• Ei tuloksia

Cardiac Lymphatics - A New Avenue for Therapeutics?

N/A
N/A
Info
Lataa
Protected

Academic year: 2022

Jaa "Cardiac Lymphatics - A New Avenue for Therapeutics?"

Copied!
25
0
0

Kokoteksti

(1)

UEF//eRepository

DSpace https://erepo.uef.fi

Rinnakkaistallenteet Terveystieteiden tiedekunta

2017

Cardiac Lymphatics - A New Avenue for Therapeutics?

Vuorio Taina

Elsevier BV

Tieteelliset aikakauslehtiartikkelit

© Elsevier Ltd

CC BY-NC-ND https://creativecommons.org/licenses/by-nc-nd/4.0/

http://dx.doi.org/10.1016/j.tem.2016.12.002

https://erepo.uef.fi/handle/123456789/7914

Downloaded from University of Eastern Finland's eRepository

(2)

Cardiac lymphatics, a new avenue for therapeutics?

1 2

Taina Vuorio1, Annakaisa Tirronen1, Seppo Ylä-Herttuala1,2 3

4

1Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular 5

Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; 2 Kuopio 6

University Hospital, Science Service Center and Gene Therapy Unit, P.O. Box 1777, FIN-70211 7

Kuopio, Finland 8

http://www.uef.fi/en/web/aivi/seppo-ylaherttuala-group 9

10

Correspondence to S.Y-H. Tel. +358 403552075, seppo.ylaherttuala@uef.fi.

11 12 13

Keywords 14

atherosclerosis, cardiac lymphatic vessels, myocardial infarction, reverse cholesterol transport, 15

therapeutics 16

17 18 19 20 21 22

(3)

Abstract 23

Progress in lymphatic vessel biology and novel imaging techniques have established the importance 24

of lymphatic vasculature as part of the cardiovascular system. Lymphatic vessel network regulates 25

many physiological processes important in the heart such as fluid balance, transport of extravasated 26

proteins and trafficking of immune cells. Therefore, lymphangiogenic therapy could be beneficial in 27

the treatment of cardiovascular diseases, for example by improving reverse cholesterol transport from 28

atherosclerotic lesions or resolving edema and fibrosis after myocardial infarction. In this review, we 29

first describe recent findings in the development and function of cardiac lymphatic vessels and 30

subsequently focus on the prospects of pro- and anti-lymphangiogenic therapies in cardiovascular 31

diseases.

32 33 34

(4)

Lymphatic vessels are novel players in cardiovascular health and disease 35

36

For decades the anatomy and function of cardiac lymphatic system was analyzed with dye injection 37

techniques in large animal models [1] and the role of cardiac lymphatics in normal physiology and 38

pathological conditions remained understudied. The identification of specific lymphatic endothelial 39

cell (LEC) (see Glossary) regulators and markers, such as Prospero Homeobox 1 (PROX1) [2], 40

Vascular Endothelial Growth Factor C (VEGF-C) [3], Vascular Endothelial Growth Factor 41

Receptor 3 (VEGFR-3) [4], Podoplanin (PDPN) [5] and Lymphatic vessel endothelial hyaluronan 42

receptor 1 (LYVE1) [6] has brought several new transgenic mouse models and sophisticated confocal 43

and multiphoton imaging methods for lymphatic research [7].

44 45

The lymphatic system is required for the maintenance of body fluid balance, chylomicron transport 46

from the intestine and immune cell transport and surveillance. The lymphatic network is composed of 47

blind-ended, thin-walled lymphatic capillaries that take up extra interstitial fluid, lymphatic collector 48

vessels dedicated to the transport of the fluid, andlymph nodes that are responsible for immune cell 49

responses. LECs are loosely connected with button-like junctions that allow the entry of fluid, 50

proteins and leukocytes into the lymphatic capillaries. Lymph flows from capillaries into the valve- 51

containing lymphatic collectors and through the thoracic duct and right lymphatic trunk into venous 52

circulation [8].

53 54

As in other parts of the body, the heart has an extensive lymphatic network that regulates and 55

maintains the correct fluid balance. The obstruction of the cardiac lymph flow in the healthy heart of 56

an experimental animal can lead to severe cardiac edema, left ventricular dysfunction and 57

hemorrhages (reviewed in [1]). On the other hand, the development of new lymphatics 58

(lymphangiogenesis) may be favorable in delaying atherosclerotic plaque formation within coronary 59

arteries [9]. In addition, therapeutically induced lymphangiogenesis has shown to be beneficial during 60

the healing process after myocardial infarction (MI) as it reduces fluid retention and improves 61

inflammatory cell clearance in the cardiac tissue [10, 11]. In this review, we will introduce the 62

development and regulation of cardiac lymphatics and subsequently focus on therapeutic applications 63

for cardiovascular pathologies 64

65 66

Regulators of lymphangiogenesis 67

68

Lymphatic vessels network is formed during embryogenesis and lymphangiogenesis is also activated 69

during pathological states, such as inflammation, tumor formation and lymphedema [12]. After more 70

than a decade of debate on the origin of LECs, in 2007 Srinivasan et al. [13] confirmed that LECs 71

(5)

originate primarily from embryonic veins. They used Tie2-Cre lineage tracing, a method that utilizes 72

inducible genetical labeling of Tie2-expressing endothelial cells that can be later traced with 73

tamoxifen induction. Lymphangiogenesis starts at embryonic day 10 (E10.0) in mice [2, 14] and 74

during weeks 6-7 in human gestation when PROX1 expression is induced in a subpopulation of 75

endothelial cells in common cardinal vein, intersomitic veins and superficial venous plexus [15].

76

However, lymphatics in specific parts of the body, such as lumbar and dorsal skin regions, cannot be 77

traced to originate from Tie2 venous endothelial source [16]. In addition, mesenteric lymphatics have 78

shown to arise from a specific pool of naïve hematopoietic cells [17]. A study by Klotz et al. [10]

79

described the yolk sac haemogenic endothelium as an alternative source of cardiac LECs in mice.

80

This finding was recently challenged by Ulvmar et al. [18] proposing that Pdgfrb-Cre lineage tracing, 81

a method used by Klotz et al. labels the yolk sac haemogenic endothelium incompletely. Thus, the 82

most common origin of LECs is the embryonic veins but the exact identity of the cardiac LEC 83

progenitor remains to be determined. The function of the key factors regulating lymphangiogenesis 84

are described in the next chapters.

