160.Iwakiri Y. The lymphatic system: A new frontier in hepatology. Hepatology. 2016 doi: 10.1002/hep.28650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Favre CJ, Mancuso M, Maas K, McLean JW, Baluk P, McDonald DM. Expression of genes involved in vascular development and angiogenesis in endothelial cells of adult lung. Am J Physiol Heart Circ Physiol. 2003;285:H1917–38. doi: 10.1152/ajpheart.00983.2002. [DOI] [PubMed] [Google Scholar]
  • 162.Kaser-Eichberger A, Schroedl F, Bieler L, Trost A, Bogner B, Runge C, Tempfer H, Zaunmair P, Kreutzer C, Traweger A, Reitsamer HA, Couillard-Despres S. Expression of Lymphatic Markers in the Adult Rat Spinal Cord. Front Cell Neurosci. 2016;10:23. doi: 10.3389/fncel.2016.00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Cho CH, Koh YJ, Han J, Sung HK, Lee HJ, Morisada T, Schwendener Ra, Brekken Ra, Kang G, Oike Y, Choi TS, Suda T, Yoo OJ, Koh GY. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circulation Research. 2007:100. doi: 10.1161/01.RES.0000259564.92792.93. [DOI] [PubMed] [Google Scholar]
  • 164.Zheng M, Kimura S, Nio-Kobayashi J, Takahashi-Iwanaga H, Iwanaga T. Three types of macrophagic cells in the mesentery of mice with special reference to LYVE-1-immunoreactive cells. Biomed Res. 2014;35:37–45. doi: 10.2220/biomedres.35.37. [DOI] [PubMed] [Google Scholar]
  • 165.Xu H, Chen M, Reid DM, Forrester JV. LYVE-1-positive macrophages are present in normal murine eyes. Invest Ophthalmol Vis Sci. 2007;48:2162–71. doi: 10.1167/iovs.06-0783. [DOI] [PubMed] [Google Scholar]
  • 166.Schledzewski K, Falkowski M, Moldenhauer G, Metharom P, Kzhyshkowska J, Ganss R, Demory A, Falkowska-Hansen B, Kurzen H, Ugurel S, Geginat G, Arnold B, Goerdt S. Lymphatic endothelium-specific hyaluronan receptor LYVE-1 is expressed by stabilin-1+, F4/80+, CD11b+ macrophages in malignant tumours and wound healing tissue in vivo and in bone marrow cultures in vitro: implications for the assessment of lymphangiogenesis. J Pathol. 2006;209:67–77. doi: 10.1002/path.1942. [DOI] [PubMed] [Google Scholar]
  • 167.Lua I, Li Y, Zagory JA, Wang KS, French SW, Sevigny J, Asahina K. Characterization of hepatic stellate cells, portal fibroblasts, and mesothelial cells in normal and fibrotic livers. J Hepatol. 2016 doi: 10.1016/j.jhep.2016.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Peters A, Pitcher LA, Sullivan JM, Mitsdoerffer M, Acton SE, Franz B, Wucherpfennig K, Turley S, Carroll MC, Sobel RA, Bettelli E, Kuchroo VK. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity. 2011;35:986–96. doi: 10.1016/j.immuni.2011.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Kamba T, Tam BY, Hashizume H, Haskell A, Sennino B, Mancuso MR, Norberg SM, O’Brien SM, Davis RB, Gowen LC, Anderson KD, Thurston G, Joho S, Springer ML, Kuo CJ, McDonald DM. VEGF-dependent plasticity of fenestrated capillaries in the normal adult microvasculature. Am J Physiol Heart Circ Physiol. 2006;290:H560–76. doi: 10.1152/ajpheart.00133.2005. [DOI] [PubMed] [Google Scholar]
  • 170.Witmer AN, van Blijswijk BC, van Noorden CJ, Vrensen GF, Schlingemann RO. In vivo angiogenic phenotype of endothelial cells and pericytes induced by vascular endothelial growth factor-A. J Histochem Cytochem. 2004;52:39–52. doi: 10.1177/002215540405200105. [DOI] [PubMed] [Google Scholar]
