Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 1.
Published in final edited form as: Cell Signal. 2020 Jun 20;73:109698. doi: 10.1016/j.cellsig.2020.109698

Implications of the PAPP-A-IGFBP-IGF-1 pathway in the pathogenesis and treatment of polycystic kidney disease

Sonu Kashyap 1, Julianna D Zeidler 1, Claudia CS Chini 1, Eduardo Nunes Chini 1,*
PMCID: PMC7387202  NIHMSID: NIHMS1608225  PMID: 32569826

Abstract

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common genetic diseases implicated in the development of end stage renal disease (ESRD). Although FDA has recently approved a drug against ADPKD, there is still a great need for development of alternative management strategies for ADPKD. Understanding the different mechanisms that lead to cystogenesis and cyst expansion in ADPKD is imperative to develop new therapies against ADPKD. Recently, we demonstrated that caloric restriction can prevent the development of cystic disease in animal models of ADPKD and through these studies identified a new role for pregnancy associated plasma protein-A (PAPP-A), a component of the insulin-like growth factors (IGF) pathway, in the pathogenesis of this disease. The PAPP-A-IGF pathway plays an important role in regulation of cell growth, differentiation, and transformation and dysregulation of this pathway has been implicated in many diseases. Several indirect studies support the involvement of IGF-1 in the pathogenesis of ADPKD. However, it was only recently that we described a direct role for a component of this pathway in pathogenesis of ADPKD, opening a new avenue for the therapeutic approaches for this cystic disease. The present literature review will critically discuss the evidence that supports the role of components of IGF pathway in the pathogenesis of ADPKD and discuss the pharmacological implications of PAPP-A-IGF axis in this disease.

Keywords: Autosomal dominant polycystic kidney disease (ADPKD), Pregnancy associated plasma protein-A (PAPP-A), Insulin like growth factors (IGFs), Insulin growth factor binding protein (IGFBP), metalloproteases

Introduction

Autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases is caused by mutations in PKD1 and PKD2 genes which encodes polycystin 1 (PC1) and polycystin 2 (PC2) proteins respectively [1]. The hyperproliferation of tubular epithelial cells causes cyst formation and the cyst expansion eventually leads to the kidney enlargement which often results in ESRD [2]. Although FDA has recently approved tolvaptan drug against ADPKD to slow the progression of cystic disease, there are several adverse effects of this drug that make it unsuitable for many patients [3]. Therefore, the management strategies for ADPKD still remain largely supportive and limited. It is extremely important to understand the different molecular mechanisms that lead to cystogenesis and cyst expansion in order to develop new therapeutic strategies for management of ADPKD. Many signaling pathways have been reported to be involved in the pathogenesis of ADPKD including cyclic AMP (cAMP), MAPK/ERK, mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK) and growth factors [46]. The present review will explore the role of components of IGF pathway in ADPKD and its potential therapeutic implications. In particular, we will discuss the complexity of IGF pathway and the regulation of IGF bioavailability by the metalloproteinase, pregnancy associated plasma protein-A (PAPP-A).

The pathogenesis of ADPKD

Mutation in PKD1 gene is responsible for about 80% ADPKD patients, whereas PKD2 gene mutation accounts for 15% cases [7]. ADPKD patients generally carry a germline mutation in one allele of either PKD1 or PKD2, and second hits like somatic inactivation of the remaining wild-type PKD1 or PKD2 allele or loss of heterozygosity should occur in order to initiate the cyst formation [8]. The activity of the polycystins complex is thought to be necessary to prevent the cell-autonomous renal epithelial cell cystogenesis and when the level of functional PC1 or PC2 is reached below a certain critical threshold level, it leads to cystogenesis [8, 9]. The polycystins form multimeric protein complexes that are involved in regulation of components of various signaling cascades including Ca2+, cAMP and mTOR. The reduction in functional PC1 or PC2 therefore leads to several cellular alterations in cystic epithelial cells including cellular processes like fluid transport, proliferation, apoptosis, cell adhesion and differentiation [5, 1012]. Reduction in Ca2+ influx, induction in cAMP levels, and abnormal activation of MAPK/ERK pathway in renal epithelial c ells are critical mediators of cyst growth and expansion [9]. Other mechanisms that have also been shown to be involved in cyst growth include dysregulated signaling of heterotrimeric G proteins, mTOR, phosphoinositide 3-kinase (PI3K)/Akt, AMPK, Janus kinase -signal transducer and activator of transcription (JAK/STAT) and nuclear factor-κB (NF-κB) pathway [1316]. Additionally, metabolic alterations including defective glucose metabolism, impaired beta-oxidation, and abnormal mitochondrial activity are also shown to be associated with cyst expansion [1722].

Therefore, cyst expansion is an important factor in the pathogenesis of ADPKD which results in compression and damage of surrounding tissue, and eventually leads to inflammation, fibrosis and kidney failure [4]. It is clear that the expansion of cysts involves several mechanisms including dysregulation in cell proliferation processes. Therefore, the possibility of involvement of growth factors pathways cannot be denied in pathogenesis of ADPKD. In fact, epidermal growth factors (EGF), fibroblast growth factor 23 (FGF23) and insulin like growth factors (IGFs) have been reported to be involved in ADPKD [2326]. However, to date the specific role and mechanisms of dysregulation of growth factor pathways in ADPKD have not been elucidated. In particular, the role of components of the IGF-1 pathway in ADPKD is largely unknown.

IGF-1 pathway in ADPKD

Circulating IGF-1 is mainly produced in liver and exerts endocrine effects, but locally produced IGF-1 acts in an autocrine/paracrine manner. Therefore, IGFs can function as circulating hormones as well as tissue growth factors. The effects of both IGF-1 and IGF-2 are largely mediated through the IGF-1 receptor. Interaction of IGF-1 with IGF-1R triggers PI3K-Akt and mitogen-activated protein kinase (MAPK) pathways which regulate processes like cellular metabolism, apoptosis, cell adhesion, and angiogenesis [27, 28]. Since these pathways have also been shown to be involved in ADPKD pathogenesis, understanding the exact role of the IGF pathway and its components in ADPKD could identify potential new therapeutic targets for this cystic disease. The IGF-1 pathway is a complex growth factor pathway with multiple regulatory components [29]. It plays a key role in several physiological and pathological conditions such as aging, cancer, and tissue growth [3034]. While it has previously been speculated that IGF-1 may play a role in ADPKD [3540], no direct causal or mechanistic in vivo data implicating a role for any of the components of the IGF1 pathway in ADPKD can be found in the literature. In fact, the role of components of the IGF-1 pathway had never been directly tested in ADPKD until recently [41].

