Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Immune Cells as a Source and Target of Angiogenic and Lymphangiogenic Factors Stefania Loffredo ⋅ Rosaria Ilaria Staiano ⋅ Francescopaolo Granata ⋅  Arturo Genovese ⋅ Gianni Marone  Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II, School of Medicine, Naples, Italy

Abstract Angiogenesis and lymphangiogenesis are distinct and complex processes requiring a finely tuned balance between stimulatory and inhibitory signals. Immune and inflammatory cells can contribute to these processes by multiple mechanisms: directly by producing a broad array of angiogenic growth factors, and indirectly by secreting several cytokines, chemokines and other mediators able to coordinate the cell-cell interactions. Immune cells can stimulate or inhibit angiogenesis/lymphangiogenesis, depending on their activation status and subset specificity. We summarize recent findings reporting the expression and activity of angiogenic and lymphangiogenic factors and their receptors and coreceptors in immune cells. It is evident that modulation of angiogenesis and lymphangiogenesis by the innate and adaptive immune cells (mast cells, macrophages, dendritic cells, basophils, eosinophils, and some subsets of T cells) is a highly complex process not yet comCopyright © 2014 S. Karger AG, Basel pletely understood.

S.L. and R.I.S. contributed equally to this paper.

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

The term angiogenesis was coined by John Hunter in 1787 to describe the growth of blood capillaries. The formation of vessels is a complex process, requiring a finely tuned balance between numerous stimulatory and inhibitory signals, such as vascular endothelial growth factors (VEGFs), integrins, angiopoietins, chemokines, oxygen sensors, miRNAs, endogenous inhibitors and many others [1–3]. In the embryo, blood vessels arise from endothelial cell precursors which share an origin with hematopoietic progenitors [4]. These progenitors assemble into a primitive vascular labyrinth of small capillaries, in a process known as vasculogenesis. During adult life, most

16

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

blood vessels remain quiescent and angiogenesis occurs only in the cycling ovary and in the placenta. However, endothelial cells (EC) retain their remarkable ability to divide rapidly in response to different stimuli, such as hypoxia or inflammation. During angiogenesis, the vascular plexus progressively expands by means of vessel sprouting and remodels into a highly organized vascular network of larger vessels ramifying into smaller ones [5]. The lymphatic system develops in parallel, but secondarily to the blood vascular system through a process known as lymphangiogenesis [6, 7]. Lymphangiogenesis is primarily involved in embryonic development, but can be reactivated in adult life in response to different stimuli. In certain disorders, these stimuli become excessive and the balance between stimulators and inhibitors shifts, resulting in a (lymph)angiogenic switch. For example, angiogenesis and lymphangiogenesis occur in chronic inflammation, wound healing, autoimmunity, allograft rejection and tumor metastasis [6, 7]. In the early 1970s, Judah Folkman observed that tumor tissue was enriched by an extraordinary high number of blood vessels that were fragile and often hemorrhagic [8]. Folkman further noted that angiogenesis is rate-limiting for tumor growth and chronic inflammation [9]. The most potent proangiogenic molecules known so far are members of the VEGF family: VEGF-A, VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PlGF). VEGF-A and VEGF-B are key regulators of blood vessel growth, whereas VEGF-C and VEGF-D modulate lymphangiogenesis [10, 11]. Some VEGFs have differentially spliced forms that differ in their ability to enhance angiogenesis. Human VEGF-A has at least six isoforms: 121, 145, 165, 183, 189 and 204 [12]. Of these, VEGF-A165 and VEGF-A121 are the most potent proangiogenic isoforms. PlGF, expressed in placenta and certain tumors, has two major isoforms: PlGF-1 (PlGF131) and PlGF-2 (PlGF152) [13]. In addition to the best known proangiogenic activity of VEGFs, recently have been identified various isoforms of VEGF-A with presumably antiangiogenic activity. The proangiogenic isoforms are identified as VEGFxxx, while the antiangiogenic isoform are VEGFxxxb [14]. These antiangiogenic VEGFs could act as competitive inhibitors of VEGFxxx isoforms. VEGFs and PlGFs signal through three human members of the VEGF receptor (VEGFR) family: VEGFR-1, VEGFR-2, and VEGFR-3. VEGF functions are also modulated through the production of an alternative mRNA variant of VEGFR-1, namely soluble VEGFR-1 (sVEGFR-1) which lacks the transmembrane region necessary to attach the receptor to the cell membrane and is devoid of kinase activity. It prevents VEGF from activating VEGFR-1 or VEGFR-2 by inhibiting their dimerization. Following the discovery of the VEGF-VEGFR system, the angiopoietins (Angs), together with their corresponding Tie receptors (Tie1 and Tie2), were identified as the second EC-specific receptor tyrosine kinase signaling system [15]. The Ang/Tie system plays a key role in remodeling and maturation of blood vessels as well as lymphatic vessels. There are four known Angs, Ang-1, Ang-2, Ang-3 and Ang-4. Ang-1 and Ang-2 are ligands for the Tie2 receptor: Ang-1 is a full agonist, whereas Ang-2 is

an antagonist. Besides their role in angiogenesis, Angs modulate several aspects of inflammation [16]. Recent advances in the understanding of cancer biology have highlighted the functional role of another class of molecules involved in angiogenesis/lymphangiogenesis: the semaphorins [17, 18]. Semaphorins form a family of molecular signals known to guide and control cell migration during embryo development and in adults. All semaphorins are characterized by an amino-terminal 500-amino acid Sema domain that is essential for signaling. Semaphorins are grouped into eight classes based on their structural domains, with classes 3–7 comprising the vertebrate semaphorins [17]. Semaphorins signal through two major receptor families, plexins and neuropilins (NRPs). In vertebrates, two NRPs (NRP-1 and NRP-2) and nine plexins have been identified [19]. Neuropilins have the ability to bind with high affinity to multiple ligand families, they function as co-receptors, binding to extracellular ligands with high affinity and complexing with other transmembrane receptors to form holoreceptors. NRPs are receptors for both the class 3 semaphorins and heparin-binding members of the VEGF family [20].

Mast cells are bone marrow-derived immune cells widely distributed throughout vascularized tissues and at interfaces with the external environment [21, 22]. They produce a wide array of mediators and cell-cell signaling molecules. Human mast cells synthesize histamine, which is stored in secretory granules as a preformed mediator. These granules also contain a variety of proteolytic enzymes (α- and β-tryptase, chymase, carboxypeptidase A, cathepsin G, secreted phospholipases A2 and proteoglycans) [21, 23]. Mast cells are a major source of arachidonic acid-derived lipid mediators and PAF. Immunologic stimulation of human mast cells activates a specific program of gene expression leading to de novo synthesis of a wide spectrum of cytokines (IL-3, IL-5, IL-6, IL-13, IL-16, IL-18, IL-25, TGF-β, SCF, GM-CSF, TNF-α) and chemokines (CXCL8, CCL3, CCL2, CCL1) [21]. This variety of mediators may account for the implication of mast cells in several chronic inflammatory diseases and in tumor growth [24]. Mast cells infiltrate the sites of chronic inflammation, which can lead to cancer in certain conditions. Despite that mast cell-produced cytokines may participate in antitumor responses by fostering tumor rejection or apoptosis, during the last decade the protumorigenic role of mast cells took priority. In fact, it has been highlighted that mast cells density is increased at the margins of various tumors in humans [25–27] and in rodents [28–35]. In addition, mast cell infiltration around human tumors correlates with angiogenesis and metastasis [36]. Initial in vitro studies demonstrated that rodent mast cells can produce certain angiogenic factors, such as VEGFs and

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

17

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Expression of VEGFs and Their VEGFRs in Mast Cells

VEGFR-2

NRP-1 NRP-2

Tie1

VEGFR-2

Tie2 NRP-1

Tie1

NRP-2

VEGFR-1

Mast cell

Basophil

VEGF-A

VEGFR-2

NRP-1

VEGF-C

Ang-1

VEGF-B

VEGFR-1 Tie1

Tie2

Tie2

VEGFR-3 Tie1

VEGF-D

NRP-2

LEC

BEC Angiogenesis

Lymphangiogenesis

Angs [29, 37] (fig. 1). Moreover, several authors described the production of VEGF by mast cells derived from cord blood progenitors and transformed human mast cell lines such as HMC-1 [38, 39]. Our group demonstrated the constitutive expression of several isoforms of angiogenic (VEGF-A and VEGF-B) and lymphangiogenic factors (VEGF-C and VEGF-D) in primary human mast cells and in two human mast cell lines (LAD-2 and HMC-1) [40]. We also demonstrated that these cells enhance the expression and the release of VEGF-A when stimulated with PGE2 and adenosine, two important molecules involved in tissue inflammation and hypoxia [41, 42]. VEGF-A secreted by human mast cells exerted proangiogenic activity in vivo [40]. These findings indicate that mast cells might be a component of the complex network involving chronic inflammation and tumor angiogenesis. Mast cells surrounding and infiltrating tumors can influence several aspects of cancer biology by releasing cytokines and protease in addition to proangiogenic molecules [43, 44]. The role of mast cells in tumor growth is also supported by the