  • 171.Gunn MD, Kyuwa S, Tam C, Kakiuchi T, Matsuzawa A, Williams LT, Nakano H. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J Exp Med. 1999;189:451–60. doi: 10.1084/jem.189.3.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Heydtmann M, Hardie D, Shields PL, Faint J, Buckley CD, Campbell JJ, Salmon M, Adams DH. Detailed analysis of intrahepatic CD8 T cells in the normal and hepatitis C-infected liver reveals differences in specific populations of memory cells with distinct homing phenotypes. J Immunol. 2006;177:729–38. doi: 10.4049/jimmunol.177.1.729. [DOI] [PubMed] [Google Scholar]
  • 173.Grant AJ, Goddard S, Ahmed-Choudhury J, Reynolds G, Jackson DG, Briskin M, Wu L, Hubscher SG, Adams DH. Hepatic expression of secondary lymphoid chemokine (CCL21) promotes the development of portal-associated lymphoid tissue in chronic inflammatory liver disease. Am J Pathol. 2002;160:1445–55. doi: 10.1016/S0002-9440(10)62570-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Lee SJ, Evers S, Roeder D, Parlow AF, Risteli J, Risteli L, Lee YC, Feizi T, Langen H, Nussenzweig MC. Mannose receptor-mediated regulation of serum glycoprotein homeostasis. Science. 2002;295:1898–901. doi: 10.1126/science.1069540. [DOI] [PubMed] [Google Scholar]
  • 175.Takahashi K, Donovan MJ, Rogers RA, Ezekowitz RA. Distribution of murine mannose receptor expression from early embryogenesis through to adulthood. Cell Tissue Res. 1998;292:311–23. doi: 10.1007/s004410051062. [DOI] [PubMed] [Google Scholar]
  • 176.Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Yla-Herttuala S, Alitalo K. Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. Embo j. 2002;21:4593–9. doi: 10.1093/emboj/cdf470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Linehan SA, Martinez-Pomares L, Stahl PD, Gordon S. Mannose receptor and its putative ligands in normal murine lymphoid and nonlymphoid organs: In situ expression of mannose receptor by selected macrophages, endothelial cells, perivascular microglia, and mesangial cells, but not dendritic cells. J Exp Med. 1999;189:1961–72. doi: 10.1084/jem.189.12.1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Gigi-Leitner O, Geiger B. Antigenic interrelationship between the 40-kilodalton cytokeratin polypeptide and desmoplakins. Cell Motil Cytoskeleton. 1986;6:628–39. doi: 10.1002/cm.970060611. [DOI] [PubMed] [Google Scholar]
  • 179.Sawa Y, Shibata K, Braithwaite MW, Suzuki M, Yoshida S. Expression of immunoglobulin superfamily members on the lymphatic endothelium of inflamed human small intestine. Microvasc Res. 1999;57:100–6. doi: 10.1006/mvre.1998.2132. [DOI] [PubMed] [Google Scholar]
  • 180.Cao Y, Chang H, Li L, Cheng RC, Fan XN. Alteration of adhesion molecule expression and cellular polarity in hepatocellular carcinoma. Histopathology. 2007;51:528–38. doi: 10.1111/j.1365-2559.2007.02820.x. [DOI] [PubMed] [Google Scholar]
  • 181.Ebata N, Nodasaka Y, Sawa Y, Yamaoka Y, Makino S, Totsuka Y, Yoshida S. Desmoplakin as a specific marker of lymphatic vessels. Microvasc Res. 2001;61:40–8. doi: 10.1006/mvre.2000.2280. [DOI] [PubMed] [Google Scholar]
  • 182.Hoye AM, Couchman JR, Wewer UM, Fukami K, Yoneda A. The newcomer in the integrin family: integrin alpha9 in biology and cancer. Adv Biol Regul. 2012;52:326–39. doi: 10.1016/j.jbior.2012.03.004. [DOI] [PubMed] [Google Scholar]