85 86

PROX1 87

Homeobox transcription factor PROX1 is a key molecule in the differentiation of blood and lymphatic 88

endothelial cells as it determines the specification of LECs from endothelial precursor cells during 89

embryogenesis [2]. Transcription factors SRY-Box 18 (SOX18) [19] and COUP transcription factor 2 90

(COUP-TFII) [20] are required for PROX1 activation. The expression of SOX18 is restricted to only 91

certain cells in venous endothelium and the expression is maintained in precursor LECs migrating 92

towards lymph sacs [19]. It has been speculated that SOX18 has an essential role in the early 93

induction of PROX1 but it is not sufficient for maintaining PROX1 expression later [21]. In contrast 94

to SOX18, COUP-TFII is present in all venous endothelial cells in the embryo, and it forms a 95

heterodimer with PROX1 in the cells determined for LEC specification [22]. PROX1 and VEGFR-3 96

form a positive feedback loop: VEGFR-3 gene is a direct target of PROX1 and in turn, VEGFR-3 97

signaling is essential in PROX1 expression in LECs [23].

98 99

VEGF-C and VEGFR-3 100

VEGFR-3 expression induces the budding and migration of LECs towards the mesenchyme that 101

expresses VEGF-C, the primary ligand for VEGFR-3 [24, 25]. Budding LECs coalesce and form 102

lymph sacs, which represent the first primitive lymphatic structures [14]. Both in embryos and adults, 103

VEGF-C–VEGFR-3 signaling is crucial for LEC proliferation, migration and survival [24, 25].

104

However, another VEGFR-3 ligand, VEGF-D, is not required for lymphatic vessel development 105

during embryogenesis [26]. Collagen And Calcium Binding EGF Domains 1 (CCBE1) is essential for 106

sprouting of the primitive LECs from the cardinal vein. It is highly expressed near developing 107

lymphatic vessels and particularly in the developing heart [27]. CCBE1 has been shown to enhance 108

(6)

the cleavage of pro-VEGF-C to its active form by ADAM Metallopeptidase With Thrombospondin 109

Type 1 Motif 3 (ADAMTS3) metalloproteinase [28]. In a recent study, Bui et al. [29] confirmed that a 110

complex of CCBE1-ADAMTS3 is required for proper proteolytic activation of VEGF-C but not 111

VEGF-D. It was speculated that VEGF-C is required for the growth of developing lymphatics 112

whereas VEGF-D serves as a growth factor for reactive, local lymphatic growth during inflammatory 113

reactions in adults [29]. In addition to CCBE1, neuropilin 2 (NRP2) can enhance VEGFR-3 signaling 114

by interacting with VEGFR-3 and thereby increasing the affinity of LECs towards VEGF-C [30].

115 116

Podoplanin 117

The expression of a transmembrane O-glycoprotein podoplanin (PDPN) distinguishes endothelial 118

cells inside the embryonic veins from fully budded LECs [31]. Studies in Pdpn-/- mice have suggested 119

that the expression of PDPN is required for platelet activation and aggregation by binding to a platelet 120

factor C-type lectin-like receptor 2 (CLEC-2). Platelets without PDPN function cannot aggregate and 121

therefore PDPN knockout embryos have incomplete lymphovenous separation and blood-filled 122

lymphatics [32, 33]. PDPN expression is sustained in mature LECs and it is widely used as a LEC 123

marker [5].

124 125

FOXC2 126

Forkhead Box C2 (FOXC2) is required for maturation and maintenance of collecting lymphatic 127

vessels and especially lymphatic valves [34]. In addition, FOXC2 sustains LEC quiescence and 128

stability by maintaining the intercellular junctions and cytoskeleton organization [35]. FOXC2 has 129

been shown to regulate Ras/ERK signaling pathway in LECs along with VEGFR-3. Therefore, it has 130

been speculated that FOXC2 fine-tunes the functions of VEGFR-3 during lymphangiogenesis [36].

131 132 133

Development, anatomy and function of the cardiac lymphatic system 134

135

In mice, cardiac lymphatic vessel formation starts at E11.0-12.0, a few days after the development of 136

coronary blood vessels. VEGFR-3 and PROX1 positive LECs migrate from extracardiac tissues 137

towards the outflow tract on the ventral surface of the heart. At E14.5, lymphatic vessels sprout from 138

the sinus venosus towards the ventricular surface [10]. During embryonic days E15.0-E18.0, LECs 139

migrate from the base of the heart towards the apex, following the coronary vasculature forming the 140

main precollector lymphatics [37]. In addition, cells that express macrophage marker F4-80 have 141

been shown to incorporate to lymphatic vessel walls during prenatal development [38]. After birth, 142

lymphangiogenesis continues and the lymphatic network is expanded from the subepicardium towards 143

the myocardium [37]. In mice, the lymphatic vasculature is fully developed by post-natal day 15 [10].

144

The development of cardiac lymphatic vessels is described inFigure 1.

145

(7)

In the adult heart, the cardiac lymphatic system forms a network of capillaries and precollecting 146

lymphatic vessels that are located most abundantly in the ventricles. The human left ventricular wall 147

possesses approximately 30 lymphatic vessels/mm2, significantly less than blood capillaries [39, 40].

148

There is some species-specific variation in the anatomy of cardiac lymphatics. For example, the 149

majority of lymphatics in rabbit and mouse hearts are located in the subepicardium and outer 150

myocardium, whereas in humans lymphatic capillaries can be visualized evenly in the myocardial, 151

subepicardial, and subendocardial areas (reviewed by Ratajska [40]). Cardiac lymphatic flow begins 152

from the subendocardium, runs through the myocardium and enters into the capillary lymphatic 153

plexus in the epicardium [41]. The diameter of the capillaries varies, depending on the species and 154

study, from 20 µm up to 400 µm [40]. Lymphatic capillaries converge into large valve-containing 155

collecting lymphatics that run along the left conal and left cardiac veins, and finally drain into 156

subaortic and paratracheal lymph nodes [37]. All cardiac valves also possess lymphatics, that are 157

mainly located in the basal part but also some are scattered peripherally [39].

158 159

Many congenital lymphatic disorders cause lymphatic dysfunction and lymphedema primarily in the 160

legs and feet of the individual but not in the heart, such as Milroy’s disease, associated with missense 161

mutations in VEGFR-3 tyrosine kinase domain [42] and lymphedema-distichiasis syndrome, caused 162

by mutations in the FOXC2 gene [43]. However, Noonan syndrome, affected by the upregulated 163

RAS-MAPK signaling, is associated with several signs of lymphatic dysfunction such as 164

lymphedema, lymphatic dysplasias, chylous reflux and intestinal lymphangiectasis, but also 165

congenital heart defects characterized by pulmonary valve stenosis, atrial septal defects and 166

hypertrophic cardiomyopathy [44]. Cardiac lymphatic anatomy or function has not been recorded in 167

these patients, but lymphatic dysfunction might affect the progression of heart defects as the 168

development of lymphatics is abnormal.