18

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Fig. 1. Schematic representation of the release of VEGFs and Ang-1 from human basophils and mast cells. Activated basophils express and release several isoforms of VEGF-A, two isoforms of VEGF-B and Ang-1. These cells express VEGFR-2, NRP-1, NRP-2 and Tie1. VEGF-A, VEGF-B and Ang-1 can modulate angiogenesis through the interaction with VEGFR-2, VEGFR-1, Tie1, Tie2, and NRP-1 present on blood endothelial cells (BEC). Activated human mast cells express and release, in addition to VEGF-A and VEGF-B, the two most important lymphangiogenic factors, namely VEGF-C and VEGF-D. These cells express both VEGFR-1 and VEGFR-2, Tie1 and Tie2, and NRP-1 and NRP-2. VEGF-A and VEGF-B modulate angiogenesis, whereas VEGF-C and VEGF-D activate VEGFR-3 preferentially expressed in lymphatic endothelial cells (LEC).

observation that carcinoma formation and growth are reduced in mast-cell-deficient KitW-sh/W-sh mutant mice [28, 31]. Additional studies have emphasized the proangiogenic and protumorigenic roles of mast cells in a mouse model of epithelial carcinogenesis [28]. Melillo et al. [25] have demonstrated that human mast cells density is increased in human thyroid cancer and correlates [45] with their invasiveness. Moreover, we have shown that VEGF-A released by thyroid tumor cells is chemoattractant for mast cells, indicating that mast cells are a target, in addition to a source, for VEGF. We also characterized how mast cells can be activated by tumor thyroid cells to release histamine and to enhance the expression of cytokines and chemokines. Interestingly, mast cell-derived mediators can stimulate the proliferation of thyroid cancer cells [25]. Recent results demonstrated that mast cells density increases in human cutaneous lymphoma, and their number correlates with microvessel density and malignancy [46]. In summary, we and others have identified a plethora of mast cell inflammatory and immunomodulating activities that affects tumorigenesis. Therefore, we have proposed that mast cells surrounding and/or infiltrating tumors should be called tumor associated mast cells [24]. There is compelling evidence that tissue hypoxia is a hallmark of both chronic inflammation and cancer. Hypoxia and inflammation are interwined at the molecular, cellular, and clinical level [47]. Inflammation is an important cause of hypoxia and hypoxia results in a proinflammatory phenotype. Hypoxia upregulates the expression of hypoxia-inducible factor 1 (HIF-1) which increases the expression of VEGF-A [48]. It has been suggested that histamine can directly induce the expression of HIF-1 in mouse bone marrow-derived mast cells and subsequently upregulates the expression of VEGF-A [49]. HIF-1 expression can also modulate other inflammatory functions of human mast cells such as histamine and chemokine release [50].

Basophils circulate in human peripheral blood where they represent less than 1% of leukocytes. IL-3 is the principal cytokine responsible for human basophil growth and differentiation from CD34+ pluripotent progenitor cells. Other cytokines (GM-CSF, IL-1, IL-5, IL-33, NGF and leptin) are also important for human basophil growth, differentiation, and mediator production [21, 51]. Basophils have been mistakenly considered the Cinderella of the immune system for several decades. In the 1990s, it was discovered that they represent a major source of IL-4 and can contribute to Th2 polarization [52–55]. Mast cells and basophils, the only inflammatory cells expressing the high affinity receptor for IgE (FcεRI), are conventionally considered key players in the pathogenesis of allergic disorders [21]. However, there is increasing evidence that basophils play a relevant role in the initiation and progression of certain autoimmune diseases [56].

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

19

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Expression of VEGFs, Their VEGFRs and NRPs in Human Basophils

Few groups have focused their attention on the role of basophils in angiogenesis and lymphangiogenesis. de Paulis et al. [57] explored the expression of VEGFs and their receptors in human basophils. Basophils constitutively express several isoforms of VEGF-A (VEGF-A121, VEGF-A165 and VEGF-A189) and their immunologic activation induces the release of VEGF-A. Interestingly, VEGF-A is chemotactic for basophils presumably through the interaction with VEGFR-2. The vast majority of these cells phenotypically expressed NRP-1 and NRP-2. It is intriguing that basophils constitutively express also sVEGFR-1 mRNA. These cells do not express VEGF-C and VEGF-D mRNAs and presumably modulate angiogenesis, but not lymphangiogenesis. Cerny-Reiterer et al. [58] reported that basophils isolated from a patient with chronic myeloid leukemia can produce another proangiogenic factor, hepatocyte growth factor (HGF). They also demonstrated that basophil-derived HGF induces EC migration in vitro. In addition, IL-3 promotes the expression and release of HGF from chronic myeloid leukemia basophils [58]. The latter findings suggest that basophils, in addition to mast cells, may play a role in the progression of certain tumors.

Expression of VEGFs and VEGFRs in Monocytes, Macrophages and Dendritic Cells

Macrophages, dendritic cells (DCs) and their circulating precursor monocytes play an important role in the resolution of inflammation and in the modulation of angiogenesis. This ability is due to their ubiquitous distribution, functional plasticity, and capacity to produce a variety of mediators and angiogenic factors [59–61].

Macrophages Macrophages are the first line of defense against infectious organisms and remove dead cells during development and reparative processes. In addition to functioning as phagocytic cells, macrophages play a pivotal role in such pathological processes as

20

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Monocytes There is compelling evidence that different bone marrow-derived myeloid cells participate in various aspects of cancer development and progression. VEGF-A165 and VEGF-A121 induce monocyte migration through the activation of VEGFR-1 [62]. Sawano et al. [63] have shown that the majority of human monocytes express V ­ EGFR-1 (83%), but not VEGFR-2. PlGF-1 promotes cytokine and chemokine gene expression in monocytes and chemotaxis in the monocytic cell line THP-1 via VEGFR-1 [64]. Monocytes are also a source, in addition to be a target, of angiogenic mediators. Monocytes express and release VEGF-A induced by M-CSF [65] and by activation of adenosine receptors (A1 and A2A) [66]. Leukotrienes, via the engagement of cysLT1 receptor, induce the expression of VEGF in human monocytes [67]. The production of VEGF-A in monocytes is also linked to both intracellular calcium influx and the production of reactive oxygen species [68].

Angiogenic Activity of Macrophages Macrophages can promote cancer progression either directly, by stimulating the proliferation of tumor cells, or indirectly, by producing angiogenic and lymphangiogenic factors [74]. These proangiogenic programs are induced in TAMs by local activation signals resulting from metabolic conditions (e.g. hypoxia, lactate, pyruvate, or hydrogen ions) or from mediators produced at sites of tissue injury (e.g. IL-10, MCSF, LPS, adenosine) [70, 74]. VEGF is produced by TAMs and also by tumor cells in a variety of human cancers [25, 75]. We have demonstrated that human lung macrophages constitutively express and synthesize different forms of angiogenic (VEGF-A and VEGF-B), and lymphangiogenic (VEGF-C and VEGF-D) factors [76]. Secreted phospholipases A2, a class of inflammatory mediators whose expression is increased in certain human tumors, induce the release of VEGF-A and VEGF-C from human lung macrophages [76]. This effect is enhanced by adenosine analogs that induce a functional switch of macrophages by increasing VEGF-A and suppressing TNF-α through a cooperation between A2A and A3 adenosine receptors [76]. VEGF contributes to the expansion of the macrophages population by recruiting circulating monocytes which express VEGFR-1 [62]. We have recently found that also primary human lung macrophages retain a functional plasticity that modulates their angiogenic profile. Figure 2 illustrates that this plasticity depends on the activating stimuli and the type of mediators produced (e.g. TNF-α vs. VEGF-A) [Loffredo et al., unpubl. data].

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

21

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

chronic inflammation and cancer by releasing a variety of growth factors, cytokines and proteolytic enzymes, in response to different stimuli. These activities of macrophages are influenced by microenvironmental factors [61]. The majority of malignant tumors are infiltrated by macrophages, which can comprise more than 50% of the total tumor mass [69]. Tumor-associated macrophages (TAMs) are recruited early at tumor sites where they frequently display protumor functions, such as activation of the neoangiogenic switch, the secretion of soluble factors that support tumor cell resistance to apoptotic stimuli and stimulate the proliferation and invasion of malignant cells [70]. Two functional phenotypes have been described for macrophages: M1 and M2. M1 or classically activated macrophages are stimulated by IFN-γ, microbial products (e.g. LPS) and opsonized particles. They produce inflammatory (reactive oxygen species and NO) and immunostimulating cytokines (IL-1β, IL-12, TNF-α) to elicit the adaptive immune response, and may exert antitumor activity [61]. Macrophages acquire an M2 phenotype in response to Th2 cytokines (IL-4, IL-13), apoptotic bodies, and immune complexes. M2 are characterized by an immunosuppressive phenotype (IL-10), and have high scavenging activity and ability to support tissue repair and remodeling [71–73]. During the early stages of tumor development, macrophages have an M1 phenotype, which contribute to tumor rejection, but once the tumor is established macrophages switch to a proangiogenic M2 phenotype, which stimulates tumor growth.

