• 沒有找到結果。

I. Introduction and literature review

1.4 PlGF and cancers

1.4.1. Placenta growth factor, PlGF

PlGF, shares 53% identity with the platelet-derived growth factor (PDGF)-like region of

VEGF (Nissen et al., 1998). The human PlGF gene has been mapped to chromosome 14q24 (Mattei

et al., 1996). PlGF is anticipated to have 149 amino acids and is encoded by 7 exons that span 800

kb (Roy et al., 2006). By alternative splicing, 4 forms of PlGF protein are generated: PlGF-1,

PlGF-2, PlGF-3, and PlGF-4 (Maglione et al., 2000). Only PlGF-2 is capable of binding heparin

(Hauser and Weich, 1993). Unlike VEGFA, which binds to both FLT1 and FLK1, PlGF binds to

FLT1 but not FLK1, and it also binds to neuropilin 1 and 2 (Persico et al., 1999; Migdal et al.,

1998)

1.4.2. Distribution of PlGF in tissues

PlGF is abundant in the trophoblasts of the placenta at the middle to late stages of pregnancy.

The amount of PlGF persistently increases toward the terminal development of the placenta, and its

41

biosynthesis seems to be limited to trophoblasts and stromal cells (Maglione et al., 1993; Hauser et

al., 1993). This temporal pattern of PlGF production may reflect a role in preventing excessive

neovascularization and overgrowth of the placenta tissue by downregulating angiogenesis. In

addition to the placenta, the gene is abundantly expressed in the thyroid under normal physiological

conditions (Viglietto et al., 1995).

1.4.3. Impact of PlGF gene knockout mice

Knockout of only one allele of the Vegfa gene, encoding VEGF-A, is lethal in the mouse

embryo (heterozygous lethality) with impaired angiogenesis and blood island formation leading to

developmental abnormalities (Ferrara et al., 1996; Carmeliet et al., 1996). In contrast, several

studies suggest that genetic deletion of either Vegfb or PlGF genes does not result in obvious

impairment of the vascular system (Luttun et al., 2002; Aase et al., 2001; Bellomo et al., 2001).

Although PlGF knockout mice under normal physiological conditions do not exhibit obvious

phenotypic changes, these animals show vascular defects under several pathological settings

(Luttun et al., 2002; Fischer et al., 2007; Carmeliet et al., 2001; Luttun et al., 2002). The response to

VEGF-A is also impaired, including reduced angiogenesis and vascular permeability under

ischemic insult in a hind limb model and recruitment of monocytes and macrophages in a skin

wound assay. Transplantation of wild-type bone marrow restores angiogenesis and collateral growth

in this knockout model under ischemic conditions, which suggests that PlGF may contribute to

42

vessel growth in adult animals through the mobilization of bone marrow–derived cells (Carmeliet et

al., 2001).

1.4.4 Mechanisms of PlGF

Several studies have analysed the molecular mechanisms of PlGF. This growth factor

stimulates angiogenesis, in part by enhancing VEGF signalling. It displaces VEGFA from FLT1,

which liberates VEGFA and allows it to activate FLK1 and enhance VEGF-driven angiogenesis

(Park et al., 1994). PlGF also upregulates the expression of VEGFA, fibroblast growth factor 2

(FGF2), platelet-derived growth factor-β (PDGFB), matrix metalloproteinases (MMPs), and other

angiogenic factors (Marcellini et al., 2006; Roy et al., 2005). Moreover, the binding of PlGF to

FLT1 leads to intermolecular crosstalk between FLT1 and FLK1, which amplifies FLK1 signalling

and consequently enhances VEGF-driven responses (Autiero et al., 2003). These effects of PlGF

suggest that endothelial cells enhance their own responsiveness to VEGFA by producing PlGF

(Autiero et al., 2003). However, PlGF is also capable of inducing its own signals through FLT1

independently of VEGFA. Indeed, gene expression profiling of endothelial cells has revealed that

PlGF signals directly through FLT1 and switches on a number of pro-angiogenic genes (Autiero et

al., 2003; Schoenfeld et al., 2004).

1.4.5. PlGF in primary human tumors

43

The first investigation conducted on PlGF expression in tumors was undertaken by Takahashi

et al. (1994), who demonstrated that PlGF is expressed in hypervascular renal cell carcinoma. PlGF

levels correlate with serosal invasion, lymph-node metastasis, tumor stage, and survival in gastric

cancer (Chen et al., 2004); with disease progression and survival in colorectal cancer (Wei et al.,

2005 ); with tumor stage in non-small cell lung carcinoma (Zhang et al., 2005 ); with recurrence,

metastasis and mortality in breast cancer (Parr et al., 2005); and with post-surgical early recurrence

of hepatocellular carcinoma (Ho et al., 2007). Plasma PlGF protein levels are also upregulated and

correlate with tumor grade and survival in patients with renal cell carcinoma (Matsumoto et al.,

2003). PlGF is not only produced by malignant cells, but also by endothelial cells, smooth muscle

cells, pericytes, cancer-associated fibroblasts, tumour-associated macrophages and various other

inflammatory cells in the tumor stroma (Fischer et al., 2007; Luttun et al., 2002; Carmeliet et al.,

2001; Yonekura et al., 1999). Tumor cells can also induce PlGF expression by fibroblasts via

crosstalk between tumor cells and the stroma. However, not all studies have found increased PlGF

levels in tumors. PlGF mRNA is downregulated in thyroid tumors (Viglietto et al., 1995), and is

undetectable in ovarian tumors (Sowter et al., 1997). PlGF expression is apparently lower in

prostatic tumors than in normal prostatic tissue (Matsumoto et al., 2003). A similar situation occurs

in the thyroid, where strong PlGF expression has been noted in normal tissue and its production is

decreased in thyroid tumors (Viglietto et al., 1995).