Components of the IGF pathway

As mentioned above, IGF pathway is a complex system consisting of two peptides: IGF-1 and IGF-2, two receptors: IGF-1 and −2 receptors (IGF-1R and IGF-2R), six IGF-binding proteins (IGFBP-1–6) and IGFBP proteases (Figure 1).

Figure-1. IGF system and its components.

Figure-1.

IGF system consists of Insulin like growth factors (IGFs), IGF binding proteins (IGFBPs), IGF1 and 2 receptors (IGF1R and IGF2R) and proteases. ALS: Acid labile subunit.

IGF ligands

IGFs are crucial for normal growth and development. They promote cell proliferation, differentiation and survival and also exert insulin-like metabolic effects in a wide range of cells and tissues. Although, hepatocytes are the main source of circulating IGF-1, IGFs are ubiquitously expressed on cells and have multiple endocrine, autocrine and paracrine effects [42]. IGF-I production is stimulated by growth hormone (GH) and insulin. IGF-I can also be released independent of GH. IGF-1 and IGF-2 belong to the same family and were first characterized as possessing insulin like activity [43]. IGFs share nearly 50% homology with insulin. In humans, Igf1 gene is located on the chromosome 12q23 [44]. It has 6 exons and four transcriptional variants that are originated by alternative splicing [45], generating different isoforms for this gene, namely IGF-IEa (MGF), IGF-IEb, and IGF-IEc. These isoforms are differentially regulated, for instance, in response to exercise or muscle regeneration [46, 47]. Igf2 gene is located on chromosome 11, possesses 9 exons and also gives rise to multiple mRNA variants and three isoforms, which have tissue specific expression [45, 48]. This transcriptional diversity contributes to IGF-1 and IGF-2 regulation, but the mechanisms involved are still mostly unknown. IGF-1 plays an important role in aging, fetal development, growth during childhood and adolescence, and tissue homeostasis. On the other hand, IGFs are risk factors in obesity and cancer due to the potent proliferative activity [31, 34, 49, 50].

IGF-1 receptors

IGFs exert their effects by interacting with the specific cell surface receptors known as IGF-1R and IGF-2R. IGFRs are tyrosine kinase receptors that are extensively expressed on mammalian tissues [5153]. IGF-1R shares approximately 60% homology with the insulin receptor (IR) and a number of their downstream molecules are the same, including insulin substrate-1, PI3K, protein kinase B (PKB/Akt), mTOR and p70S6 kinase. IGF-1R is a heterotetrameric receptor comprising two α and two β subunits (α2β2). Similar to the IR, IGFRs require a series of post-translational modifications such as glycosylation, disulfide linkage, and proteolytic cleavage before reaching their mature form consisting of two α subunits, containing the ligand biding site, and two β subunits, consisting the transmembrane and tyrosine kinase domains [54, 55]. Upon binding to IGFs, IGFR undergoes conformational changes that bring β subunits to close proximity triggering trans-autophosphorylation of the β subunits and activating the cytoplasmic tyrosine kinase domain. IGF-2 also binds to IGF-1R but with lower affinity compared with IGF-1. The IGF-2R, also known as the cation independent mannose 6-phosphate receptor, is a large single transmembrane protein with no homology with the IGF-IR. It binds to IGF-2 with higher affinity compared to IGF-1 [56]. The signaling pathways activated by the IGF-2R are not well defined. Most of the biological actions of IGF-2 are believed to be mediated by IGF-IR.

IGF-binding proteins

IGF-biding proteins (IGFBPs) are the important members of the IGF system and association between IGFs and IGFBPs is a major regulatory step in the IGF signaling pathway [57]. These proteins are present in the extracellular space as well as in the circulation and possess high affinity to IGFs [58, 59]. Over 95% of IGFs in serum are bound to IGFBPs. As the affinity of IGFBPs for IGFs is higher than the affinity of IGF to the cell -surface IGF-IR, IGFs which are bound to IGFBPs do not bind to IGF-IR. Thus, IGFBPs, in addition to stabilizing IGF by increasing its half-life in tissues and blood, play an important role in preventing the interaction between IGFs and their receptors for downstream signaling [57]. For example, in vascular smooth muscle cells (VSMCs), IGFBP-4 binds to IGF-1 and blocks its interaction with the IGF1R inhibiting IGF-1-stimulated DNA synthesis [60]. In addition, liver specific deletion of IGF-1 resulted in 80% reduction in circulating IGF-I, but did not change postnatal growth, indicating the importance of local IGF-1 [61].

Six members of IGFBP family, IGFBP-1 to IGFBP-6 have been identified [57]. IGFBPs share structurally similar features such as a three-domain structure cysteine-rich C and N-terminals, which are stabilized by multiple disulfide bounds, with a central linker domain in between [62]. Both N- and C- terminal domains are highly conserved across IGFBP family and form the IGF-binding site. Their structural domains include binding sites to components of the extracellular matrix, proteolytic cleavage sites, and sites for post-translational modifications [57, 62]. Although structurally similar, the different IGFBP proteins have distinct functions and their expression and mechanisms of regulation are cell and tissue specific. For instance, IGFBP-3, and to some extent IGFBP-5, can form a ternary complex of 150 kDa with IGF and the glycoprotein acid labile subunit (ALS) [63, 64]. ALS stabilizes this complex extending its half-life in the circulation; therefore this association is important to keep IGF in the circulation for a longer period of time compared to its binary complex due to its high molecular size. While the majority of circulating IGFBP-3 and IGFBP-1 are produced in the liver, IGFBP-3 and other IGFBPs are also expressed in many peripheral tissues [57]. Hepatic expression is regulated by different stimuli e.g. IGFBP-3 expression is regulated in response to GH stimulation [65, 66], whereas IGFBP-1 expression is increased by starvation, hypoxia, and glucocorticoids and decreased in response to insulin [6770]. In some circumstances, some IGFBPs may also potentiate IGF actions. For instance, when associated with extracellular matrix components, IGFBP-5 affinity to IGF can decrease upon biding to some components of the extracellular matrix which further regulates the effects of IGF-1 [71, 72]. Interestingly, IGFBPs also have IGF-independent functions like cell migration promotion by binding to cell surface proteins such as integrins [73, 74]; antiangiogenic activity promoted by IGFBP-6 interaction with vascular endothelial growth factor (VEGF) [75]; and transcriptional transactivator functions in the nuclei [7678].