TNF-α (fold increase vs. unstimulated)

*

a

* LPS + INF-γ

LPS

LPS + IL-10

* LPS + INF-γ

LPS

LPS + IL-10

VEGF-A (fold increase vs. unstimulated)

*

b

Fig. 2. Production of TNF-α (a) and VEGF-A (b) during human lung macrophage polarization. Macrophages, isolated from lung parenchyma of patients undergoing thoracic surgery, were incubated with LPS (1 µg/ml) plus IFN-γ (1,000 U/ml), LPS plus IL-10 (10 ng/ml) or LPS alone for 24 h at 37 ° C. At the end of incubation, the production of VEGF-A and TNF-α was evaluated in supernatants by ELISA. a IFN-γ enhances the production of TNF-α induced by LPS; by contrast IL-10 blocks the release of this cytokine induced by LPS. b Opposite effects were obtained when the production of VEGF-A was evaluated. In this case, INF-γ inhibits VEGF-A release induced by LPS, whereas IL-10 increases its production. This figure illustrates the results obtained in six separate experiments. * p < 0.001 when compared to LPS alone.

In tumor areas of low oxygen tension, where TAMs usually accumulate, hypoxia associated with tissue injury results in breakdown of ATP and release of adenosine [77]. Activation of adenosine receptors (AR) on the surface of macrophages induce anti-inflammatory signals, reduce TNF-α, IL-12 and MIP-1α release and upregulate the expression of VEGF [78]. It has been suggested that TLR and AR signaling work in concert to induce an angiogenic phenotype because macrophages knocked-down for TLR are unable to produce VEGF in response to AR agonists [78, 79]. In addition, hypoxic conditions induce an overexpression of HIFs in macrophages. HIF-1 and HIF-2α play a central role in the production of VEGF (VEGF-A, VEGF-C and VEGFD) and of CXCL8 [70]. Macrophages produce TNF-α which is an activating mediator and sustains the growth of tumor cells and blood vessels. TNF-α stimulates the production of several angiogenic factors (VEGF, bFGF and IL-8) and the activation of matrix degrading enzymes (MMP-9) [69]. MMP-9 promotes the release of VEGF from extracellular matrix storage sites through proteolytic mechanisms [80]. IL-8 is a potent angiogenic

22

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

   

chemokine produced by TAMs in human malignancies [69]. The interaction between infiltrated macrophages and tumor cells is also known to upregulate IL-8 (and VEGF) expression in both cell types in a paracrine manner [81]. IL-8 expression has been correlated negatively with patient survival and positively with macrophage infiltration in tumors, suggesting that this chemokine might be dominantly derived from TAMs [69]. IL-6, released by TAMs, plays a key role in sustaining the survival and proliferation of malignant cells in tumors of epithelial and hematopoietic origin [70]. IL-1 enhances invasiveness of the tumor and induces angiogenesis and immune suppression in the host [82]. IL-1 has been shown to selectively induce TNF-α, angiogenin and HIF-1, thereby promoting VEGF production [69]. In addition to the earlier-mentioned molecules, TAMs can also produce proteolytic enzymes which mobilize VEGF from extracellular matrix stores, indirectly sustaining tumor angiogenesis [83]. bFGF promotes every phase of the angiogenic ­process, including synthesis of proteinases and endothelial cell migration [69]. ­Furthermore, TAMs express urokinase-type plasminogen activator receptor [84], macrophage-inhibitory factor, and PAF [85] that modulate angiogenesis.

Lymphangiogenic Activity of Macrophages Macrophages play an important role also in the formation of lymphatic vessels. Both tumor cells and macrophages express lymphangiogenic factors, such as VEGF-C and VEGF-D, which activate VEGFR-3 present on lymphatic endothelial cells [76, 93]. Macrophages apparently may play a dual role in lymphangiogenesis. Their circulating precursors, monocytes, migrate from blood vessels to tumor tissues where these cells are exposed to TNF-α and other proinflammatory mediators, and are converted into VEGF-C-secreting macrophages. In addition, macrophages may transdifferentiate into lymphatic endothelial cells by forming cell aggregates that integrate into an existing lymphatic vessel [94, 95].

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

23

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Antiangiogenic Activity of Macrophages In addition to angiogenic induction, macrophages, under certain conditions, can exert antiangiogenic activity. These cells can promote vascular regression by activating EC apoptosis [86]. Moreover, macrophages can produce IL-12 and IL-18 which are known for their antitumor and antimetastatic activity [87], inhibiting angiogenesis through diverse mechanisms [88]. Inhibition of angiogenesis by IL-12 and IL-18 is not due to a direct interaction with EC, but through induction of IFN-γ production by Th1 cells and NK cells [69], IFN-γ, in turn, induces production of angiostatic molecules such as CXCL10 and CXCL9 [89]. Another angiostatic effect of IL-18 is its ability to inhibit bFGF-induced EC proliferation [90]. Interestingly, IL-18 and IL-12 synergistically exert antitumor effects and inhibit angiogenesis [91]. Finally, macrophages can release metalloelastase (MMP-12), which, through proteolytic degradation of plasminogen, generates angiostatin, an endogenous inhibitor of angiogenesis [92].

Dendritic Cells DCs are professional APC and have long been known as critical regulators of adaptive immune responses. There is evidence that DCs can influence the angiogenic process. DCs are not often found in tumor infiltrates [69], and, if present, they are mostly in an immature state and incapable of inducing an effective immune response [69]. Conflicting data demonstrate that DCs, especially immature DCs (iDCs), can produce both pro- and antiangiogenic mediators. iDCs have been shown to silence immunity and induce tolerance by deleting T cells or by expanding Treg [69, 86]. This phenomenon appears to be mediated by tumor-derived factors such as VEGF, IL-6 and M-CSF. Moreover, DCs can also stimulate angiogenesis indirectly by enhancing the angiogenic potential of other cell types. NRP-1 on DC surface plays a crucial role in angiogenic sprouting in endothelial cells. NRP-1 can be transferred from DCs to T cells by trogocytosis. As a result, T cells capture and carry VEGF to endothelial cells at sites of angiogenic stimulation [96]. Conversely, DC infiltration in tumor lesions is associated with improved survival rates and reduced incidence of recurrent disease in different types of malignancies [97]. Moreover, DCs have the unique ability to present tumor-specific antigens and subsequently, activate a specific antitumor T cell response in vivo. The antiangiogenic activity of DCs is mediated by the production of IL-12 and by sVEGFR-1. Kishuku et al. [98] have shown that this receptor, expressed by DCs, effectively inhibits angiogenesis in vitro and in vivo. DCs can also modulate lymphangiogenesis through the expression of VEGFR-3 and the production of VEGF-C [99].

Foxp3+ regulatory T cells (Treg) are a population of CD4+ T cells that play a central role in maintaining self-tolerance and in regulating responses to infectious agents and tumor antigens [100]. Both natural Treg cells (nTreg), which develop early in the thymus, and induced Treg cells (iTreg), which are generated later in the periphery, express Foxp3 and have complementary functions that lead to suppression of immune reactivity [101]. Until few years ago, it was impossible to distinguish phenotypically these two Treg subsets and to understand the specific contribution of each of them to immune regulation. Recently, the differential expression of NRP-1 on the surface of nTreg and iTreg has allowed a better characterization of these subsets. NRP-1 is a transmenbrane protein involved in angiogenesis as a coreceptor of Sema3 and VEGFs. NRP-1 participates in the priming of CD4+ T cells by DCs and in the regulation of the immunological synapse and response [102]. Using a combination of a novel transgenic mice, two groups have independently demonstrated the existence of two subsets of Foxp3+ Treg cells: a NRP-1high subset (70–80% of total cells) and a NRP-1low subset (20–30% of total cells) [103, 104]. NRP-1high Treg cells are the nTreg cells because their NRP-1 level is correlated with expression of Helios (a tran-

24

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Expression of NRPs in Regulatory T Cells

scription factor used as a marker of nTreg cells). Weiss et al. [103] showed that ­NRP-1low Treg cells express the protein DAPL1 (death-associated protein-like 1) and are iTreg cells. The levels of NRP-1 are generally stable on both Treg cell subsets after Treg activation and proliferation in response to TCR stimulation. However, TGF-β induces NRP-1 expression on Treg cells, whereas IL-6 prevents TGF-β-induced NRP-1 upregulation in vitro [103]. Collectively, these data indicate that NRP-1 provides an excellent marker to distinguish Treg subsets. Both populations appear similarly efficient in suppressing T cell proliferation in vitro, antibody responses in vivo and the development of diabetes in genetically predisposed mice [103, 104].