44

1.4.6 PlGF as a positive regulator of tumorigenesis and metastasis

In the transgenic PLGF-expressing mice, overexpression of PlGF in keratinocytes is also

linked to an increase in tumor growth, invasiveness, and the numbers and sizes of metastases in

animals inoculated intradermally with B16-BL6 melanoma cells (Marcellini et al., 2006).

Experiments with cultured cancer cell lines suggest that PlGF has a role in controlling cell motility

and invasiveness. In breast cancer cell lines, exogenous addition of PlGF-2, but not VEGF-A, to

culture medium stimulates two key characteristics associated with metastatic potential: motility

(detected by cell migration assay) and invasiveness (detected by Matrigel spheroid assay) (Taylor et

al., 2007). The PlGF-2–associated stimulation of motility and invasion is suppressed in an in vitro

system by the addition of a peptide that blocks the heparin-binding site of VEGFR1 or by an

antibody to PlGF (Taylor et al., 2007).

1.4.7 PlGF as a negative regulator of tumor growth angiogenesis.

Xu and colleagues (2006) reported that overexpression of PlGF impairs tumor growth in

xenograft models. In above study, a full-length PlGF-2 plasmid expression construct was stably

transfected into three human cell lines (lung carcinoma, colon carcinoma, and glioblastoma) that

produced high amounts of VEGF-A, and clones with the highest amounts of PlGF-2 were selected

for implantation (Xu et al., 2006). Although there was no effect on the growth rate of the cell lines

in culture, subcutaneous or orthotopic implantation of the PlGF-overexpressing cell lines into mice

45

revealed an inhibition of tumor growth and a reduction in tumor-associated angiogenesis. The

antagonism of VEGF-induced angiogenesis by production of PlGF within the same population of

cells has also been demonstrated in a murine model of fibrosarcoma (Eriksson et al., 2002).

VEGF-A–PlGF-1 heterodimers fails to activate VEGFR2-mediated signaling and fails to induce

angiogenesis in vitro and in vivo. Although overexpression of human PlGF-1 in murine

fibrosarcoma cells does not alter the growth rate of the cells in culture, forced production of PlGF-1

markedly reduces the rate of growth of tumors arising from subcutaneously implanted inoculates.

1.4.8 Epression of PlGF in non-cancerous disease

Pre-eclampsia is characterized by high blood pressure and elevated protein levels in urine

during pregnancy. In a normal pregnancy, angiogenesis, and vascular transformation lead to normal

placental development, whereas pre-eclamptic pregnancies are subject to abnormal angiogenesis

and vascular transformation. Since PlGF is a factor in angiogenesis, testing of the PlGF levels

between 15 and 18 weeks allows a doctor to determine whether a patient is at high risk, since the

PlGF level in a patient with pre-eclampsia will be lower than average PlGF levels during a normal

pregnancy (Schmidt et al., 2009). The new tests identify PlGF in blood earlier and more effectively

than do traditional testing measures.

The pathogenesis of chronic obstructive pulmonary disease (COPD) is hypothesized to result

from an imbalance of proteases and antiproteases in the lung. The expression of PlGF increases as a

46

response of airway epithelial cells to proinflammatory cytokines. The sustained stimuli of cytokines

and PlGF subsequently reduce VEGF expression and promote the apoptosis of airway epithelial

cells through VEGFR. The apoptosis of epithelial cells is considered essential for the pathogenesis

of pulmonary emphysema (Cheng et al., 2008).

As a risk-predicting biomarker in patients with acute coronary syndrome (ACS), placental

growth factor (PlGF) (Lenderink et al., 2006) was recently shown to be important in early and

advanced atherosclerotic lesions and coronary plaque rupture. This biomarker could be up-regulated

in the crucial mechanism of atherosclerosis in ACS patients (Heeschen et al., 2004) based on

subsequent platelet aggregation and systematic thrombosis, which may lead to the acute myocardial

infarction (AMI) or sudden cardiac death. Hence, as a member of the vascular endothelial growth

factor family (VEGF) (Carmeliet et al., 2001) PlGF may be a primary inflammatory instigator of

atherosclerotic plaque instability during the acute phase of ACS.

In our previous study, we had used an immunohistochemical technique to examine the

expression of PlGF in 100 specimens of oral squamous cell carcinoma (OSCC) (Cheng et al., 2010).

We found that the higher mean PLGF labeling index was significantly associated with OSCCs with

positive lymph node metastasis or with more advanced clinical stages. Positive lymph node

metastasis and PlGF labeling index > 40% are identified as independent unfavorable prognosis

factors. Several previous studies have reported an increased PlGF mRNA expression in tumor

tissues and increased serum PlGF protein levels in patients with several human cancers.Thus, in

47

this study we assessed whether the PlGF mRNA expression in OSCC tissues and serum PlGF

protein level in OSCC patients could be valuable biomarkers to predict the therapeutic effect,

progression, recurrence and prognosis of OSCC patients.

Specific goals:

1. To assess whether the expression of PlGF mRNA in OSCC tissues could be used to predict the

progression, recurrence and prognosis of OSCCs in Taiwan.

2. To evaluate whether the serum PlGF protein level in OSCC patients could be used to predict the

progression, recurrence and prognosis of OSCCs in Taiwan.This work was published by Oral

Oncology and the article is included as appendix B1.

48

相關文件