169 170

In addition to hereditable lymphatic dysfunctions, lymphatic vessels play a role in several heart- 171

related conditions and might have a therapeutical potential. These will be discussed in the further 172

sections of the review.

173 174 175

Lymphatic vessels in heart diseases 176

177

Heart diseases affect nearly half of the population in Western societies [45]. They are primarily 178

caused byatherosclerosis, a chronic inflammatory disease of the arteries that can cause blockage of 179

the blood flow leading to ischemia and MI. In the following sections we review the current knowledge 180

of lymphatics in the cardiac pathologies and highlight the therapeutic potential of lymphangiogenesis 181

(Table 1).

182

(8)

Atherosclerosis 183

Atherosclerosis is characterized by the formation of arterial fatty streaks and plaques loaded with 184

macrophages containing cholesterol (foam cells). A continuous recruitment of monocytes and 185

differentiation into foamy macrophages drives disease progression [46]. The so-called vulnerable 186

plaques, are especially dangerous because they can break and cause acute occlusion in large arteries in 187

the heart and other organs [47]. An attractive treatment option for atherosclerosis is to reduce 188

macrophage burden and decrease cholesterol build up within artery wall by way of a reverse 189

cholesterol transport (RCT).

190 191

RCT is a pathway responsible for cholesterol mobilization on high density lipoprotein (HDL) 192

particles from extravascular tissues such as artery wall and muscle to the liver for excretion [48]. RCT 193

is a key player in maintaining peripheral and total body cholesterol homeostasis and it could lead to 194

regression of atherosclerosis by reducing the cholesterol content within the plaques. A strong inverse 195

correlation has been established between plasma HDL cholesterol and the risk for cardiovascular 196

disease [49]. During RCT, apolipoprotein A1 containing pre- HDL removes cholesterol from 197

macrophages through the ABCA1 and ABCG1 transporters, and then travels through the bloodstream 198

to the liver for biliary excretion [48]. The exact route and mechanism for HDL RCT from peripheral 199

tissue to the circulation still remains unclear.

200 201

Recent studies suggest that lymphatic vessels could play an important role in RCT by transporting 202

HDL from interstitial tissues to the bloodstream [9, 50]. It has been shown in mice that blocking 203

lymphatic growth within the aortic wall leads to greater cholesterol retention in the aortae [9].

204

Furthermore, a surgical ablation of lymphatic vessels in the mouse skin blocked RCT without 205

impairing cholesterol efflux from macrophages indicating the importance of lymphatics in HDL 206

transport [9]. Soluble decoy VEGFR-3×Ldlr /ApoB100/100 mice and Chy×Ldlr /ApoB100/100 mice 207

both display inhibited VEGF-C – VEGFR-3 signaling thus leading to impaired lymphangiogenesis.

208

Studies with these mouse models further confirmed that lymphatic insufficiency leads to accelerated 209

atherosclerotic lesion development highlighting the importance of lymphatic vessel function in 210

cholesterol metabolism [51].

211 212

Additionally, it has been shown that restoration of lymphatic function in hypercholesterolemic mice 213

improves lipid clearance. Hypercholesterolemic ApoE mice carrying excess cholesterol in VLDL 214

and chylomicron remnant fraction displayed structural alterations in lymphatics and lower expression 215

of VEGF-C and FOXC2, which both are important factors maintaining lymphatic vasculature.

216

However, treatment of ApoE mice with a local injection of recombinant VEGF-C growth factor 217

restored lymphatic function and reduced the accumulation of cholesterol in the skin and improved 218

(9)

RCT, further emphasizing the potential of lymphangiogenic therapy as a tool for cholesterol clearance 219

[50].

220 221

Proprotein convertase subtilisin/kexin type 9 (PCSK9) down-regulates of LDL receptor (LDLR) by 222

binding the receptor and causing its lysosomal degradation in cells. A study employing Pcsk9 mice 223

suggests that LDL receptor (LDLR) could also play a role in lymphatic dysfunction. Atherosclerosis 224

protected Pcsk9 mice exhibited improved collecting lymphatic vessel function combined with 225

enhanced expression of LDLR on LECs whereas mice deficient of LDLR (Ldlr /hApoB100/100) were 226

shown to have defects in collecting lymphatic vessels before the atherosclerotic lesion formation in 227

LDLR-dependent manner. Treatment of Ldlr /hApoB100/100 mice with selective VEGFR-3 agonist, 228

VEGF-C152S mutant, significantly increased lymphatic vasculature in the adventitia of the aortic sinus 229

and enhanced lymphatic cellular transport. However, there is no data whether this could alter the risk 230

for atherosclerosis [52].

231 232

Most of the studies have utilized mouse models but in 2011 Kholova et al. [39] demonstrated that 233

lymphatics are also present in human coronary arteries as well as in atherosclerotic plaques.

234

Lymphatics were found in intima, media and adventitia of the arterial wall, and moreover, increased 235

medial lymphangiogenesis was present in progressive atherosclerotic lesions, which could be a natural 236

response to resolve increased inflammation and cholesterol build up [39]. Interestingly, it has been 237

shown that ApoA1 and HDL concentration in human artery wall is as high as in the plasma, thus 238

emphasizing a potential role of arterial lymphatics in carrying excess cholesterol out from 239

atherosclerotic plaques [53].

240 241

As mentioned earlier, the mechanism for HDL transport from peripheral tissue to the circulation has 242

remained unknown. Recently, it was suggested that Scavenger receptor class B member 1 (SR-BI) is 243

expressed in the lymphatic endothelium where it mediates the internalization and transport of HDL.

244

Furthermore, downregulation of SR-BI by small interfering RNAs resulted in 80% inhibition of HDL 245

uptake by LECsin vitro. It was also shown that treatment of wild type mice with an SR-BI blocking 246

antibody inhibits the transport of HDL via lymphatics by 75%. This is the first study demonstrating 247

that lymphatics can actively transport HDL cholesterol via SR-BI dependent mechanism [50].

248 249

All aforementioned studies show that functional lymphatic vasculature is responsible for cholesterol 250

removal from the artery wall and therefore, influences the atherosclerotic plaque formation.

251

Additionally, it has been proposed that HDL enters the lymphatic vasculature via SR-BI dependent 252

mechanism before reaching the bloodstream. These results suggest that therapies aimed at reducing 253

atherosclerosis by way of induced lymphangiogenesis and enhanced HDL transport may be beneficial.