Expression of Angiopoietins in Immune Cells

Mast Cells and Basophils Murine mast cells express Ang-1 and Tie2, but not Ang-2 [29]. These cells promote the growth of plasma cell tumors through secretion of Ang-1, which stimulates neovascularization in conjunction with tumor derived VEGF-A [29]. Recently we have observed that human lung mast cells and LAD-2 constitutively express Ang-1 and Ang-2. In our experimental conditions, neither Ang-1 nor Ang-2 are released from these cells [106]. Tie1 is expressed on 63% of human lung mast cells whereas Tie2 was highly expressed (99%) on these cells. We also found that low concentrations of ­Ang-1 selectively induced chemotaxis of mast cells [106]. We have also found that human basophils express both Ang-1 and Ang-2 mRNAs. These cells release Ang-1, spontaneously and upon cellular activation by PMA and bryostatin 1. By contrast, none of the stimuli examined induces Ang-2 release [106]. Moreover, basophils express Tie1 at both mRNA and protein level. These findings led us to suggest the possibility of a crosstalk between basophils and mast cells: activated

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

25

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Angiopoietins (Angs) play an important role in regulating both the quiescent and the angiogenic microvasculature. The Ang-Tie system consists of two cell-surface tyrosine kinase receptors (Tie1 and Tie2) and three ligands (Ang-1, Ang-2, and Ang-3/ Ang-4). Tie2 is primarily expressed on ECs and binds all the known angiopoietins with comparable affinity, whereas Tie1 is still an orphan receptor that can regulate Tie2 activity via heterodimerization with it [105]. Ang-1, primarily expressed by perivascular cells, stabilizes the blood vascular endothelium, maintaining EC survival and limiting the angiogenic response in pathological conditions. Ang-2 is released by activated ECs and promotes plasticity in the sites of vascular remodeling and angiogenesis by antagonizing Ang-1-mediated EC quiescence, although the functions of Ang2 appear to vary in a context-dependent manner [15]. The Ang-Tie system functions as a key regulator of vascular quiescence system. In mature vasculature, the Ang-Tie system controls endothelial homeostasis during acute injury, inflammation and pathological angiogenesis [16].

basophils can release Ang-1 that, in turn, might attract mast cells through Tie2 engagement. Thus, mast cells and basophils could play a coordinated role in the complex orchestration of inflammatory and tumor angiogenesis. Eosinophils Few studies have focused on the role of eosinophils in the modulation of angiogenesis and lymphangiogenesis and on their interactions with angiopoietins. The group of Francesca Levi-Schaffer demonstrated that human eosinophils produce at least two angiogenic molecules, namely VEGF-A [107] and osteopontin [108]. Moreover, they found that hypoxia induces the release of VEGF-A and IL-8 [109]. Eosinophil infiltration is a characteristic feature of allergic inflammation and levels of VEGF correlate well with the percentage of eosinophils and eosinophil cationic proteins in induced sputum of patients with asthma [110]. Moreover, Ang-1 and Ang2 levels are higher in sputum of patients with severe asthma compared to moderate asthma and healthy subjects [111]. VEGF, as well as Ang-1, are potent chemoattractant for human eosinophils [112, 113]. Ang-1 can induce eosinophil migration with a maximal response in the range of 10 pg/ml to 100 ng/ml, which is the range of Ang-1 concentration in human plasma. By contrast, Ang-2 exerts only minor chemotactic property on eosinophils [112]. Moreover, they report that Ang-1, but not Ang-2, can inhibit VEGF-induced eosinophil chemotaxis [112]. A comprehensive review on the role of eosinophil in angiogenesis is included in this volume [114].

Macrophages Tie2 is specifically expressed on a subtype of Tie2+ monocytes/macrophages called Tie2-expressing monocytes/macrophages (TEMs) implicated in angiogenesis [122]. Tie2 is weakly expressed by monocytes, but is upregulated upon their homing to a variety of tumors [105]. The relationship between TEMs and tumors suggests that these cells may crosstalk with ECs to provide paracrine support to the angiogenic vasculature. Mouse TEMs present a unique genetic profile that consists in an increased

26

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Neutrophils Neutrophils are the primary leukocytes to be recruited at inflammatory sites and modulate a wide range of inflammatory activities [115]. These cells can release VEGF and metalloproteinases, which contribute to extracellular matrix degradation [116]. Human neutrophils express Tie2 receptor and both Ang-1 and Ang-2 exert pro-inflammatory activities inducing PAF synthesis from ECs and neutrophils [117]. In addition, Ang-1, but not Ang-2, binds to Tie2 and prolongs neutrophil viability mainly through the release of IL-8 [116, 118]. The effect of Angs on neutrophil recruitment is still under debate. In fact, while Sturn et al. [119] reported that Ang-1 inhibits VEGF-mediated neutrophil migration, Brkovic et al. [120] showed that both Ang-1 and Ang-2 can increase neutrophil chemotaxis. A comprehensive review on the role of neutrophils in tumor angiogenesis is included in this volume [121].

proangiogenic activity and lower proinflammatory activity compared with Tie2-negative tumor macrophages [105, 123]. Ang-2 induces Tie2 phosphorylation in human monocytes and increases their expression of proangiogenic genes [124]. In addition to promoting angiogenesis, Ang-2 may stimulate TEMs also to acquire an immunosuppressive function in tumors. Ang2 stimulates the release of the IL-10 and of CCL17 [125], suppressing T cell proliferation and promoting the expansion of Tregs, respectively [124, 126]. Ang-1-Tie2 signaling inhibits LPS-induced macrophage activation, which is shown by decreased cell migration and production of TNF-α [125]. Mouse macrophages express mRNA for Ang-2, but not for Ang-1 and Ang-3. Constitutive Ang-2 secretion is enhanced by LPS, IFN-γ, PGE2, and VEGF [127]. Mouse macrophages also express angiopoietin-like protein 4 (AngTL4) and its expression is inhibited by TLR activation induced by LPS [128].

The IL-17 cytokine family includes six members, namely IL-17A, IL-17B, IL-17C, IL17D, IL-17E/IL-25, and IL-17F. IL-17A and IL-17F are produced primarily by a unique lineage of CD4+ T helper type 17 cells (Th17) [129]. These cells express IL-23 and develop in response to IL-23, conditions in which Th1 and Th2 development are suppressed and express the nuclear receptor RORγt. IL-17 plays a role in host defense and is a key mediator of autoimmune (rheumatoid arthritis, psoriasis, inflammatory bowel disease), inflammatory disorders (asthma), and tumorigenesis [130, 131]. IL-17A and IL-17F are mainly expressed by Th17 cells, whereas IL-17E/IL-25 is expressed by murine mast cells [132]. IL-17 receptors (IL-17R) are ubiquitously expressed in epithelial cells, ECs, hematopoietic cells, fibroblasts, and osteoblasts [133]. In addition to its proinflammatory activity, IL-17 is an angiogenic factor in tumors and in rheumatoid arthritis [134, 135]. Numasaki and collaborators have demonstrated that IL-17 is a potent inducer of angiogenic chemokines from a number of cells [136, 137]. Indeed, IL-17 has the ability to induce the production of a variety of proangiogenic factors, including VEGF and HGF [138]. Moreover, IL-17 potentiates the mitogenic action of β-FGF, HGF and VEGF for vascular endothelial cells [139]. The precise mechanisms by which IL-17 elicits new vessel formation in vivo have not been completely understood yet. It has been suggested that airway exposure to LPS induces the production of VEGF and that T cell priming and Th1 and Th17 responses to LPS-containing allergens depend on VEGF [140]. In a mouse model of noneosinophilic asthma (high-dose LPS model), it has been shown that T cell priming depends on VEGFR-1-mediated signaling, mainly via the maturation and migration of lung DCs. In this model, Th17 polarization depends on VEGFR-2-mediated signaling mainly via the production of Th17polarizing cytokine such as IL-6 [141].

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

27

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Direct and Indirect Angiogenic Activity of IL-17

IL-17E/IL-25 Promotes Angiogenesis in Asthma

Different members of the IL-17 family are involved in various waves of airway inflammatory diseases. IL-17A has been reported to play an important role in the pathogenesis of chronic obstructive pulmonary disease, bacterial pneumoniae, and asthma [142], where it might be involved in airway remodeling by inducing growth and mucin production of airway epithelial cells. IL-17A and IL-17F influence chemokine release by lung ECs in response to proinflammatory stimuli such as IL-1β and TNF-α [143]. IL-25 induces a Th2-type inflammatory reaction in the airways characterized by local upregulation of IL-4, IL-5 and IL-13, overexpression of eotaxin and tissue eosinophilia [144]. It has been suggested that exposure of lung fibroblast to IL-25 may contribute to the recruitment and enhanced survival of inflammatory cells (e.g. eosinophils) [145]. Through all these effects, IL-25 may participate in the maintenance of airway inflammation in asthma and other respiratory inflammatory diseases. IL-17E/IL-25 Production by Human Eosinophils and Basophils IL-17E/IL-25 was found to be expressed by mouse mast cells [132]. However, it remains unclear whether these cells can secrete bioactive IL-25 protein. Human eosinophils and basophils express IL-25 transcript and are the only source of this cytokine compared to other cell lineage (T and B cells, NK cells, monocytes, and DCs) [146]. Wang et al. [146] have demonstrated that human eosinophils and basophils release IL-25 upon activation. Interestingly, while eosinophils isolated from normal or allergic subjects produce similar levels of IL-25, basophils from allergic patients produce twofold more IL-25 than that produced by basophils from normal subjects. They suggested that IL-25 may regulate Th2 memory cells both enhancing the Th2 expansion and polarization. These results suggest that eosinophils and basophils are the major cell types that produce IL-25 in humans.