  • 183.Halin C, Tobler NE, Vigl B, Brown LF, Detmar M. VEGF-A produced by chronically inflamed tissue induces lymphangiogenesis in draining lymph nodes. Blood. 2007;110:3158–67. doi: 10.1182/blood-2007-01-066811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wuest TR, Carr DJ. VEGF-A expression by HSV-1-infected cells drives corneal lymphangiogenesis. J Exp Med. 2010;207:101–15. doi: 10.1084/jem.20091385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Kubo H, Cao R, Brakenhielm E, Makinen T, Cao Y, Alitalo K. Blockade of vascular endothelial growth factor receptor-3 signaling inhibits fibroblast growth factor-2-induced lymphangiogenesis in mouse cornea. Proc Natl Acad Sci U S A. 2002;99:8868–73. doi: 10.1073/pnas.062040199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Baluk P, Tammela T, Ator E, Lyubynska N, Achen MG, Hicklin DJ, Jeltsch M, Petrova TV, Pytowski B, Stacker SA, Yla-Herttuala S, Jackson DG, Alitalo K, McDonald DM. Pathogenesis of persistent lymphatic vessel hyperplasia in chronic airway inflammation. J Clin Invest. 2005;115:247–57. doi: 10.1172/JCI22037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Kim KE, Koh YJ, Jeon BH, Jang C, Han J, Kataru RP, Schwendener RA, Kim JM, Koh GY. Role of CD11b+ macrophages in intraperitoneal lipopolysaccharide-induced aberrant lymphangiogenesis and lymphatic function in the diaphragm. Am J Pathol. 2009;175:1733–45. doi: 10.2353/ajpath.2009.090133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Yan ZX, Jiang ZH, Liu NF. Angiopoietin-2 promotes inflammatory lymphangiogenesis and its effect can be blocked by the specific inhibitor L1-10. Am J Physiol Heart Circ Physiol. 2012;302:H215–23. doi: 10.1152/ajpheart.00895.2011. [DOI] [PubMed] [Google Scholar]
  • 189.Yuen D, Grimaldo S, Sessa R, Ecoiffier T, Truong T, Huang E, Bernas M, Daley S, Witte M, Chen L. Role of angiopoietin-2 in corneal lymphangiogenesis. Invest Ophthalmol Vis Sci. 2014;55:3320–7. doi: 10.1167/iovs.13-13779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Saito Y, Nakagami H, Morishita R, Takami Y, Kikuchi Y, Hayashi H, Nishikawa T, Tamai K, Azuma N, Sasajima T, Kaneda Y. Transfection of human hepatocyte growth factor gene ameliorates secondary lymphedema via promotion of lymphangiogenesis. Circulation. 2006;114:1177–84. doi: 10.1161/CIRCULATIONAHA.105.602953. [DOI] [PubMed] [Google Scholar]
  • 191.Furtado GC, Marinkovic T, Martin AP, Garin A, Hoch B, Hubner W, Chen BK, Genden E, Skobe M, Lira SA. Lymphotoxin beta receptor signaling is required for inflammatory lymphangiogenesis in the thyroid. Proc Natl Acad Sci U S A. 2007;104:5026–31. doi: 10.1073/pnas.0606697104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Mounzer RH, Svendsen OS, Baluk P, Bergman CM, Padera TP, Wiig H, Jain RK, McDonald DM, Ruddle NH. Lymphotoxin-alpha contributes to lymphangiogenesis. Blood. 2010;116:2173–82. doi: 10.1182/blood-2009-12-256065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Watari K, Nakao S, Fotovati A, Basaki Y, Hosoi F, Bereczky B, Higuchi R, Miyamoto T, Kuwano M, Ono M. Role of macrophages in inflammatory lymphangiogenesis: Enhanced production of vascular endothelial growth factor C and D through NF-kappaB activation. Biochem Biophys Res Commun. 2008;377:826–31. doi: 10.1016/j.bbrc.2008.10.077. [DOI] [PubMed] [Google Scholar]