254

Thus far, clinical trials aiming to increase HDL levels have failed to reduce the risk of recurrent 255

(10)

cardiovascular events [54, 55]. This accentuates that simply increasing the HDL levels is not a 256

sufficient treatment, thus targeting the mechanisms behind HDL transport could provide a better 257

therapeutic outcome.

258 259

Myocardial infarction 260

As in other parts of the body, cardiac lymphatic vessels are required for the maintenance of fluid 261

balance and immune surveillance, both important factors during and after MI. Briefly, MI is primarily 262

a manifestation of coronary artery disease where a part of the heart muscle is damaged by a blockage 263

in blood flow. In most cases, the rupture of the atherosclerotic plaques in the coronary artery wall lead 264

to a clotting cascade and complete blockage of the arterial lumen. If this ischemic condition is 265

prolonged, cardiac cells within the region die leading to the formation of an infarction scar [56].

266

Recruited inflammatory cells clear the affected area from dead cells and matrix debris and stimulate 267

regenerative processes. However, inflammatory reaction can also cause deleterious cardiac 268

remodeling [57].

269 270

Only a few studies have addressed the role of lymphatic vessels in MI. Recently, it was shown that 271

adverse remodeling of epicardial collector lymphatics in the infarcted area causes reduced lymphatic 272

flow and persistent edema [11]. Cardiac edema has been shown to induce myocardial stiffness, 273

fibrosis and LV dysfunction by increasing the amount of collagens in the ventricle walls [58]. As a 274

response to the fluid accumulation into the myocardium, lymphangiogenesis has been observed after 275

experimental LAD ligation in mice [10] and rats [11] and also in post-mortem human MI samples 276

[59].

277 278

Therapeutical lymphangiogenesis appears to be beneficial for post-MI healing. Klotz et al. showed 279

that left ventricular ejection fraction was significantly improved 14 and 21 days after MI, when 280

VEGFR-3 specific recombinant protein VEGF-C156S therapy was utilized [10]. Henri et al. [11]

281

studied the effect of microparticles carrying VEGF-CC152Sprotein, another selective VEGFR-3 ligand, 282

in an MI rat model. While, no effect was seen in infarct scar size, cardiac hypertrophy was decreased.

283

In addition, precollector remodeling was attenuated and fluid balance was improved. Both VEGF-C 284

and VEGF-D therapy has been shown to promote blood flow and healing in the rabbit and mouse hind 285

limb after ischemia, but the role of lymphangiogenesis in these studies remains questionable [60-63].

286

In addition, gene therapy trial in humans showed beneficial effects of VEGF-C therapy on angina 287

score [64, 65]. Unfortunately, lymphangiogenesis was not analyzed in these studies.

288 289

Diseases of heart valves 290

Lymphatic vessels have been shown to be present both in normal and diseased human heart valves.

291

As compared to blood vessels, lymphatic vasculature is proportionately more prominent in valves 292

(11)

than in the other areas of the heart [39]. A few publications have reported that increased 293

lymphangiogenesis is observed during valve pathologies [39, 66, 67]. Human heart valve endocarditis 294

is characterized by inflammation and abnormal growth, as well as inflammation-associated 295

lymphangiogenesis. Lymphatic vasculature within the diseased heart valves was shown to be 296

increased, both in vessel size and density [67]. However, it is not clear whether lymphangiogenesis 297

within the valves is enough to resolve the inflammation and would lymphangiogenic therapy be a 298

sufficient treatment.

299 300

Valvular stenosis involves narrowing of the valve caused by calcification and accumulation of lipids 301

and inflammatory cells [66]. Syvaranta et al. [66] showed that the lymphatic vasculature is present in 302

human stenotic aortic valves and lymphatic capillaries are associated with areas rich in inflammatory 303

cells and neovascularization. Furthermore, VEGF-D and VEGFR-3 mRNA levels in the stenotic 304

aortic valves were upregulated and conceivably are responsible for the increased lymphangiogenesis.

305

Lymphangiogenesis may provide a pathway for lipid and inflammatory cell clearance leading to 306

reduced valvular thickening.

307 308

Heart transplantation 309

The lymphangiogenesis observed after MI appears to be beneficial in resolving inflammation by 310

increasing the clearance of fluid and immune cells as well as inflammatory mediators from the injured 311

heart. However, sustained lymphangiogenesis may also provoke unwanted adverse effects by 312

supporting the transport of immune cells and antigens. Lymphatic vasculature is an important bridge 313

between the innate and adaptive immunity, and in the case of heart transplantation, it may evoke 314

adaptive immunity with serious consequences [68]. Activated antigen-presenting cells such as 315

dendritic cells migrate to the recipient´s draining lymph nodes to trigger adaptive immune responses 316

by presenting foreign antigens to T cells, thus aggravating inflammation. Because increased 317

lymphangiogenesis enables antigen-presenting cell migration, lymphangiogenesis can serve as a 318

increased exposure of lymph nodes target to modulate adverse immune reactions [68].

319 320

A recent study demonstrated that cardiac allograft ischemia-reperfusion injury in the rat heart 321

increased graft VEGF-C expression and lymphatic vessel activation. Treatment with a VEGFR-3 322

inhibitor led to reduced lymphatic vessel activation and dendritic cell maturation, subsequently 323

reducing chronic inflammation and resulting in attenuated acute and chronic rejection. Furthermore, 324

mouse studies using transplanted hearts carrying a LEC specific VEGFR-3 deletion confirmed the 325

previous results that VEGFR-3 inhibition leads to prolonged cardiac allograft survival [69].

326 327

Another method to prolong allograft heart transplant survival is to destroy the bridge between innate 328

and adaptive immunity. Azzi et al. (2016) injected microparticles containing T cell proliferation 329

(12)

inhibitors directly to the transplanted mouse heart. These microparticles mimic lymphocyte migration 330

to the lymph nodes and therefore target the activation of the adaptive immunity. Microparticles 331

delivered to draining lymph nodes successfully prolonged heart allograft survival [70].

332 333

From mice to humans?