In addition, IGFBP-related proteins (IGFBP-rP1–9) have been identified, which have several fold lower binding affinity for IGFs than IGFBPs, but their physiological and pathophysiological functions are still not clear [79].

IGFBP proteases

IGFBP proteases cleave IGFBPs into fragments that have lower affinity for IGFs. The reduced affinity of IGFBP fragments towards IGFs leads to the increased bioavailability of IGFs and thus activates IGF receptors and downstream signaling. Although, there are three major groups of proteases which cleave IGFBPs and modulate the availability of free IGF, consisting serine proteases, cathepsins and matrix metalloproteinases [8085] (Table 1), this review will mainly focus on metalloproteinase, PAPP-A.

Table 1.

Major groups of IGFBP proteases and their substrates.

Enzyme IGFBPs
Serine proteinases Prostate specific antigen (PSA) [80, 154] IGFBP-3,5
Kallikreinlike proteinase, Y-nerve growth factor [80, 155] IGFBP-3,4, 6
Plasmin [156, 157] IGFBP-1, 3, 5
Complement protein 1S (C1s) [83] IGFBP-5
Thrombin [158] [156] IGFBP-5, 3
Matrix Metalloproteinases Pappalysins [88, 90, 92, 159] PAPP-A IGFBP-2, 4, 5
PAPP-A2 IGFBP-3, 5
Adamalysins [84, 160, 161] IGFBP-3, 5
Matrixins/MMPs [80, 162165] IGFBP-2, 3, 5,
Cathepsins Cathepsin D [166169] IGFBP 1–5

Pappalysins are proteases in the metzincin superfamiliy of metalloproteinases, and pregnancy associated plasma protein-A (PAPP-A), alias pappalysin-1, is the founding member of this protease family [86, 87]. PAPP-A specifically cleaves IGFBP4 in an IGF-dependent manner [88] and may be the only protease responsible for IGFBP-4 cleavage under physiological condition [89]. Although IGFBP-4 is the main substrate of PAPP-A, it has also been reported to cleave IGFBP-5 [90] and IGFBP-2 [91] in an IGF-independent manner. PAPP-A2 a paralog of PAPP-A, is the second member of pappalysin family and cleaves IGFBP-5 and IGFBP-3 in an IGF-independent manner [92].

Role of PAPP-A in regulation of local IGF signaling in tissues

PAPP-A was first identified as one of the placental proteins present at higher levels in serum of pregnant women [93, 94]. During pregnancy, it is primarily produced by placental syncytiotrophoblasts and secreted into the circulation, increasing its levels from early detection at first trimester until delivery at term. Initially, despite being recognized as a clinically relevant marker used in screening for Down’s syndrome [95], PAPP-A was considered a pregnancy protein with no physiological function. In 1990s, several studies reported the novel IGF-dependent protease activity of PAPP-A towards IGFBP-4 in cell culture media from various cell types [9698], however, Lawrence et al were first to prove that PAPP-A was the protease responsible for the IGF-dependent protease activity towards IGFBP-4 [88]. Subsequently, PAPP-A was reported to specifically cleave IGFBP-4 in human, ovine, bovine, equine, and porcine ovarian follicular fluid and shown to be expressed in granulosa cells of these mammals and in culture media from osteoblasts, lung cells, smooth muscle cells [99] [100104]. Over the past years, PAPP-A has been reported to be ubiquitously expressed in multiple tissues [86, 94] and shown to have important roles in several physiological functions outside of pregnancy [105, 106].

PAPP-A exists in two major forms. During pregnancy, it circulates as a disulfide bound 500kDa heterotetrameric complex consisting of 2 PAPP-A subunits covalently bound to two proform of eosinophil major basic protein (pro-MBP) [107] [105]. ProMBP is the endogenous inhibitor of PAPP-A, thus in this complex, PAPP-A is proteolytic inactive. proMBP recognizes cell bound PAPP-A and readily forms a complex which is unable to bind to cell surface, thus it enters the circulation [108]. On the other hand, the non-pregnant form of PAPP-A is a proteolytically active homodimer not covalently linked with pro-MBP.

Human PAPP-A sequence is comprised of 1547 amino acid residues with a distinct set of protein modules (Figure 2). In each subunit, at N-terminal, there is a 250-residue laminin G-like module with unspecified function followed by a 350 residue proteolytic domain containing the so-called elongated zinc binding consensus sequence and a short sequence for Met-turn formation, which are the characteristics of the metzincin super family of metalloproteinases [87]. The proteolytic domain is then followed by two regions M1 and M2 which are not very well characterized yet. The C-terminal module has five short consensus repeat (SCR) modules or complement control protein (CCP) modules, SCR 1–5. SCR3 and SCR4 are responsible for binding to glycosaminoglycans (GAGs) present on cell surfaces, enabling interactions between PAPP-A and cell surfaces [86]. PAPP-A also contains three Lin12-Notch repeat (LNR) modules, and each binds a calcium ion and determines the proteolytic specificity. LNR1–2 is present in the proteolytic domain and LNR-3 is in the C domain.

Figure-2. Schematic representation of primary structure of PAPP-A subunit.

Figure-2.