  • 194.Al-Rawi MA, Watkins G, Mansel RE, Jiang WG. Interleukin 7 upregulates vascular endothelial growth factor D in breast cancer cells and induces lymphangiogenesis in vivo. Br J Surg. 2005;92:305–10. doi: 10.1002/bjs.4832. [DOI] [PubMed] [Google Scholar]
  • 195.Al-Rawi MA, Watkins G, Mansel RE, Jiang WG. The effects of interleukin-7 on the lymphangiogenic properties of human endothelial cells. Int J Oncol. 2005;27:721–30. [PubMed] [Google Scholar]
  • 196.Choi I, Lee YS, Chung HK, Choi D, Ecoiffier T, Lee HN, Kim KE, Lee S, Park EK, Maeng YS, Kim NY, Ladner RD, Petasis NA, Koh CJ, Chen L, Lenz HJ, Hong YK. Interleukin-8 reduces post-surgical lymphedema formation by promoting lymphatic vessel regeneration. Angiogenesis. 2013;16:29–44. doi: 10.1007/s10456-012-9297-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chauhan SK, Jin Y, Goyal S, Lee HS, Fuchsluger TA, Lee HK, Dana R. A novel pro-lymphangiogenic function for Th17/IL-17. Blood. 2011;118:4630–4. doi: 10.1182/blood-2011-01-332049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Hammer T, Tritsaris K, Hubschmann MV, Gibson J, Nisato RE, Pepper MS, Dissing S. IL-20 activates human lymphatic endothelial cells causing cell signalling and tube formation. Microvasc Res. 2009;78:25–32. doi: 10.1016/j.mvr.2009.02.007. [DOI] [PubMed] [Google Scholar]
  • 199.Oka M, Iwata C, Suzuki HI, Kiyono K, Morishita Y, Watabe T, Komuro A, Kano MR, Miyazono K. Inhibition of endogenous TGF-beta signaling enhances lymphangiogenesis. Blood. 2008;111:4571–9. doi: 10.1182/blood-2007-10-120337. [DOI] [PubMed] [Google Scholar]
  • 200.Clavin NW, Avraham T, Fernandez J, Daluvoy SV, Soares MA, Chaudhry A, Mehrara BJ. TGF-beta1 is a negative regulator of lymphatic regeneration during wound repair. Am J Physiol Heart Circ Physiol. 2008;295:H2113–27. doi: 10.1152/ajpheart.00879.2008. [DOI] [PubMed] [Google Scholar]
  • 201.Avraham T, Daluvoy S, Zampell J, Yan A, Haviv YS, Rockson SG, Mehrara BJ. Blockade of transforming growth factor-beta1 accelerates lymphatic regeneration during wound repair. Am J Pathol. 2010;177:3202–14. doi: 10.2353/ajpath.2010.100594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Dunworth WP, Cardona-Costa J, Bozkulak EC, Kim JD, Meadows S, Fischer JC, Wang Y, Cleaver O, Qyang Y, Ober EA, Jin SW. Bone morphogenetic protein 2 signaling negatively modulates lymphatic development in vertebrate embryos. Circ Res. 2014;114:56–66. doi: 10.1161/CIRCRESAHA.114.302452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Shao X, Liu C. Influence of IFN- alpha and IFN- gamma on lymphangiogenesis. J Interferon Cytokine Res. 2006;26:568–74. doi: 10.1089/jir.2006.26.568. [DOI] [PubMed] [Google Scholar]
  • 204.Shin K, Kataru RP, Park HJ, Kwon BI, Kim TW, Hong YK, Lee SH. TH2 cells and their cytokines regulate formation and function of lymphatic vessels. Nat Commun. 2015;6:6196. doi: 10.1038/ncomms7196. [DOI] [PubMed] [Google Scholar]
  • 205.Nielsen SR, Hammer T, Gibson J, Pepper MS, Nisato RE, Dissing S, Tritsaris K. IL-27 inhibits lymphatic endothelial cell proliferation by STAT1-regulated gene expression. Microcirculation. 2013;20:555–64. doi: 10.1111/micc.12055. [DOI] [PubMed] [Google Scholar]
  • 206.Heinz M, Niederleithner HL, Puujalka E, Soler-Cardona A, Grusch M, Pehamberger H, Loewe R, Petzelbauer P. Activin A is anti-lymphangiogenic in a melanoma mouse model. J Invest Dermatol. 2015;135:212–21. doi: 10.1038/jid.2014.328. [DOI] [PubMed] [Google Scholar]

RESOURCES