334 335

Most of the successful cardiac studies within the lymphatic field have utilized rodent models (see 336

Table 1), which leads us to question whether the methods and results from the small animal studies 337

can be translated to complex and chronic human diseases. To resolve this question, the next important 338

step is large animal studies. There are only a few publications utilizing pig models, which demonstrate 339

that lymphedema can be treated with local injections of adenoviruses encoding lymphangiogenic 340

VEGF-C [71-73], VEGF-C156S [72] or VEGF-D [73]. The same treatment strategies could be 341

employed to treat cardiac pathologies in large animal models. There is an ongoing clinical trial for 342

enhancing cardiac angiogenesis, which emphasizes that the methods to treat MI patients are already 343

available [74]. The NOGA electroanatomical mapping and injection catheter provides a tool for 344

lymphangiogenic therapy by enabling to detect and target the borderline areas of the infarction scar 345

and to deliver the intramyocardial treatment simultaneously [74, 75]. Clearly, the tools to move on to 346

large animal studies and even to clinical trials in cardiac lymphatic field in the near future are readily 347

available.

348 349

The activation of lymphangiogenesis may result in increased exposure of lymph nodes to 350

inflammatory mediators and promote the development of metastasis (reviewed in [68]). Therefore, the 351

safety and efficacy of the lymphangiogenic therapy need to be carefully evaluated within the context 352

of each disorders and the desired therapeutical goals. Most current studies have used recombinant 353

proteins or plasmids for treatments but these generally suffer from short duration and low efficacy.

354

Another option for gene delivery would be viral vectors which efficiently carry and transduce the 355

therapeutic factor into the target cells and tissues [76]. Also, biodegradable microparticles have been 356

utilized as carriers of therapeutic factors into lymph nodes [70] and myocardial wall [11, 70]. Most 357

commonly used pro-lymphangiogenic factors are VEGFR-3 ligands VEGF-C and VEGF-D and their 358

splice variants. Other options, such as angiopoietins and fibroblast growth factors and their 359

combinations require further evaluation in this context [12, 77].

360 361

Concluding Remarks and Future Perspectives 362

363

After years of ignorance cardiac lymphatic system has become an active target for research and novel 364

findings are published at increasing pace. Recent advances in the lymphatic field have provided new 365

insights in the treatment of cardiovascular diseases (See Outstanding Questions box and Figure 2, Key 366

(13)

figure). It has been shown that the lymphatic vessels are present around atherosclerotic lesions and 367

provide an important pathway for cholesterol transportation. Therefore, novel lymphangiogenic 368

therapies could accelerate RCT leading to inhibition or regression of atherosclerosis.

369

Lymphangiogenic therapy has also been successfully utilized to resolve edema formation, 370

inflammatory cell accumulation and fibrosis during MI in mice [10, 11]. On the other hand, anti- 371

lymphangiogenic therapy could be beneficial in preventing undesired inflammation in heart 372

transplants.

373 374

In addition, modulation of lymphatic function could be beneficial in treating lipid-related diseases, 375

such as weight-gain and hypercholesterolemia. One of the first reports describing the connection 376

between lymphatics and lipids was in 2005, when Harvey et al. [78] showed that lymphatic 377

dysfunction in Prox1+/- mice caused leakage of lymph into surrounding tissues and resulted in adult- 378

onset obesity. Fat accumulation has also been observed in humans with surgical ablation of lymph 379

nodes [79] but not in other mouse models of lymphatic deficiency [8]. Evidence from the RCT studies 380

indicate that lymphatics are also required for lipoprotein transport and metabolism. It is well 381

established that lymphatics mediate chylomicron transport from intestine into bloodstream, but the 382

function of lymphatics in regulating lipid and lipoprotein uptake into the lacteals has not yet been 383

characterized. Further, the question still remains if lymphatics participate in the trafficking and 384

regulation of endogenous lipoproteins, namely VLDL and atherogenic LDL. There are many open 385

questions in the relationship of lymphatics and lipid metabolism and when clarified, new avenues 386

might open for the treatment of lipid-related diseases, primary causes of many heart pathologies.

387 388

Acknowledgements: This work was supported by Finnish Academy Center of Excellence in 389

Cardiovascular and Metabolic Diseases, Erkko Foundation and Urho Känkänen Foundation.

390 391 392

(14)

393

r: recombinant, p: plasmid, Ad: adenoviral, ip: intraperitoneal 394

395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411

Table 1. Modulation of lymphangiogenesis in cardiovascular diseases

Model Species Treatment Delivery Outcome Ref

Lipoprotein

transport mouse rVEGF-C skin improved RCT [50]

Coronary artery disease

human pVEGF-2

(i.e pVEGF-C) endocardium

improved symptoms of

angina

[64]

Myocardial infarction

mouse rVEGF-C systemic (ip) improved cardiac

function [10]

rat microparticles carrying

VEGF-CC152S myocardium reduced edema,

reduced fibrosis [11]

pig AdVEGF-C myocardium prevention of MI

progression [80]

Peripheral

ischemia rabbit pVEGF-C and

rVEGF-C

angioblasty balloon and intra-arterial

improved flow [60]

Intact muscle

mouse AdVEGF-D hind limb angiogenesis,

lymphangiogenesis [61]

rabbit AdVEGF-D,

AdVEGF-C hind limb lymphangiogenesis [62]

Cardiac

allograft rat VEGF-C/D trap,

VEGFR-3 antibody intra-arterial improved allograft

survival [69]

(15)

Figure Legends 412

Figure 1. Development of cardiac lymphatic vessels 413

(A) Cardiac lymphatic endothelial cells (LECs) originate from common cardinal vein (CCV) and the 414

yolk sac hemogenic endothelium. (B) PROX1 expression is activated by COUPTF-II and SOX18 in a 415

subset of endothelial cells that are then designated for LEC specification. (C) PROX1 activates 416

VEGFR-3 expression and initiates budding of LECs from CCV. (D) LECs migrate towards the 417

systemic venous sinus along the VEGF-C gradient. NRP2 and a complex of CCBE1 and ADAMTS3 418

enhance VEGF-C signaling through VEGFR-3. (E) LECs enter the heart by E12.5 and (F) begin to 419

form the first primitive lymphatic capillaries. (G) Lymphatic network expands from base towards the 420

apex following the coronary veins. (H) By P15, cardiac lymphatic network is fully developed. Panels 421

E-H adapted from Klotz et al. [10].

422 423 424

Figure 2. Key figure. Potential lymphangiogenic therapies in cardiac diseases 425

A schematic diagram illustrating the role of lymphangiogenesis in atherosclerosis and MI. (A) 426

Atherosclerosis is characterized by lipid and inflammatory cell build up within arterial walls. (B) 427

Lymphangiogenic therapy could enhance RCT leading to greater lipid clearance from atherosclerotic 428

plaques and regression of atherosclerosis through accelerated HDL turnover. HDL removes 429

cholesterol from macrophages through ABCA1 and ABCG1 and exits via SR-BI to the adventitial 430

lymphatic vessels to the blood stream. (C) Atherosclerotic plaque rupture leads to coronary artery 431

occlusion and MI. MI is followed by adverse remodeling of epicardial collector lymphatics and 432

subsequently edema. Additionally, severe inflammation and fibrosis is developed. (D) Therapeutic 433

lymphangiogenesis after MI increases the lymph flow and resolves inflammation leading to improved 434

cardiac function and healing of MI.