At N-terminal, there is a 250-residue laminin G-like module with unspecified function followed by a 350 residue proteolytic domain containing the so-called elongated zinc binding consensus sequence and a short sequence for Met-turn formation. The proteolytic domain is followed by two ill-defined regions M1 and M2. The C-terminal module has five short consensus repeat (SCR) modules or complement control protein (CCP) modules, SCR 1–5. SCR3 and SCR4 are responsible for binding to glycosaminoglycans (GAGs) present on cell surfaces, enabling interactions between PAPP-A and cell surfaces. PAPP-A also contains three Lin12-Notch repeat (LNR) modules, and each binds a calcium ion and determines the proteolytic specificity. LNR1–2 is present in the proteolytic domain and LNR-3 is in the C domain.

PAPP-A proteolytic activity cleaves IGFBP-4 at Met-135/Lys-136 in the linking domains and this cleavage only happens when IGFBP-4 is bound to IGF-1, thus showing its dependency on IGF-1. N- and C-terminal domains of IGFBPs have reduced affinity for IGFs, therefore this cleavage of IGFBP-4 by PAPP-A allows dissociation of bound IGF and increases its local bioavailability [109]. Proteolytically active PAPP-A is cell surface bound; therefore cleavage of IGFBP-4 happens in close proximity to IGF1R and released bioactive IGFs can interact with its receptors and trigger downstream signaling. This in turn enhances the effects of IGFs on cell proliferation, survival, and differentiation. Therefore, proteolysis of IGFBP-4 by cell surface bound PAPP-A is the final modulating step delivering IGF to its receptor.

The effects of PAPP-A are reported to be mainly local. For example, PAPP-A deficient mice, which are about 40% smaller than their wild-type littermates, showed no proteolytic activity against IGFBP-4 and no changes in circulating IGF levels, suggesting that PAPP-A plays a role in the regulation of autocrine/paracrine (but not endocrine) effects of IGF and local IGF bioavailability [89]. Furthermore, transgenic mice over-expressing PAPP-A in skeletal muscle and osteoblasts have increased skeletal muscle mass and bone formation respectively [110, 111]. However, dual overexpression of a protease-resistant IGFBP-4 along with PAPP-A completely diminished these anabolic effects of PAPP-A in mice [112] indicating that an increase in local IGF bioavailability through IGFBP proteolysis is the primary reason for the anabolic effects caused by PAPP-A. In summary, PAPP-A is not a direct inhibitor of IGF signaling, but acts as an important regulator in local bioavailability in IGF through proteolysis of IGFBP-4. PAPPA activity can be regulated by different protein binding inhibitors such as stanniocalcins (STNC). Both STNC1 and 2 have been shown to bind to PAPP-A and inhibit its proteolytic activity providing an extra layer of regulation for the IGF system [113].

PAPP-A expression in cultured cells is shown to be upregulated by proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 [114, 115]. PAPP-A is also upregulated by TGFβ, IL-4, as well as by the cAMP-inducing agent forskolin and other factors [106].

Several lines of evidence indicate an important role for PAPP-A in a number of physiological and pathological conditions. PAPP-A plays a crucial role during pregnancy [103, 116] by regulating the IGF bioavailability. The IGFs play an important role in regulating fetal growth [117] [118], autocrine and paracrine control of trophoblast invasion of decidua and early development and vascularization of placenta [119]. Thus, during pregnancy, high levels of un-complexed PAPP-A could be required locally for placental development, whereas in circulation PAPP-A:proMBP complex inhibits the proteolytic activity of PAPP-A which would otherwise lead to unusual systemic higher IGFBP-4 proteinase activity. Low levels of PAPP-A in maternal plasma during the first trimester have been linked with Down syndrome, as well as many abnormal pregnancy outcomes including pregnancy induced hypertensive disorders, premature delivery, and still birth [120122]. Low levels of PAPP-A in pregnancy can be associated with high IGFBPs and consequently less availability of free IGF. Therefore, reduced availability of free IGF that can interact with its receptor could lead to abnormal pregnancy outcome. Outside pregnancy, PAPP-A expression has been shown to be upregulated in vascular injury models in pigs [101], and mice [123] and also in healing skin wounds in human [124] indicating a role for PAPP-A in vascular injury and tissue remodeling. PAPP-A also plays an important role in healthy ovarian follicular development [99, 125, 126] and regulates prenatal or postnatal growth and skeletal muscle formation [110]. In addition, PAPP-A has also been implicated in the pathogenesis of many diseases like atherosclerosis and cancer, as well as age-related disorders [127133]. Plasma PAPP-A is present at higher levels in patients on dialysis, and is an independent predictor of mortality of patients on hemodialysis [134, 135]. Elevated PAPP-A levels have also been associated with diabetes [136] and cerebrovascular diseases [137].

What is known about IGF pathway in ADPKD.

Several indirect lines of evidence over the years have indicated that the IGF-I could be involved in progression of cystic lesions in ADPKD (Table 2). Initially, a study by Nakamura et al [35] had reported that renal Igf1 mRNA expression increased progressively with age in the DBA/2FG-pcy mouse model of PKD compared to the control DBA/2 mice. The authors suggested that decreased mRNA expression of renal epidermal growth factor and increased mRNA expression of IGF-l, PCNA, TGFβ, PDGF-A and PDGF-B chains as well as bFGF might be involved in progression of cystic disease in DBA/2FG-pcy mice. Later, Aukema and Housini also reported that in Han:SPRD-cy rat model of PKD, IGF-1 levels were 32 to 76% higher in kidneys of cy/+ (heterozygotes for PKD mutation) compared with control rats (+/+). Treatment with dietary soy protein delayed the progression of cystic disease and reduced the kidney IGF-1 levels in male and female cy/+ rats [36]. Both of these studies indicated that the levels of IGF-1 are increased in rodent ADPKD models.

Table 2.

Evidences in support for the role of IGF-1 pathway in pathogenesis of autosomal dominant polycystic kidney disease (ADPKD).