There is increasing evidence that cells of innate and adaptive immunity can participate in the highly complex processes of angiogenesis and lymphangiogenesis (fig. 3). It is now well established that inflammatory and immune cells can release a wide spectrum of angiogenic factors. By contrast, the role of immune cells in the modulation of lymphangiogenesis is still in its infancy and warrant additional investigations. However, we and others have demonstrated that activated human macrophages, mast cells and DCs can express and release the two most important lymphangiogenic factors, namely VEGF-C and VEGF-D [40, 76, 99, 147]. The role of different populations and subpopulations of B and T cells (e.g. Th17, Treg) in the modulation of angiogenesis/ lymphangiogenesis is incompletely understood. There is some evidence that B cells

28

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Concluding Remarks

Ba

PMN

PMN

Ba

Eos

s

Mono

s Eos



DC

Blo

od

ve

sse

MC ls

m Ly

ph

at

ic

s ve

se

ls

can secrete VEGF-A [148], whereas T lymphocytes negatively regulate lymph node lymphatic vessel formation mainly through IFN-γ [149]. It is now evident that there are striking differences in the production of angiogenic and lymphangiogenic molecules among different immune cells. For instance, activated human basophils are a major source of several isoforms of VEGF-A [57]. By contrast, human mast cells [40] and macrophages [76] produce both angiogenic and lymphangiogenic factors. This suggests that different immune cells express distinct repertories of molecules influencing angiogenesis and lymphangiogenesis. Interestingly, certain immune cells (e.g. macrophages) have the potential to produce proangiogenic or antiangiogenic molecules depending on tissue setting or experimental conditions.

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

29

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

Fig. 3. Schematic representation of immune cells participating in inflammatory angiogenesis and lymphangiogenesis. Circulating immune cells such as neutrophils (PMN), basophils (Bas), eosinophils (Eos), and monocytes (Mono) can be recruited (dotted lines) at sites of inflammation by chemoattractants as well as by angiogenic factors (e.g. VEGFs and Angs). These cells, together with resident immune cells such as monocyte-derived macrophages (MΦ), DCs, and mast cells (MC), contribute to the formation of new blood vessels by producing several angiogenic factors (large continuous lines). Tissue mast cells, macrophages, and dendritic cells have also the capacity to produce lymphangiogenic factors (small continuous lines) that contribute to the formation of new lymphatic vessels. The role of different populations and subpopulations of B and T cells in the modulation of angiogenesis/lymphangiogenesis is incompletely understood. During chronic inflammation and tumor growth several immune cells can play a dual role being both source and target of angiogenic and lymphangiogenic factors.

During the last years we have learned that immune cells are not only a source of angiogenic molecules, but also their target. In fact, different immune cells express selectively distinct receptors (VEGFR-1 and/or VEGFR-2, Tie1 and/or Tie2) and coreceptors (NRP-1 and/or NRP-2) for angiogenic factors. These molecules (e.g. VEGFs, PlGF, Angs) can exert pro-inflammatory effects (e.g. chemotaxis) by engaging specific receptors and/or coreceptors on immune cells. Thus, it appears that angiogenic/ lymphangiogenic molecules produced by immune cells play a dual role: they can influence angiogenesis/lymphangiogenesis and can contribute to chronic inflammation at least by recruiting other immune cells. Folkman was a pioneer in suggesting that immune cells can influence angiogenesis not only in tumor growth [8], but also in chronic inflammation [9]. There is now evidence that inflammatory angiogenesis can exert a prominent role in the initiation and progression of several chronic inflammatory disorders such as rheumatoid arthritis [150], skin [151] and allergic disorders [45]. Although angiogenesis/lymphangiogenesis regulation by immune cells is a highly complex process incompletely understood, we suggest that these events warrant consideration for future therapeutic intervention on chronic inflammatory disorders and tumors.

Acknowledgements This work was supported in part by grants from the Ministero dell’Istruzione, Università e Ricerca (MIUR), the Istituto Superiore di Sanità (AIDS Project) and Regione Campania (CISI Lab Project and CREME Project).

  1 Mammoto A, Connor KM, Mammoto T, Yung CW, Huh D, Aderman CM, Mostoslavsky G, Smith LE, Ingber DE: A mechanosensitive transcriptional mechanism that controls angiogenesis. Nature 2009; 457:1103–1108.   2 Weis SM, Cheresh DA: Alphav integrins in angiogenesis and cancer. Cold Spring Harb Perspect Med 2011;1:a006478.   3 Landskroner-Eiger S, Moneke I, Sessa WC: miRNAs as modulators of angiogenesis. Cold Spring Harb Perspect Med 2013;3:a006643.   4 Carmeliet P: Angiogenesis in life, disease and medicine. Nature 2005;438:932–936.   5 Carmeliet P, Jain RK: Molecular mechanisms and clinical applications of angiogenesis. Nature 2011; 473:298–307.   6 Oliver G, Alitalo K: The lymphatic vasculature: recent progress and paradigms. Annu Rev Cell Dev Biol 2005;21:457–483.

30

  7 Kim H, Kataru RP, Koh GY: Regulation and implications of inflammatory lymphangiogenesis. Trends Immunol 2012;33:350–356.   8 Folkman J, Merler E, Abernathy C, Williams G: Isolation of a tumor factor responsible for angiogenesis. J Exp Med 1971;133:275–288.   9 Folkman J, Brem H: Angiogenesis and Inflammation; in Gallin JI, Goldstein IM, Snyderman R (eds): Inflammation: Basic Principles and Clinical Correlates. New York, Raven Press, 1992, pp 821. 10 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 USA 1998; 95: 548–553. 11 Joukov V, Sorsa T, Kumar V, Jeltsch M, ClaessonWelsh L, Cao Y, Saksela O, Kalkkinen N, Alitalo K: Proteolytic processing regulates receptor specificity and activity of VEGF-C. EMBO J 1997;16:3898–3911.

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

References

25 Melillo RM, Guarino V, Avilla E, Galdiero MR, Liotti F, Prevete N, Rossi FW, Basolo F, Ugolini C, de Paulis A, Santoro M, Marone G: Mast cells have a protumorigenic role in human thyroid cancer. Oncogene 2010;29:6203–6215. 26 Ribatti D, Guidolin D, Marzullo A, Nico B, Annese T, Benagiano V, Crivellato E: Mast cells and angiogenesis in gastric carcinoma. Int J Exp Pathol 2010; 91:350–356. 27 Yano H, Kinuta M, Tateishi H, Nakano Y, Matsui S, Monden T, Okamura J, Sakai M, Okamoto S: Mast cell infiltration around gastric cancer cells correlates with tumor angiogenesis and metastasis. Gastric Cancer 1999;2:26–32. 28 Coussens LM, Raymond WW, Bergers G, Laig-Webster M, Behrendtsen O, Werb Z, Caughey GH, Hanahan D: Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev 1999;13:1382–1397. 29 Nakayama T, Yao L, Tosato G: Mast cell-derived angiopoietin-1 plays a critical role in the growth of plasma cell tumors. J Clin Invest 2004;114:1317–1325. 30 Gounaris E, Erdman SE, Restaino C, Gurish MF, Friend DS, Gounari F, Lee DM, Zhang G, Glickman JN, Shin K, Rao VP, Poutahidis T, Weissleder R, McNagny KM, Khazaie K: Mast cells are an essential hematopoietic component for polyp development. Proc Natl Acad Sci USA 2007;104:19977–19982. 31 Soucek L, Lawlor ER, Soto D, Shchors K, Swigart LB, Evan GI: Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors. Nat Med 2007;13:1211–1218. 32 Huang B, Lei Z, Zhang GM, Li D, Song C, Li B, Liu Y, Yuan Y, Unkeless J, Xiong H, Feng ZH: SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment. Blood 2008;112:1269–1279. 33 Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, Korets L, Lam J, Tawfik D, DeNardo DG, Naldini L, de Visser KE, De Palma M, Coussens LM: FcRgamma activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell 2010; 17:121–134. 34 Tucker T, Riccardi VM, Sutcliffe M, Vielkind J, Wechsler J, Wolkenstein P, Friedman JM: Different patterns of mast cells distinguish diffuse from encapsulated neurofibromas in patients with neurofibromatosis 1. J Histochem Cytochem 2011;59:584–590. 35 Chang DZ, Ma Y, Ji B, Wang H, Deng D, Liu Y, Abbruzzese JL, Liu YJ, Logsdon CD, Hwu P: Mast cells in tumor microenvironment promotes the in vivo growth of pancreatic ductal adenocarcinoma. Clin Cancer Res 2011;17:7015–7023. 36 Acikalin MF, Oner U, Topcu I, Yasar B, Kiper H, Colak E: Tumour angiogenesis and mast cell density in the prognostic assessment of colorectal carcinomas. Dig Liver Dis 2005;37:162–169.