435 436 437 438 439 440 441 442 443 444 445 446 447 448

(16)

Glossary box 449

Atherosclerosis: A progressive disease characterized by accumulation of cholesterol, extracellural 450

matrix, fibrosis, calcium deposits and inflammatory cells within the arterial wall.

451

Edema:The abnormal accumulation of fluid in the interstitium leading to tissue swelling and pain.

452

High density lipoprotein (HDL): A small, ApoA-rich lipoprotein responsible for reverse cholesterol 453

transport of cholesterol from extrahepatic tissues to liver.

454

Lymphangiogenesis:The formation of new lymphatic vessels from pre-existing lymphatic vessels 455

Lymphatic endothelial cells (LECs):specialized form of epithelium that lines the lymphatic vessels.

456

Lymph node: An organ linked to lymphatic vessels. It is full of immune cells that recognize foreign 457

particles and remove bacteria, viruses, toxins as well as cancer cells from the body.

458

Macrophage: A white blood cell that engulfs and digests microbes, cellular debris and any foreign 459

substances. Also acts as an antigen-presenting cell to activate immune responses.

460

Myocardial infarction (MI): Cardiac muscle damage and/or necrosis due to an occlusion of a 461

coronary artery caused by atherosclerotic plaques.

462

Reverse cholesterol transport (RCT): Movement of cholesterol from peripheral tissues back to the 463

liver via plasma lipoproteins.

464

Vascular endothelial growth factor 3 (VEGFR-3): A tyrosine kinase receptor that binds vascular 465

endothelial growth factors C and D. It is essential for the development and maintenance of lymphatic 466

vessels.

467 468 469 470

Trends Box 471

Cardiac lymphatic endothelial cells originate from multiple sources. In the heart, they form a 472

network of capillary lymphatic vessels and larger collector lymphatics that drain fluid, 473

macromolecules and inflammatory cells.

474

The role of lymphatic vessels in chylomicron metabolism is well-established and lymphatics 475

are also required for reverse cholesterol transport. Therefore, lymphatics may play even a 476

more significant role in lipid and lipoprotein metabolism than previously thought.

477

Lymphangiogenic therapy may become a useful option for the treatment of cardiovascular 478

diseases, such as atherosclerosis and myocardial ischemia. In addition, anti-lymphangiogenic 479

therapy could be utilized to resolve inflammatory reaction in cardiac allografts.

480 481 482 483

(17)

Outstanding Questions Box 484

What is the role of lymphatics in lipoprotein metabolism?

485

How could arterial wall lymphangiogenesis be enhanced? Would it increase reverse 486

cholesterol transport and could it be used as a treatment for atherosclerosis?

487

What is the most applicable delivery route and method for lymphangiogenic therapy for 488

myocardial ischemia?

489

Could attenuation of lymphatic vessel activation through VEGFR-3 inhibition therapy be 490

employed in the clinics to treat heart transplantation patients to prolong cardiac allograft 491

survival?

492

What are the side effects of lymphangiogenic therapy and how could they be avoided?

493 494495 496497 498499 500501 502503 504505 506507 508509 510511 512513 514 515516 517518 519 520521 522523 524525 526527 528529 530

(18)

References 531532

1 Cui, Y. (2010) Impact of lymphatic vessels on the heart.Thorac. Cardiovasc. Surg.58, 1-7 533

2 Wigle, J.T. and Oliver, G. (1999) Prox1 function is required for the development of the murine 534

lymphatic system.Cell98, 769-778 535

3 Joukov, V. et al. (1996) A novel vascular endothelial growth factor, VEGF-C, is a ligand for the 536

Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases.Embo J.15, 290-298 537

4 Kukk, E. et al. (1996) VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests 538

a role in lymphatic vascular development.Development122, 3829-3837 539

5 Breiteneder-Geleff, S. et al. (1999) Angiosarcomas express mixed endothelial phenotypes of blood 540

and lymphatic capillaries: podoplanin as a specific marker for lymphatic endothelium.Am. J. Pathol.

541

154, 385-394 542

6 Banerji, S. et al. (1999) LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific 543

receptor for hyaluronan.J. Cell Biol.144, 789-801 544

7 Lavina, B. and Gaengel, K. (2015) New imaging methods and tools to study vascular biology.Curr.

545

Opin. Hematol.22, 258-266 546

8 Aspelund, A. et al. (2016) Lymphatic System in Cardiovascular Medicine.Circ. Res.118, 515-530 547

9 Martel, C. et al. (2013) Lymphatic vasculature mediates macrophage reverse cholesterol transport in 548

mice.J. Clin. Invest.123, 1571-1579 549

10 Klotz, L. et al. (2015) Cardiac lymphatics are heterogeneous in origin and respond to injury.

550

Nature522, 62-67 551

(19)

11 Henri, O. et al. (2016) Selective Stimulation of Cardiac Lymphangiogenesis Reduces Myocardial 552

Edema and Fibrosis Leading to Improved Cardiac Function Following Myocardial Infarction.

553

Circulation133, 1484-1497 554

12 Zheng, W. et al. (2014) Lymphangiogenic factors, mechanisms, and applications.J. Clin. Invest.

555

124, 878-887 556

13 Srinivasan, R.S. et al. (2007) Lineage tracing demonstrates the venous origin of the mammalian 557

lymphatic vasculature.Genes Dev.21, 2422-2432 558

14 Yang, Y. et al. (2012) Lymphatic endothelial progenitors bud from the cardinal vein and 559

intersomitic vessels in mammalian embryos.Blood120, 2340-2348 560

15 Alitalo, K. et al. (2005) Lymphangiogenesis in development and human disease.Nature438, 946- 561

562 953

16 Martinez-Corral, I. et al. (2015) Nonvenous origin of dermal lymphatic vasculature.Circ. Res.

563

116, 1649-1654 564

17 Stanczuk, L. et al. (2015) cKit Lineage Hemogenic Endothelium-Derived Cells Contribute to 565

Mesenteric Lymphatic Vessels.Cell. Rep.