Study ADPKD model Findings
1 Nakamura et al. 1993 [35] DBA/2FG-pcy mice
  • Increase in IGF-1 mRNA

2 Aukema et al. 2001 [36] Han:SPRD-cy rat
  • IGF-1 levels higher in cy/+

  • Dietary soy protein delays the disease progression

3 Parker et al. 2007 [38] Conditional immortalized cystic cells
  • IGF-1 induces hyperproliferation via ras/raf pathway

4 Song et al. 2009 [37] Cysts of different sizes and minimally cystic tissue from human ADPKD kidney
  • Upregulation of IGF-1/IGFR1

5 Liu et al. 2013 (39)[170] Immortalized epithelial cells from ADPKD patients
  • Rosiglitazone inhibits IGF-1-induced cyst lining epithelial cell proliferation

6 Warner et al. 2016 [40] Pkd1RC/RC mice
  • Food restriction reduced the serum IGF-1 levels

  • Food restriction also reduced renal Igf 1 mRNA expression.

7 Kashyap et al. 2020 [41] Human ADPKD kidney tissue sections and cystic fluids, 9–12 ADPKD tubular epithelial cells,
Pkd1RC/RC, Pkd2WS25/−, Pkd1RC/RC-Pappa mutant mice and metanephros
  • PAPP-A significantly higher in human and experimental ADPKD

  • PAPP-A genetic deficiency as well as antibody treatment ameliorates the cystic disease.

Using the conditionally immortalized tubular epithelial cells from human ADPKD patients with defined germline PKD1 mutations, Parker et al showed that polycystin-1 deficiency was linked to increased sensitivity to IGF-1. In cystic cells, IGF-1 stimulated cell proliferation in a dose-dependent manner (38). These authors also showed that IGF-1, as well as cAMP, stimulated proliferation in these cystic cells in a PI3K- and ERK-activity dependent manner. Higher IGF-1-stimulated GTP-Ras levels were observed in PKD1 cystic cells compared to control cells, indicating that PC1 deficiency could lower the threshold for activation of Ras-Raf-mediated signaling; leading to IGF induced hyper proliferation.

A later study employed a systems biology approach to explore the growth-modulating gene pathways in renal cyst growth in ADPKD [37]. Song et al performed global gene profiling on cysts of different sizes and minimally cystic tissues (MCT) from five PKD1 human polycystic kidneys and used gene set enrichment analysis to identify the dysregulated signaling pathways, as well as key transcription factors, between cysts and MCT. They observed that human PKD1 cysts showed downregulation of kidney epithelial differentiation genes and up-regulation of developmental and mitogenic signaling pathways. Interestingly, the up-regulation of IGF-1/IGF-1R pathway was observed along with Wnt/β-catenin, G-protein-coupled receptor signaling, and was associated with renal cystic growth. This further supported the role of the IGF axis in renal cyst growth.

In addition to these studies, Liu et al [39] also reported that in cell culture studies, IGF-1 increased the cystic epithelial cells growth by 15–20% in a dose-dependent manner compared to normal cortical tubular epithelia cells. Rosiglitazone, a thiazolidinediones (TZD) inhibited the proliferation-inducing activity of IGF-1 in cystic cells, in part by inhibiting the IGF-1-induced p70S6K activation.

In a previous study, we showed that food restriction ameliorates the cystic disease in murine models of ADPKD [40]. Interestingly, in that study we also found that food restriction, which significantly reduced the cystic disease, also reduced the serum IGF-1 levels, as well as renal Igf1 mRNA expression, indicating a potential role for IGF-1 in the pathogenesis of ADPKD.

All these studies together indirectly support the idea that the IGF-1 pathway is involved in ADPKD. However, because the IGF system is complex and consists of several components, it is imperative to determine which components of the IGF pathway are involved in the pathogenesis of PKD. It wasn’t until recently that a direct role for any component of the IGF pathway was clearly demonstrated in ADPKD [41] (Table 2). In fact, our recent work demonstrated that IGF-1 and other components of this pathway are upregulated in ADPKD, and reported that the metalloproteinase PAPP-A plays a crucial role in ADPKD pathogenesis by increasing the local bioavailability of IGF-1 [41].

PAPP-A as a therapeutical target in ADPKD

As mentioned above, several studies provided indirect evidences that the IGF-1 pathway is involved in pathogenesis of ADPKD. Our recent study, for the first time, evaluated different IGF pathway components in ADPKD and demonstrated that PAPP-A plays a central role in pathogenesis of ADPKD. A significant increase in gene expression of several components of IGF-1 pathway was observed in ADPKD, including Igf1, Igf1r, and Igfbp5. Interestingly, the greatest upregulation was observed in the expression of Pappa, and this increase in Pappa was specific to kidney [41]. The increased renal Pappa expression was observed in several murine models of PKD [41]. To assess if PAPP-A expression was induced in human, we analyzed cystic fluids, as well as kidney tissues from ADPKD patients. Indeed, PAPP-A was significantly higher in human cystic fluid, and kidney tissues showed stronger expression of PAPP-A in cystic epithelia and renal tubules compared to normal kidney. Additionally, ADPKD cystic epithelial cells 9–12, derived from ADPKD patients, also showed higher PAPP-A expression compared to normal renal cortical tubular epithelial cells (RCTE), clearly showing that upregulation of PAPP-A was a common feature of several ADPKD models. Interestingly, PAPP-A expression was reduced in food restricted (FR) mice that shown reduced cystic disease. Forskolin (FSK), a cAMP inducing agent, significantly induced the PAPP-A expression in 9–12 cells in a time- and dose- dependent manner. Interestingly, we also reported that the expression of PAPP-A in these models was positively regulated by the cAMP/CREB/CBP/p300 pathway (Figure 3) [41].

Figure 3. Role of PAPP-A in pathogenesis of ADPKD.

Figure 3.

Higher levels of PAPP-A in ADPKD leads to increased proteolytic activity towards IGF bound IGFBP-4; which consequently results in the increased bioavailability of IGF. This free IGF leads to interaction with its receptor, IGF1R and cause the activation of downstream signaling. Inhibition of PAPP-A provides the novel therapeutic target in ADPKD. PAPP-A: pregnancy associated plasma protein, cAMP: cyclic AMP, CREB: cAMP response element-binding protein, IGF: Insulin like growth factor, PKA: Protein kinase A.