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

31

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

12 Keyt BA, Berleau LT, Nguyen HV, Chen H, Heinsohn H, Vandlen R, Ferrara N: The carboxyl-terminal domain (111–165) of vascular endothelial growth factor is critical for its mitogenic potency. J Biol Chem 1996;271:7788–7795. 13 Maglione D, Guerriero V, Viglietto G, Delli-Bovi P, Persico MG: Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci USA 1991;88:9267– 9271. 14 Woolard J, Wang WY, Bevan HS, Qiu Y, Morbidelli L, Pritchard-Jones RO, Cui TG, Sugiono M, Waine E, Perrin R, Foster R, Digby-Bell J, Shields JD, Whittles CE, Mushens RE, Gillatt DA, Ziche M, Harper SJ, Bates DO: VEGF165b, an inhibitory vascular endothelial growth factor splice variant: mechanism of action, in vivo effect on angiogenesis and endogenous protein expression. Cancer Res 2004;64:7822–7835. 15 Augustin HG, Koh GY, Thurston G, Alitalo K: Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol 2009;10:165–177. 16 Fiedler U, Augustin HG: Angiopoietins: a link between angiogenesis and inflammation. Trends Immunol 2006;27:552–558. 17 Sakurai A, Doci CL, Gutkind JS: Semaphorin signaling in angiogenesis, lymphangiogenesis and cancer. Cell Res 2012;22:23–32. 18 Serini G, Bussolino F, Maione F, Giraudo E: Class 3 semaphorins: physiological vascular normalizing agents for anti-cancer therapy. J Intern Med 2013; 273:138–155. 19 Neufeld G, Kessler O: The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nat Rev Cancer 2008;8:632–645. 20 Wild JR, Staton CA, Chapple K, Corfe BM: Neuropilins: expression and roles in the epithelium. Int J Exp Pathol 2012;93:81–103. 21 Marone G, Triggiani M, Genovese A, De Paulis A: Role of human mast cells and basophils in bronchial asthma. Adv Immunol 2005;88:97–160. 22 Galli SJ, Tsai M: IgE and mast cells in allergic disease. Nat Med 2012;18:693–704. 23 Triggiani M, Giannattasio G, Calabrese C, Loffredo S, Granata F, Fiorello A, Santini M, Gelb MH, Marone G: Lung mast cells are a source of secreted phospholipases A2. J Allergy Clin Immunol 2009; 124: 558–565. 24 Marone G, Galdiero MR, Melillo RM: Mast cells and basophils in angiogenesis: bridging the gap between inflammation and tumors; in Marone G, Triggiani M, Genovese A (eds): Translational Science: from Basic to Clinical Immunology and Allergy. Pisa, Pacini Editore, 2012, pp 21–35.

32

50 Sumbayev VV, Yasinska I, Oniku AE, Streatfield CL, Gibbs BF: Involvement of hypoxia-inducible factor-1 in the inflammatory responses of human LAD2 mast cells and basophils. PLoS One 2012;7:e34259. 51 Voehringer D: Basophil modulation by cytokine instruction. Eur J Immunol 2012;42:2544–2550. 52 MacGlashan D, Jr., White JM, Huang SK, Ono SJ, Schroeder JT, Lichtenstein LM: Secretion of IL-4 from human basophils. The relationship between IL-4 mRNA and protein in resting and stimulated basophils. J Immunol 1994;152:3006–3016. 53 Ochensberger B, Daepp GC, Rihs S, Dahinden CA: Human blood basophils produce interleukin-13 in response to IgE-receptor-dependent and -independent activation. Blood 1996;88:3028–3037. 54 Patella V, Giuliano A, Bouvet JP, Marone G: Endogenous superallergen protein Fv induces IL-4 secretion from human Fc epsilon RI+ cells through interaction with the VH3 region of IgE. J Immunol 1998; 161:5647–5655. 55 Genovese A, Borgia G, Bjorck L, Petraroli A, de Paulis A, Piazza M, Marone G: Immunoglobulin superantigen protein L induces IL-4 and IL-13 secretion from human Fc epsilon RI+ cells through interaction with the kappa light chains of IgE. J Immunol 2003; 170:1854–1861. 56 Charles N, Hardwick D, Daugas E, Illei GG, Rivera J: Basophils and the T helper 2 environment can promote the development of lupus nephritis. Nat Med 2010;16:701–707. 57 de Paulis A, Prevete N, Fiorentino I, Rossi FW, Staibano S, Montuori N, Ragno P, Longobardi A, Liccardo B, Genovese A, Ribatti D, Walls AF, Marone G: Expression and functions of the vascular endothelial growth factors and their receptors in human basophils. J Immunol 2006;177:7322–7331. 58 Cerny-Reiterer S, Ghanim V, Hoermann G, Aichberger KJ, Herrmann H, Muellauer L, Repa A, Sillaber C, Walls AF, Mayerhofer M, Valent P: Identification of basophils as a major source of hepatocyte growth factor in chronic myeloid leukemia: a novel mechanism of BCR-ABL1-independent disease progression. Neoplasia 2012;14:572–584. 59 Granucci F, Zanoni I, Ricciardi-Castagnoli P: Central role of dendritic cells in the regulation and deregulation of immune responses. Cell Mol Life Sci 2008;65:1683–1697. 60 Noonan DM, De Lerma Barbaro A, Vannini N, Mortara L, Albini A: Inflammation, inflammatory cells and angiogenesis: decisions and indecisions. Cancer Metastasis Rev 2008;27:31–40. 61 Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M: Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2013; 229: 176– 185.

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

37 Boesiger J, Tsai M, Maurer M, Yamaguchi M, Brown LF, Claffey KP, Dvorak HF, Galli SJ: Mast cells can secrete vascular permeability factor/ vascular endothelial cell growth factor and exhibit enhanced release after immunoglobulin E-dependent upregulation of fc epsilon receptor I expression. J Exp Med 1998;188:1135–1145. 38 Abdel-Majid RM, Marshall JS: Prostaglandin E2 induces degranulation-independent production of vascular endothelial growth factor by human mast cells. J Immunol 2004;172:1227–1236. 39 Cao J, Papadopoulou N, Kempuraj D, Boucher WS, Sugimoto K, Cetrulo CL, Theoharides TC: Human mast cells express corticotropin-releasing hormone (CRH) receptors and CRH leads to selective secretion of vascular endothelial growth factor. J Immunol 2005;174:7665–7675. 40 Detoraki A, Staiano RI, Granata F, Giannattasio G, Prevete N, de Paulis A, Ribatti D, Genovese A, Triggiani M, Marone G: Vascular endothelial growth factors synthesized by human lung mast cells exert angiogenic effects. J Allergy Clin Immunol 2009; 123: 1142–1149. 41 Fraisl P, Mazzone M, Schmidt T, Carmeliet P: Regulation of angiogenesis by oxygen and metabolism. Dev Cell 2009;16:167–179. 42 Xia D, Wang D, Kim SH, Katoh H, DuBois RN: Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nat Med 2012;18:224–226. 43 Kalesnikoff J, Galli SJ: New developments in mast cell biology. Nat Immunol 2008;9:1215–1223. 44 Yang FC, Ingram DA, Chen S, Zhu Y, Yuan J, Li X, Yang X, Knowles S, Horn W, Li Y, Zhang S, Yang Y, Vakili ST, Yu M, Burns D, Robertson K, Hutchins G, Parada LF, Clapp DW: Nf1-dependent tumors require a microenvironment containing Nf1+/–- and c-kit-dependent bone marrow. Cell 2008; 135: 437– 448. 45 Detoraki A, Granata F, Staibano S, Rossi FW, Marone G, Genovese A: Angiogenesis and lymphangiogenesis in bronchial asthma. Allergy 2010; 65: 946– 958. 46 Rabenhorst A, Schlaak M, Heukamp LC, Forster A, Theurich S, von Bergwelt-Baildon M, Buttner R, Kurschat P, Mauch C, Roers A, Hartmann K: Mast cells play a protumorigenic role in primary cutaneous lymphoma. Blood 2012;120:2042–2054. 47 Eltzschig HK, Carmeliet P: Hypoxia and inflammation. N Engl J Med 2011;364:656–665. 48 Liao D, Johnson RS: Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev 2007;26: 281–290. 49 Jeong HJ, Oh HA, Nam SY, Han NR, Kim YS, Kim JH, Lee SJ, Kim MH, Moon PD, Kim HM: The critical role of mast cell-derived hypoxia-inducible factor-1alpha in human and mice melanoma growth. Int J Cancer 2012.

75 Bingle L, Brown NJ, Lewis CE: The role of tumourassociated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 2002;196:254–265. 76 Granata F, Frattini A, Loffredo S, Staiano RI, Petraroli A, Ribatti D, Oslund R, Gelb MH, Lambeau G, Marone G, Triggiani M: Production of vascular endothelial growth factors from human lung macrophages induced by group IIA and group X secreted phospholipases A2. J Immunol 2010;184:5232–5241. 77 Dusseau JW, Hutchins PM: Hypoxia-induced angiogenesis in chick chorioallantoic membranes: a role for adenosine. Respir Physiol 1988;71:33–44. 78 Pinhal-Enfield G, Ramanathan M, Hasko G, Vogel SN, Salzman AL, Boons GJ, Leibovich SJ: An angiogenic switch in macrophages involving synergy between Toll-like receptors 2, 4, 7 and 9 and adenosine A(2A) receptors. Am J Pathol 2003;163:711–721. 79 Macedo L, Pinhal-Enfield G, Alshits V, Elson G, Cronstein BN, Leibovich SJ: Wound healing is impaired in MyD88-deficient mice: a role for MyD88 in the regulation of wound healing by adenosine A2A receptors. Am J Pathol 2007;171:1774–1788. 80 Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, Tanzawa K, Thorpe P, Itohara S, Werb Z, Hanahan D: Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2000;2:737–744. 81 Chen JJ, Yao PL, Yuan A, Hong TM, Shun CT, Kuo ML, Lee YC, Yang PC: Up-regulation of tumor interleukin-8 expression by infiltrating macrophages: its correlation with tumor angiogenesis and patient survival in non-small cell lung cancer. Clin Cancer Res 2003;9:729–737. 82 Song X, Voronov E, Dvorkin T, Fima E, Cagnano E, Benharroch D, Shendler Y, Bjorkdahl O, Segal S, Dinarello CA, Apte RN: Differential effects of IL-1 alpha and IL-1 beta on tumorigenicity patterns and invasiveness. J Immunol 2003;171:6448–6456. 83 Kessenbrock K, Plaks V, Werb Z: Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 2010;141:52–67. 84 Hildenbrand R, Wolf G, Bohme B, Bleyl U, Steinborn A: Urokinase plasminogen activator receptor (CD87) expression of tumor-associated macrophages in ductal carcinoma in situ, breast cancer, and resident macrophages of normal breast tissue. J Leukoc Biol 1999;66:40–49. 85 Seo KH, Ko HM, Choi JH, Jung HH, Chun YH, Choi IW, Lee HK, Im SY: Essential role for platelet-activating factor-induced NF-kappaB activation in macrophage-derived angiogenesis. Eur J Immunol 2004; 34:2129–2137. 86 David Dong ZM, Aplin AC, Nicosia RF: Regulation of angiogenesis by macrophages, dendritic cells, and circulating myelomonocytic cells. Curr Pharm Des 2009;15:365–379.