566

18 Ulvmar, M.H. et al. (2016) Pdgfrb-Cre targets lymphatic endothelial cells of both venous and non- 567

venous origins.Genesis54, 350-358 568

19 Francois, M. et al. (2008) Sox18 induces development of the lymphatic vasculature in mice.

569

Nature456, 643-647 570

20 Srinivasan, R.S. et al. (2010) The nuclear hormone receptor Coup-TFII is required for the initiation 571

and early maintenance of Prox1 expression in lymphatic endothelial cells.Genes Dev.24, 696-707 572

(20)

21 Francois, M. et al. (2011) The transcriptional control of lymphatic vascular development.

573

Physiology (Bethesda)26, 146-155 574

22 Aranguren, X.L. et al. (2013) COUP-TFII orchestrates venous and lymphatic endothelial identity 575

by homo- or hetero-dimerisation with PROX1.J. Cell. Sci.126, 1164-1175 576

23 Srinivasan, R.S. et al. (2014) The Prox1-Vegfr3 feedback loop maintains the identity and the 577

number of lymphatic endothelial cell progenitors.Genes Dev.28, 2175-2187 578

24 Karkkainen, M.J. et al. (2004) Vascular endothelial growth factor C is required for sprouting of the 579

first lymphatic vessels from embryonic veins.Nat. Immunol.5, 74-80 580

25 Nurmi, H. et al. (2015) VEGF-C is required for intestinal lymphatic vessel maintenance and lipid 581

absorption.EMBO Mol. Med.7, 1418-1425 582

26 Baldwin, M.E. et al. (2005) Vascular endothelial growth factor D is dispensable for development 583

of the lymphatic system.Mol. Cell. Biol.25, 2441-2449 584

27 Bos, F.L. et al. (2011) CCBE1 is essential for mammalian lymphatic vascular development and 585

enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo.Circ. Res.109, 586

486-491 587

28 Jeltsch, M. et al. (2014) CCBE1 enhances lymphangiogenesis via A disintegrin and 588

metalloprotease with thrombospondin motifs-3-mediated vascular endothelial growth factor-C 589

activation.Circulation129, 1962-1971 590

29 Bui, H.M. et al. (2016) Proteolytic activation defines distinct lymphangiogenic mechanisms for 591

VEGFC and VEGFD.J. Clin. Invest.

592

30 Xu, Y. et al. (2010) Neuropilin-2 mediates VEGF-C-induced lymphatic sprouting together with 593

VEGFR3.J. Cell Biol.188, 115-130 594

(21)

31 Schacht, V. et al. (2003) T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature 595

formation and causes lymphedema.Embo J.22, 3546-3556 596

32 Uhrin, P. et al. (2010) Novel function for blood platelets and podoplanin in developmental 597

separation of blood and lymphatic circulation.Blood115, 3997-4005 598

33 Herzog, B.H. et al. (2013) Podoplanin maintains high endothelial venule integrity by interacting 599

with platelet CLEC-2.Nature502, 105-109 600

34 Yang, Y. and Oliver, G. (2014) Development of the mammalian lymphatic vasculature.J. Clin.

601

Invest.124, 888-897 602

35 Sabine, A. et al. (2015) FOXC2 and fluid shear stress stabilize postnatal lymphatic vasculature.J.

603

Clin. Invest.125, 3861-3877 604

36 Fatima, A. et al. (2016) Foxc1 and Foxc2 deletion causes abnormal lymphangiogenesis and 605

correlates with ERK hyperactivation.J. Clin. Invest.126, 2437-2451 606

37 Flaht-Zabost, A. et al. (2014) Cardiac mouse lymphatics: developmental and anatomical update.

607

Anat. Rec. (Hoboken)297, 1115-1130 608

38 Flaht, A. et al. (2012) Cellular phenotypes and spatio-temporal patterns of lymphatic vessel 609

development in embryonic mouse hearts.Dev. Dyn.241, 1473-1486 610

39 Kholova, I. et al. (2011) Lymphatic vasculature is increased in heart valves, ischaemic and 611

inflamed hearts and in cholesterol-rich and calcified atherosclerotic lesions.Eur. J. Clin. Invest.41, 612

487-497 613

40 Ratajska, A. et al. (2014) Comparative and developmental anatomy of cardiac lymphatics.

614

ScientificWorldJournal2014, 183170 615

(22)

41 Lupinski, R.W. (2009) Aortic fat pad and atrial fibrillation: cardiac lymphatics revisited.ANZ J.

616

Surg.79, 70-74 617

42 Brice, G.W. et al. (1993) Milroy Disease. InGeneReviews(R)(Pagon, R.A. et al, ed.), University 618

of Washington, Seattle 619

43 Mansour, S. et al. (1993) Lymphedema-Distichiasis Syndrome. InGeneReviews(R)(Pagon, R.A.

620

et al, ed.), University of Washington, Seattle 621

44 Romano, A.A. et al. (2010) Noonan syndrome: clinical features, diagnosis, and management 622

guidelines.Pediatrics126, 746-759 623

45 Townsend, N. et al. (2015) Cardiovascular disease in Europe--epidemiological update 2015.Eur.

624

Heart J.36, 2696-2705 625

46 Randolph, G.J. (2014) Mechanisms that regulate macrophage burden in atherosclerosis.Circ. Res.

626

114, 1757-1771 627

47 Yla-Herttuala, S. et al. (2013) Stabilization of atherosclerotic plaques: an update.Eur. Heart J.34, 628

3251-3258 629

48 Rader, D.J. et al. (2009) The role of reverse cholesterol transport in animals and humans and 630

relationship to atherosclerosis.J. Lipid Res.50 Suppl, S189-94 631

49 Gordon, T. et al. (1977) High density lipoprotein as a protective factor against coronary heart 632

disease. The Framingham Study.Am. J. Med.62, 707-714 633

50 Lim, H.Y. et al. (2013) Lymphatic vessels are essential for the removal of cholesterol from 634

peripheral tissues by SR-BI-mediated transport of HDL.Cell. Metab.17, 671-684 635

51 Vuorio, T. et al. (2014) Lymphatic vessel insufficiency in hypercholesterolemic mice alters 636

lipoprotein levels and promotes atherogenesis.Arterioscler. Thromb. Vasc. Biol.34, 1162-1170 637

(23)

52 Milasan, A. et al. (2016) Effects of LDL Receptor Modulation on Lymphatic Function.Sci. Rep.6, 638

27862 639

53 Yla-Herttuala, S. et al. (1987) Glycosaminoglycans and apolipoproteins B and A-I in human 640

aortas. Chemical and immunological analysis of lesion-free aortas from children and adults.