Furthermore, genetic deletion of PAPP-A showed significant reduction in cyst development and significantly improved the inflammation, kidney injury and fibrosis in the Pkd1RC/RC mouse model. Even a single copy deletion of PAPP-A was able to improve the cystic disease, glomerular filtration rate and survival in ADPKD mice, confirming the role of PAPP-A in pathogenesis of ADPKD. PAPP-A deficiency led to the reduced expression of downstream signaling components of IGF-1R like ERK, Akt, and PCNA, whereas it also showed an induction in AMPK expression. There was no difference in the IGF levels measured in circulation. In addition, preclinical studies demonstrated that treatment with a monoclonal antibody that blocks the proteolytic activity of PAPP-A against IGFBP4 ameliorated ADPKD cystic disease in vivo in Pkd1RC/RC mice, as well as ex vivo in embryonic kidneys [41]. Therefore, this study demonstrated that PAPP-A–IGF-1 axis plays an important role in the cystogenesis and introduced a new therapeutic strategy for ADPKD involving the inhibition of PAPP-A (Figure 3). Moreover, there exists a cross talk between IGF and EGF receptors pathway not only on cell surface level but downstream too [138], and IGF-1 mediated transactivation of EGFR and subsequent downstream signaling have been reported earlier [139]. Therefore, although future studies are needed but it could be speculated that PAPP-A inhibition might regulate the local IGF-1 levels and thus inhibit the crosstalk with these receptors further inhibiting these signaling pathway responsible for cystic disease.

Pharmacological approaches to inhibit PAPP-A

Although, direct inhibition of IGF signaling targeting IGF1R has been proposed in diseases like cancer, phase III clinical trials did not demonstrate a clear benefit and shown some disadvantages due to the lack of specificity [140142]. PAPP-A could be an alternative therapeutic target to inhibit the IGF signaling, since inhibition of PAPP-A does not inhibit the IGF-1/IGF1R signaling directly, but limits the tissue specific bioavailability of IGF. Moreover, PAPP-A is an ecto-enzyme and therefore it can be targeted by specific monoclonal antibodies that can inhibit its proteolytic activity [143]. For example, Mikkelsen et al showed the selective inhibition of PAPP-A proteolytic activity against IGFBP-4 in vitro by using monoclonal antibodies that target the unique substrate binding site exosite of PAPP-A [143]. Later, they also reported that IGF signaling can be targeted in vivo using monoclonal antibodies against PAPP-A in a murine xenograft model [144]. Similarly, these neutralizing antibodies inhibited ovarian cancer growth and ascites accumulation in human tumor avatar models in mice [145]. Recently, Mohrin et al also reported the development of a neutralizing antibody against PAPP-A (anti-PAPP-A) and demonstrated that short-term treatment with anti-PAPP-A leads to a reduction in IGF signaling in mesenchymal stromal cells (MSCs) which causes functional changes at the tissue level [146]. All these studies clearly provide the evidence that inhibition of proteolytic activity of PAPP-A using monoclonal antibodies can be utilized to modulate IGF signaling in tissues.

Therefore, the development of therapeutic monoclonal antibodies against PAPP-A will provide a novel approach to target the IGF signaling. There are several advantages in using monoclonal antibodies for therapeutical interventions. For example, monoclonal antibodies can provide highly specific targeting, therefore decreasing the potential for off- target effects and increasing its therapeutic window. In addition, antibodies have long and reliable half-lives [147].

Somatostatin analogues like octreotide and lanreotide have also been implicated in the treatment of ADPKD, and several clinical trials have been conducted with varying outcomes [148151]. Somatostatin acts on 5 G-protein coupled receptors (GPCRs) and binding to these receptors blocks the mitogen activated protein kinase, cell proliferation and also suppresses the expression of IGF-1, as well as other growth factors [152, 153]. Thus, somatostatin analogues could block the cystic growth via multiple mechanisms, including suppression of IGF-1. Therefore, combination therapies including somatostatin analogs and PAPP-A antibodies could provide an alternative approach to treat ADPKD patients.

Conclusions

Recently, we described a direct role for PAPP-A, a component of the IGF pathway, in the pathogenesis of ADPKD. Targeting PAPP-A-IGFBP-IGF axis could provide a new avenue for the management of this cystic disease. For example, therapeutic monoclonal antibodies could be used to provide specific target engagement and higher efficacy with reduced toxicity. Therefore, pharmacological inhibition of PAPP-A using therapeutic monoclonal antibodies could be a novel approach to target the IGF pathway in ADPKD.

Funding

This work was supported in part by grants from the Mayo Clinic Center for Biomedical Discovery and grant DK090728 from the Mayo Translational PKD Center.