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

33

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

62 Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D: Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 1996;87:3336–3343. 63 Sawano A, Iwai S, Sakurai Y, Ito M, Shitara K, Nakahata T, Shibuya M: Flt-1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte-macrophages in humans. Blood 2001;97:785–791. 64 Selvaraj SK, Giri RK, Perelman N, Johnson C, Malik P, Kalra VK: Mechanism of monocyte activation and expression of proinflammatory cytochemokines by placenta growth factor. Blood 2003;102:1515–1524. 65 Eubank TD, Galloway M, Montague CM, Waldman WJ, Marsh CB: M-CSF induces vascular endothelial growth factor production and angiogenic activity from human monocytes. J Immunol 2003;171:2637– 2643. 66 Clark AN, Youkey R, Liu X, Jia L, Blatt R, Day YJ, Sullivan GW, Linden J, Tucker AL: A1 adenosine receptor activation promotes angiogenesis and release of VEGF from monocytes. Circ Res 2007;101: 1130– 1138. 67 Poulin S, Thompson C, Thivierge M, Veronneau S, McMahon S, Dubois CM, Stankova J, Rola-Pleszczynski M: Cysteinyl-leukotrienes induce vascular endothelial growth factor production in human monocytes and bronchial smooth muscle cells. Clin Exp Allergy 2011;41:204–217. 68 Hill LM, Gavala ML, Lenertz LY, Bertics PJ: Extracellular ATP may contribute to tissue repair by rapidly stimulating purinergic receptor X7-dependent vascular endothelial growth factor release from primary human monocytes. J Immunol 2010;185:3028–3034. 69 Dirkx AE, Oude Egbrink MG, Wagstaff J, Griffioen AW: Monocyte/macrophage infiltration in tumors: modulators of angiogenesis. J Leukoc Biol 2006; 80: 1183–1196. 70 Allavena P, Mantovani A: Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment. Clin Exp Immunol 2012;167:195–205. 71 Gordon S, Martinez FO: Alternative activation of macrophages: mechanism and functions. Immunity 2010;32:593–604. 72 Mantovani A, Sica A, Locati M: Macrophage polarization comes of age. Immunity 2005;23:344–346. 73 Qian BZ, Pollard JW: Macrophage diversity enhances tumor progression and metastasis. Cell 2010; 141: 39–51. 74 Lamagna C, Aurrand-Lions M, Imhof BA: Dual role of macrophages in tumor growth and angiogenesis. J Leukoc Biol 2006;80:705–713.

34

  99 Hamrah P, Chen L, Zhang Q, Dana MR: Novel expression of vascular endothelial growth factor receptor (VEGFR)-3 and VEGF-C on corneal dendritic cells. Am J Pathol 2003;163:57–68. 100 Matarese G, Procaccini C, De Rosa V: At the crossroad of T cells, adipose tissue, and diabetes. Immunol Rev 2012;249:116–134. 101 Papatriantafyllou M: T cells: neuropilin 1 – distinguishing TReg cell subsets. Nat Rev Immunol 2012; 12:746. 102 Parker MW, Guo HF, Li X, Linkugel AD, Vander Kooi CW: Function of members of the neuropilin family as essential pleiotropic cell surface receptors. Biochemistry 2012;51:9437–9446. 103 Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, Xiong H, Dolpady J, Frey AB, Ruocco MG, Yang Y, Floess S, Huehn J, Oh S, Li MO, Niec RE, Rudensky AY, Dustin ML, Littman DR, Lafaille JJ: Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3+ T reg cells. J Exp Med 2012;209:1723–1742. 104 Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-Bucktrout S, Anthony BA, Sverdrup FM, Head R, Kuster DJ, Ruminski P, Weiss D, Von Schack D, Bluestone JA: Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med 2012;209:1713–1722. 105 De Palma M, Naldini L: Angiopoietin-2 TIEs up macrophages in tumor angiogenesis. Clin Cancer Res 2011;17:5226–5232. 106 Prevete N, Staiano RI, Granata F, Detoraki A, Necchi V, Ricci V, Triggiani M, de Paulis A, Marone G, Genovese A: Expression and function of angiopoietins and their Tie receptors in human basophils and mast cells. J Biol Reg Homeos Agents 2013;in press. 107 Puxeddu I, Alian A, Piliponsky AM, Ribatti D, Panet A, Levi-Schaffer F: Human peripheral blood eosinophils induce angiogenesis. Int J Biochem Cell Biol 2005;37:628–636. 108 Puxeddu I, Berkman N, Ribatti D, Bader R, Haitchi HM, Davies DE, Howarth PH, Levi-Schaffer F: Osteopontin is expressed and functional in human eosinophils. Allergy 2010;65:168–174. 109 Nissim Ben Efraim AH, Eliashar R, Levi-Schaffer F: Hypoxia modulates human eosinophil function. Clin Mol Allergy 2010;8:10. 110 Asai K, Kanazawa H, Kamoi H, Shiraishi S, Hirata K, Yoshikawa J: Increased levels of vascular endothelial growth factor in induced sputum in asthmatic patients. Clin Exp Allergy 2003;33:595–599. 111 Tseliou E, Bakakos P, Kostikas K, Hillas G, Mantzouranis K, Emmanouil P, Simoes D, Alchanatis M, Papiris S, Loukides S: Increased levels of angiopoietins 1 and 2 in sputum supernatant in severe refractory asthma. Allergy 2012;67:396–402.

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

87 Belardelli F, Ferrantini M: Cytokines as a link between innate and adaptive antitumor immunity. Trends Immunol 2002;23:201–208. 88 Del Vecchio M, Canova S, Messina A, Bajetta E: Impressive objective response in a patient with extensive metastatic melanoma including the brain. Melanoma Res 2007;17:332–334. 89 Kanegane C, Sgadari C, Kanegane H, Teruya-Feldstein J, Yao L, Gupta G, Farber JM, Liao F, Liu L, Tosato G: Contribution of the CXC chemokines IP10 and Mig to the antitumor effects of IL-12. J Leukoc Biol 1998;64:384–392. 90 Cao R, Farnebo J, Kurimoto M, Cao Y: Interleukin-18 acts as an angiogenesis and tumor suppressor. FASEB J 1999;13:2195–2202. 91 Tatsumi T, Huang J, Gooding WE, Gambotto A, Robbins PD, Vujanovic NL, Alber SM, Watkins SC, Okada H, Storkus WJ: Intratumoral delivery of dendritic cells engineered to secrete both interleukin (IL)-12 and IL-18 effectively treats local and distant disease in association with broadly reactive Tc1-type immunity. Cancer Res 2003;63:6378–6386. 92 Gorrin-Rivas MJ, Arii S, Mori A, Takeda Y, Mizumoto M, Furutani M, Imamura M: Implications of human macrophage metalloelastase and vascular endothelial growth factor gene expression in angiogenesis of hepatocellular carcinoma. Ann Surg 2000;231: 67–73. 93 Coffelt SB, Hughes R, Lewis CE: Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta 2009;1796:11–18. 94 Kerjaschki D: The crucial role of macrophages in lymphangiogenesis. J Clin Invest 2005;115:2316–2319. 95 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;115:2363–2372. 96 Bourbie-Vaudaine S, Blanchard N, Hivroz C, Romeo PH: Dendritic cells can turn CD4+ T lymphocytes into vascular endothelial growth factor-carrying cells by intercellular neuropilin-1 transfer. J Immunol 2006;177:1460–1469. 97 Dadabayev AR, Sandel MH, Menon AG, Morreau H, Melief CJ, Offringa R, van der Burg SH, Janssen-van Rhijn C, Ensink NG, Tollenaar RA, van de Velde CJ, Kuppen PJ: Dendritic cells in colorectal cancer correlate with other tumor-infiltrating immune cells. Cancer Immunol Immunother 2004;53:978–986. 98 Kishuku M, Nishioka Y, Abe S, Kishi J, Ogino H, Aono Y, Azuma M, Kinoshita K, Batmunkh R, Makino H, Ranjan P, Minakuchi K, Sone S: Expression of soluble vascular endothelial growth factor receptor-1 in human monocyte-derived mature dendritic cells contributes to their antiangiogenic property. J Immunol 2009;183:8176–8185.