641

Arteriosclerosis7, 333-340 642

54 Schwartz, G.G. et al. (2012) Effects of dalcetrapib in patients with a recent acute coronary 643

syndrome.N. Engl. J. Med.367, 2089-2099 644

55 Barter, P.J. and Rye, K.A. (2012) Cholesteryl ester transfer protein inhibition as a strategy to 645

reduce cardiovascular risk.J. Lipid Res.53, 1755-1766 646

56 Thygesen, K. et al. (2012) Third universal definition of myocardial infarction.Nat. Rev. Cardiol.

647

9, 620-633 648

57 Frangogiannis, N.G. (2012) Regulation of the inflammatory response in cardiac repair.Circ. Res.

649

110, 159-173 650

58 Davis, K.L. et al. (2000) Effects of myocardial edema on the development of myocardial 651

interstitial fibrosis.Microcirculation7, 269-280 652

59 Ishikawa, Y. et al. (2007) Lymphangiogenesis in myocardial remodelling after infarction.

653

Histopathology51, 345-353 654

60 Witzenbichler, B. et al. (1998) Vascular endothelial growth factor-C (VEGF-C/VEGF-2) promotes 655

angiogenesis in the setting of tissue ischemia.Am. J. Pathol.153, 381-394 656

61 Kholova, I. et al. (2007) Adenovirus-mediated gene transfer of human vascular endothelial growth 657

factor-d induces transient angiogenic effects in mouse hind limb muscle.Hum. Gene Ther.18, 232- 658

659 244

(24)

62 Rissanen, T.T. et al. (2003) VEGF-D is the strongest angiogenic and lymphangiogenic effector 660

among VEGFs delivered into skeletal muscle via adenoviruses.Circ. Res.92, 1098-1106 661

63 Rutanen, J. et al. (2004) Adenoviral catheter-mediated intramyocardial gene transfer using the 662

mature form of vascular endothelial growth factor-D induces transmural angiogenesis in porcine 663

heart.Circulation109, 1029-1035 664

64 Losordo, D.W. et al. (2002) Phase 1/2 placebo-controlled, double-blind, dose-escalating trial of 665

myocardial vascular endothelial growth factor 2 gene transfer by catheter delivery in patients with 666

chronic myocardial ischemia.Circulation105, 2012-2018 667

65 Reilly, J.P. et al. (2005) Long-term (2-year) clinical events following transthoracic intramyocardial 668

gene transfer of VEGF-2 in no-option patients.J. Interv. Cardiol.18, 27-31 669

66 Syvaranta, S. et al. (2012) Lymphangiogenesis in aortic valve stenosis--novel regulatory roles for 670

valvular myofibroblasts and mast cells.Atherosclerosis221, 366-374 671

67 Niinimaki, E. et al. (2016) Lymphangiogenesis is increased in heart valve endocarditis.Int. J.

672

Cardiol.219, 317-321 673

68 Kim, H. et al. (2012) Regulation and implications of inflammatory lymphangiogenesis.Trends 674

Immunol.33, 350-356 675

69 Dashkevich, A. et al. (2016) Ischemia-Reperfusion Injury Enhances Lymphatic Endothelial 676

VEGFR3 and Rejection in Cardiac Allografts.Am. J. Transplant.16, 1160-1172 677

70 Azzi, J. et al. (2016) Targeted Delivery of Immunomodulators to Lymph Nodes.Cell. Rep.15, 678

1202-1213 679

71 Honkonen, K.M. et al. (2013) Lymph node transfer and perinodal lymphatic growth factor 680

treatment for lymphedema.Ann. Surg.257, 961-967 681

(25)

72 Visuri, M.T. et al. (2015) VEGF-C and VEGF-C156S in the pro-lymphangiogenic growth factor 682

therapy of lymphedema: a large animal study.Angiogenesis18, 313-326 683

73 Lahteenvuo, M. et al. (2011) Growth factor therapy and autologous lymph node transfer in 684

lymphedema.Circulation123, 613-620 685

74 Hassinen, I. et al. (2016) Intramyocardial gene therapy directed to hibernating heart muscle using a 686

combination of electromechanical mapping and positron emission tomography.Hum. Gene Ther.

687

75 Nurro, J. et al. (2016) AdVEGF-B186 and AdVEGF-DDeltaNDeltaC induce angiogenesis and 688

increase perfusion in porcine myocardium.Heart 689

76 Laakkonen, J.P. and Yla-Herttuala, S. (2015) Recent Advancements in Cardiovascular Gene 690

Therapy and Vascular Biology.Hum. Gene Ther.26, 518-524 691

77 Davydova, N. et al. (2016) Differential receptor binding and regulatory mechanisms for the 692

lymphangiogenic growth factors VEGF-C and VEGF-D.J. Biol. Chem.

693

78 Harvey, N.L. et al. (2005) Lymphatic vascular defects promoted by Prox1 haploinsufficiency 694

cause adult-onset obesity.Nat. Genet.37, 1072-1081 695

79 Brorson, H. et al. (2009) Breast cancer-related chronic arm lymphedema is associated with excess 696

adipose and muscle tissue.Lymphat Res. Biol.7, 3-10 697

80 Patila, T. et al. (2006) Vascular endothelial growth factor C-induced collateral formation in a 698

model of myocardial ischemia.J. Heart Lung Transplant.25, 206-213 699

700

Viittaukset

LIITTYVÄT TIEDOSTOT

Tekijänoikeudet ja niiden loukkaaminen sekä oman osaamisen suojaaminen on yksi erilaisten sosiaalisen median palveluiden hyödyntämiseen liittyvistä avainkysymyksistä, joka

Liite 3: Kriittisyysmatriisit vian vaikutusalueella olevien ihmisten määrän suhteen Liite 4: Kriittisyysmatriisit teollisen toiminnan viansietoherkkyyden suhteen Liite 5:

(1995) ovat tutkineet lämpötilan ja kosteuspitoisuuden säätöä yhden huo- neen kokoisessa testiympäristössä sekä sumean logiikan että karkeiden joukkojen teo- rian (rough

The functionality of the chamber was tested with oxidation experiments of toluene, resulting in secondary organic aerosol (SOA) yields of 12–42 %, de- pending on the initial

Here we show that VEGFR3 has an essential role in cardiac lymphatic vessel morphology and attenuated VEGFR3 signaling exposes mice to higher mortality, hemorrhages and a

Recent findings: Studies have shown that chylomicron entry to lacteals and lymph movement in intestinal lymphatic capillaries is an active process.. Regulators of

In chapter eight, The conversational dimension in code- switching between ltalian and dialect in Sicily, Giovanna Alfonzetti tries to find the answer what firnction

The new European Border and Coast Guard com- prises the European Border and Coast Guard Agency, namely Frontex, and all the national border control authorities in the member