Footnotes

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

References

  • 1.Tan Y-C, Blumenfeld J, and Rennert H, Autosomal dominant polycystic kidney disease: Genetics, mutations and microRNAs. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 2011. 1812(10): p. 1202–1212. [DOI] [PubMed] [Google Scholar]
  • 2.Torres VE, Harris PC, and Pirson Y, Autosomal dominant polycystic kidney disease. The Lancet, 2007. 369(9569): p. 1287–1301. [DOI] [PubMed] [Google Scholar]
  • 3.Gansevoort RT, et al. , Recommendations for the use of tolvaptan in autosomal dominant polycystic kidney disease: a position statement on behalf of the ERA-EDTA Working Groups on Inherited Kidney Disorders and European Renal Best Practice. Nephrology Dialysis Transplantation, 2016. 31(3): p. 337–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Harris PC, 2008 Homer W. Smith Award: Insights into the Pathogenesis of Polycystic Kidney Disease from Gene Discovery. Journal of the American Society of Nephrology, 2009. 20(6): p. 1188–1198. [DOI] [PubMed] [Google Scholar]
  • 5.Rowe I and Boletta A, Defective metabolism in polycystic kidney disease: potential for therapy and open questions. Nephrology Dialysis Transplantation, 2014. 29(8): p. 1480–1486. [DOI] [PubMed] [Google Scholar]
  • 6.Kashyap S. Chini EN, Energy Metabolism, Metabolic Sensors, and Nutritional Interventions in Polycystic Kidney Disease, in Polycystic Kidney Disease, Jinghua Hu YY, Editor. 2019, CRC Press; p. 161–170. [Google Scholar]
  • 7.Cornec-Le Gall E, Torres VE, and Harris PC, Genetic Complexity of Autosomal Dominant Polycystic Kidney and Liver Diseases. Journal of the American Society of Nephrology, 2018. 29(1): p. 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Qian F, et al. , The Molecular Basis of Focal Cyst Formation in Human Autosomal Dominant Polycystic Kidney Disease Type I. Cell, 1996. 87(6): p. 979–987. [DOI] [PubMed] [Google Scholar]
  • 9.Bergmann C, et al. , Polycystic kidney disease. Nature Reviews Disease Primers, 2018. 4(1): p. 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Yamaguchi T, et al. , Cyclic AMP activates B-Raf and ERK in cyst epithelial cells from autosomal-dominant polycystic kidneys. Kidney International, 2003. 63(6): p. 1983–1994. [DOI] [PubMed] [Google Scholar]
  • 11.Lee K, Battini L, and Gusella GL, Cilium, centrosome and cell cycle regulation in polycystic kidney disease. Biochimica et biophysica acta, 2011. 1812(10): p. 1263–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nauli SM, et al. , Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nature Genetics, 2003. 33(2): p. 129–137. [DOI] [PubMed] [Google Scholar]
  • 13.Parnell SC, et al. , The Polycystic Kidney Disease-1 Protein, Polycystin-1, Binds and Activates Heterotrimeric G-Proteinsin Vitro. Biochemical and Biophysical Research Communications, 1998. 251(2): p. 625–631. [DOI] [PubMed] [Google Scholar]
  • 14.Bhunia AK, et al. , PKD1 Induces p21waf1 and Regulation of the Cell Cycle via Direct Activation of the JAK-STAT Signaling Pathway in a Process Requiring PKD2. Cell, 2002. 109(2): p. 157–168. [DOI] [PubMed] [Google Scholar]
  • 15.Boca M, et al. , Polycystin-1 Induces Resistance to Apoptosis through the Phosphatidylinositol 3-Kinase/Akt Signaling Pathway. Journal of the American Society of Nephrology, 2006. 17(3): p. 637–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Takiar V, et al. , Activating AMP-activated protein kinase (AMPK) slows renal cystogenesis. Proceedings of the National Academy of Sciences, 2011. 108(6): p. 2462–2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Padovano V, et al. , Metabolism and mitochondria in polycystic kidney disease research and therapy. Nature Reviews Nephrology, 2018. 14(11): p. 678–687. [DOI] [PubMed] [Google Scholar]
  • 18.Rowe I, et al. , Defective glucose metabolism in polycystic kidney disease identifies a new therapeutic strategy. Nature Medicine, 2013. 19: p. 488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chiaravalli M, et al. , 2-Deoxy-D-Glucose Ameliorates PKD Progression. Journal of the American Society of Nephrology, 2016. 27(7): p. 1958–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Riwanto M, et al. , Inhibition of Aerobic Glycolysis Attenuates Disease Progression in Polycystic Kidney Disease. PLOS ONE, 2016. 11(1): p. e0146654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Menezes LF, et al. , Fatty Acid Oxidation is Impaired in An Orthologous Mouse Model of Autosomal Dominant Polycystic Kidney Disease. EBioMedicine, 2016. 5: p. 183–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lin C-C, et al. , A cleavage product of Polycystin-1 is a mitochondrial matrix protein that affects mitochondria morphology and function when heterologously expressed. Scientific Reports, 2018. 8(1): p. 2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hanudel MR, et al. , Erythropoietin and Fibroblast Growth Factor 23 in Autosomal Dominant Polycystic Kidney Disease Patients. Kidney International Reports, 2019. 4(12): p. 1742–1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chonchol M, et al. , Fibroblast Growth Factor 23 and Kidney Disease Progression in Autosomal Dominant Polycystic Kidney Disease. Clinical Journal of the American Society of Nephrology, 2017. 12(9): p. 1461–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Du J and Wilson PD, Abnormal polarization of EGF receptors and autocrine stimulation of cyst epithelial growth in human ADPKD. American Journal of Physiology-Cell Physiology, 1995. 269(2): p. C487–C495. [DOI] [PubMed] [Google Scholar]
  • 26.Zheleznova NN, Wilson PD, and Staruschenko A, Epidermal growth factor-mediated proliferation and sodium transport in normal and PKD epithelial cells. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 2011. 1812(10): p. 1301–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.LeRoith D, et al. , Molecular and Cellular Aspects of the Insulin-Like Growth Factor I Receptor. Endocrine Reviews, 1995. 16(2): p. 143–163. [DOI] [PubMed] [Google Scholar]
  • 28.Jones JI and Clemmons DR, Insulin-Like Growth Factors and Their Binding Proteins: Biological Actions*. Endocrine Reviews, 1995. 16(1): p. 3–34. [DOI] [PubMed] [Google Scholar]
  • 29.Annunziata M, Granata R, and Ghigo E, The IGF system. Acta Diabetologica, 2011. 48(1): p. 1–9. [DOI] [PubMed] [Google Scholar]
  • 30.Vitale G, et al. , ROLE of IGF-1 System in the Modulation of Longevity: Controversies and New Insights From a Centenarians’ Perspective. Frontiers in Endocrinology, 2019. 10(27). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bowers LW, et al. , The Role of the Insulin/IGF System in Cancer: Lessons Learned from Clinical Trials and the Energy Balance-Cancer Link. Frontiers in Endocrinology, 2015. 6(77). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yu H and Rohan T, Role of the Insulin-Like Growth Factor Family in Cancer Development and Progression. JNCI: Journal of the National Cancer Institute, 2000. 92(18): p. 1472–1489. [DOI] [PubMed] [Google Scholar]