124 Coffelt SB, Tal AO, Scholz A, De Palma M, Patel S, Urbich C, Biswas SK, Murdoch C, Plate KH, Reiss Y, Lewis CE: Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res 2010;70:5270–5280. 125 Gu H, Cui M, Bai Y, Chen F, Ma K, Zhou C, Guo L: Angiopoietin-1/Tie2 signaling pathway inhibits lipopolysaccharide-induced activation of RAW264.7 macrophage cells. Biochem Biophys Res Commun 2010;392:178–182. 126 Coffelt SB, Chen YY, Muthana M, Welford AF, Tal AO, Scholz A, Plate KH, Reiss Y, Murdoch C, De Palma M, Lewis CE: Angiopoietin 2 stimulates TIE2-expressing monocytes to suppress T cell activation and to promote regulatory T cell expansion. J Immunol 2011;186:4183–4190. 127 Hubbard NE, Lim D, Mukutmoni M, Cai A, Erickson KL: Expression and regulation of murine macrophage angiopoietin-2. Cell Immunol 2005; 234: 102–109. 128 Feingold KR, Shigenaga JK, Cross AS, Moser A, Grunfeld C: Angiopoietin like protein 4 expression is decreased in activated macrophages. Biochem Biophys Res Commun 2012;421:612–615. 129 Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT: Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 2006;441:231–234. 130 Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, Housseau F, Pardoll DM, Sears CL: A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 2009;15:1016–1022. 131 Maddur MS, Miossec P, Kaveri SV, Bayry J: Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol 2012;181:8–18. 132 Ikeda K, Nakajima H, Suzuki K, Kagami S, Hirose K, Suto A, Saito Y, Iwamoto I: Mast cells produce interleukin-25 upon Fc epsilon RI-mediated activation. Blood 2003;101:3594–3596. 133 Gaffen SL: Structure and signalling in the IL-17 receptor family. Nat Rev Immunol 2009;9:556–567. 134 Rohn TA, Jennings GT, Hernandez M, Grest P, Beck M, Zou Y, Kopf M, Bachmann MF: Vaccination against IL-17 suppresses autoimmune arthritis and encephalomyelitis. Eur J Immunol 2006; 36: 2857–2867. 135 Murugaiyan G, Saha B: Protumor vs. antitumor functions of IL-17. J Immunol 2009;183:4169–4175. 136 Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T, Robbins PD, Tahara H, Lotze MT: Interleukin-17 promotes angiogenesis and tumor growth. Blood 2003;101:2620–2627.

Immune Cells in Angiogenesis and Lymphangiogenesis Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

35

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

112 Feistritzer C, Kaneider NC, Sturn DH, Mosheimer BA, Kahler CM, Wiedermann CJ: Expression and function of the vascular endothelial growth factor receptor FLT-1 in human eosinophils. Am J Respir Cell Mol Biol 2004;30:729–735. 113 Feistritzer C, Mosheimer BA, Sturn DH, Bijuklic K, Patsch JR, Wiedermann CJ: Expression and function of the angiopoietin receptor Tie2 in human eosinophils. J Allergy Clin Immunol 2004;114:1077–1084. 114 Nissim Ben Efraim AH, Levi-Schaffer F: The roles of eosinophils in the modulation of angiogenesis; in Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 138–154. 115 Mantovani A, Cassatella MA, Costantini C, Jaillon S: Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol 2011;11:519–531. 116 Dumas E, Martel C, Neagoe PE, Bonnefoy A, Sirois MG: Angiopoietin-1 but not angiopoietin-2 promotes neutrophil viability: role of interleukin-8 and platelet-activating factor. Biochim Biophys Acta 2012;1823:358–367. 117 Lemieux C, Maliba R, Favier J, Theoret JF, Merhi Y, Sirois MG: Angiopoietins can directly activate endothelial cells and neutrophils to promote proinflammatory responses. Blood 2005;105:1523–1530. 118 Neagoe PE, Dumas E, Hajjar F, Sirois MG: Angiopoietin-1 but not angiopoietin-2 induces IL-8 synthesis and release by human neutrophils. J Cell Physiol 2012;227:3099–3110. 119 Sturn DH, Feistritzer C, Mosheimer BA, Djanani A, Bijuklic K, Patsch JR, Wiedermann CJ: Angiopoietin affects neutrophil migration. Microcirculation 2005;12:393–403. 120 Brkovic A, Pelletier M, Girard D, Sirois MG: Angiopoietin chemotactic activities on neutrophils are regulated by PI-3K activation. J Leukoc Biol 2007; 81:1093–1101. 121 Tecchio C, Cassatella MA: Neutrophil-derived cytokines involved in angiogenesis; in Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 123–137. 122 Venneri MA, De Palma M, Ponzoni M, Pucci F, Scielzo C, Zonari E, Mazzieri R, Doglioni C, Naldini L: Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood 2007;109:5276–5285. 123 Pucci F, Venneri MA, Biziato D, Nonis A, Moi D, Sica A, Di Serio C, Naldini L, De Palma M: A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood ‘resident’ monocytes, and embryonic macrophages suggests common functions and developmental relationships. Blood 2009;114:901–914.

144 Letuve S, Lajoie-Kadoch S, Audusseau S, Rothenberg ME, Fiset PO, Ludwig MS, Hamid Q: IL-17E upregulates the expression of proinflammatory cytokines in lung fibroblasts. J Allergy Clin Immunol 2006;117:590–596. 145 Hurst SD, Muchamuel T, Gorman DM, Gilbert JM, Clifford T, Kwan S, Menon S, Seymour B, Jackson C, Kung TT, Brieland JK, Zurawski SM, Chapman RW, Zurawski G, Coffman RL: New IL-17 family members promote Th1 or Th2 responses in the lung: in vivo function of the novel cytokine IL-25. J Immunol 2002;169:443–453. 146 Wang YH, Angkasekwinai P, Lu N, Voo KS, Arima K, Hanabuchi S, Hippe A, Corrigan CJ, Dong C, Homey B, Yao Z, Ying S, Huston DP, Liu YJ: IL-25 augments type 2 immune responses by enhancing the expansion and functions of TSLP-DC-activated Th2 memory cells. J Exp Med 2007;204:1837–1847. 147 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;161:947–956. 148 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;24:203–215. 149 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. 150 Lee SS, Joo YS, Kim WU, Min DJ, Min JK, Park SH, Cho CS, Kim HY: Vascular endothelial growth factor levels in the serum and synovial fluid of patients with rheumatoid arthritis. Clin Exp Rheumatol 2001;19:321–324. 151 Genovese A, Detoraki A, Granata F, Galdiero MR, Spadaro G, Marone G: Angiogenesis, lymphangiogenesis and atopic dermatitis. Chem Immunol Allergy 2012;96:50–60.

Prof. Gianni Marone Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research, University of Naples Federico II School of Medicine Via S. Pansini 5, IT–80131 Naples (Italy) E-Mail marone @ unina.it  

36

 

Loffredo · Staiano · Granata · Genovese · Marone Marone G, Granata F (eds): Angiogenesis, Lymphangiogenesis and Clinical Implications. Chem Immunol Allergy. Basel, Karger, 2014, vol 99, pp 15–36 (DOI: 10.1159/000353316)

Downloaded by: UCSF Library & CKM 169.230.243.252 - 3/1/2015 2:06:16 PM

137 Numasaki M, Watanabe M, Suzuki T, Takahashi H, Nakamura A, McAllister F, Hishinuma T, Goto J, Lotze MT, Kolls JK, Sasaki H: IL-17 enhances the net angiogenic activity and in vivo growth of human non-small cell lung cancer in SCID mice through promoting CXCR-2-dependent angiogenesis. J Immunol 2005;175:6177–6189. 138 Numasaki M, Lotze MT, Sasaki H: Interleukin-17 augments tumor necrosis factor-alpha-induced elaboration of proangiogenic factors from fibroblasts. Immunol Lett 2004;93:39–43. 139 Takahashi H, Numasaki M, Lotze MT, Sasaki H: Interleukin-17 enhances bFGF-, HGF- and VEGFinduced growth of vascular endothelial cells. Immunol Lett 2005;98:189–193. 140 Kim YS, Hong SW, Choi JP, Shin TS, Moon HG, Choi EJ, Jeon SG, Oh SY, Gho YS, Zhu Z, Kim YK: Vascular endothelial growth factor is a key mediator in the development of T cell priming and its polarization to type 1 and type 17 T helper cells in the airways. J Immunol 2009;183:5113–5120. 141 Kim YS, Choi SJ, Tae YM, Lee BJ, Jeon SG, Oh SY, Gho YS, Zhu Z, Kim YK: Distinct roles of vascular endothelial growth factor receptor-1- and receptor2-mediated signaling in T cell priming and Th17 polarization to lipopolysaccharide-containing allergens in the lung. J Immunol 2010; 185: 5648– 5655. 142 Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Page N, Olivenstein R, Elias J, Chakir J: IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J Allergy Clin Immunol 2001;108:430–438. 143 Fujie H, Niu K, Ohba M, Tomioka Y, Kitazawa H, Nagashima K, Ohrui T, Numasaki M: A distinct regulatory role of Th17 cytokines IL-17A and IL17F in chemokine secretion from lung microvascular endothelial cells. Inflammation 2012; 35: 1119– 1131.

Immune cells as a source and target of angiogenic and lymphangiogenic factors.

Angiogenesis and lymphangiogenesis are distinct and complex processes requiring a finely tuned balance between stimulatory and inhibitory signals. Imm...
2MB Sizes 0 Downloads 0 Views