  • 33.Laron Z, Insulin-like growth factor 1 (IGF-1): a growth hormone. Molecular Pathology, 2001. 54(5): p. 311–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Christopoulos PF, Msaouel P, and Koutsilieris M, The role of the insulin-like growth factor-1 system in breast cancer. Molecular Cancer, 2015. 14(1): p. 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nakamura T, et al. , Growth factor gene expression in kidney of murine polycystic kidney disease. Journal of the American Society of Nephrology, 1993. 3(7): p. 1378–86. [DOI] [PubMed] [Google Scholar]
  • 36.Aukema HM and Housini I, Dietary soy protein effects on disease and IGF-I in male and female Han:SPRD-cy rats. Kidney International, 2001. 59(1): p. 52–61. [DOI] [PubMed] [Google Scholar]
  • 37.Song X, et al. , Systems biology of autosomal dominant polycystic kidney disease (ADPKD): computational identification of gene expression pathways and integrated regulatory networks. Human Molecular Genetics, 2009. 18(13): p. 2328–2343. [DOI] [PubMed] [Google Scholar]
  • 38.Parker E, et al. , Insulin-like growth factor-1 induces hyperproliferation of PKD1 cystic cells via a Ras/Raf dependent signalling pathway. Kidney international, 2007. 72(2): p. 157–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liu C, et al. , Rosiglitazone inhibits insulin-like growth factor-1-induced polycystic kidney disease cell growth and p70S6 kinase activation. Molecular medicine reports, 2013. 8. [DOI] [PubMed] [Google Scholar]
  • 40.Warner G, et al. , Food Restriction Ameliorates the Development of Polycystic Kidney Disease. Journal of the American Society of Nephrology : JASN, 2016. 27(5): p. 1437–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kashyap S, et al. , Metalloproteinase PAPP-A regulation of IGF-1 contributes to polycystic kidney disease pathogenesis. JCI Insight, 2020. 5(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Daughaday WH and Rotwein P, Insulin-like growth factors I and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocr Rev, 1989. 10(1): p. 68–91. [DOI] [PubMed] [Google Scholar]
  • 43.Rinderknecht E and Humbel RE, Polypeptides with nonsuppressible insulin-like and cell-growth promoting activities in human serum: isolation, chemical characterization, and some biological properties of forms I and II. Proc Natl Acad Sci U S A, 1976. 73(7): p. 2365–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Brissenden JE, Ullrich A, and Francke U, Human chromosomal mapping of genes for insulin-like growth factors I and II and epidermal growth factor. Nature, 1984. 310(5980): p. 781–4. [DOI] [PubMed] [Google Scholar]
  • 45.Rotwein P, Structure, evolution, expression and regulation of insulin-like growth factors I and II. Growth Factors, 1991. 5(1): p. 3–18. [DOI] [PubMed] [Google Scholar]
  • 46.Hameed M, et al. , Expression of IGF-I splice variants in young and old human skeletal muscle after high resistance exercise. J Physiol, 2003. 547(Pt 1): p. 247–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Philippou A, et al. , Expression of IGF-1 isoforms after exercise-induced muscle damage in humans: characterization of the MGF E peptide actions in vitro. In Vivo, 2009. 23(4): p. 567–75. [PubMed] [Google Scholar]
  • 48.Monk D, et al. , Imprinting of IGF2 P0 transcript and novel alternatively spliced INS-IGF2 isoforms show differences between mouse and human. Hum Mol Genet, 2006. 15(8): p. 1259–69. [DOI] [PubMed] [Google Scholar]
  • 49.Rodríguez S, et al. , Haplotypic analyses of the IGF2-INS-TH gene cluster in relation to cardiovascular risk traits. Hum Mol Genet, 2004. 13(7): p. 715–25. [DOI] [PubMed] [Google Scholar]
  • 50.Bergman D, et al. , Insulin-Like Growth Factor 2 in Development and Disease: A Mini-Review. Gerontology, 2013. 59(3): p. 240–249. [DOI] [PubMed] [Google Scholar]
  • 51.Ullrich A, et al. , Insulin-like growth factor I receptor primary structure: comparison with insulin receptor suggests structural determinants that define functional specificity. EMBO J, 1986. 5(10): p. 2503–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bailyes EM, et al. , Insulin receptor/IGF-I receptor hybrids are widely distributed in mammalian tissues: quantification of individual receptor species by selective immunoprecipitation and immunoblotting. Biochem J, 1997. 327 (Pt 1): p. 209–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Federici M, et al. , Distribution of insulin/insulin-like growth factor-I hybrid receptors in human tissues. Mol Cell Endocrinol, 1997. 129(2): p. 121–6. [DOI] [PubMed] [Google Scholar]
  • 54.Laron Z, Insulin-like growth factor 1 (IGF-1): a growth hormone. Mol Pathol, 2001. 54(5): p. 311–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pollak M, The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer, 2012. 12(3): p. 159–69. [DOI] [PubMed] [Google Scholar]
  • 56.Braulke T, Type-2 IGF Receptor: A Multi-Ligand Binding Protein. Horm Metab Res, 1999. 31(02/03): p. 242–246. [DOI] [PubMed] [Google Scholar]
  • 57.Allard JB and Duan C, IGF-Binding Proteins: Why Do They Exist and Why Are There So Many? Front Endocrinol (Lausanne), 2018. 9: p. 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mohseni-Zadeh S and Binoux M, Insulin-like growth factor (IGF) binding protein-3 interacts with the type 1 IGF receptor, reducing the affinity of the receptor for its ligand: an alternative mechanism in the regulation of IGF action. Endocrinology, 1997. 138(12): p. 5645–8. [DOI] [PubMed] [Google Scholar]
  • 59.Salahifar H, et al. , Characterization of an amino-terminal fragment of insulin-like growth factor binding protein-3 and its effects in MCF-7 breast cancer cells. Growth Horm IGF Res, 2000. 10(6): p. 367–77. [DOI] [PubMed] [Google Scholar]
  • 60.Duan C and Clemmons DR, Differential Expression and Biological Effects of Insulin-like Growth Factor-binding Protein-4 and −5 in Vascular Smooth Muscle Cells. Journal of Biological Chemistry, 1998. 273(27): p. 16836–16842. [DOI] [PubMed] [Google Scholar]
  • 61.Yakar S, et al. , Normal growth and development in the absence of hepatic insulin-like growth factor I. Proceedings of the National Academy of Sciences, 1999. 96(13): p. 7324–7329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Forbes BE, McCarthy P, and Norton RS, Insulin-like growth factor binding proteins: a structural perspective. Front Endocrinol (Lausanne), 2012. 3: p. 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Baxter RC, Characterization of the acid-labile subunit of the growth hormone-dependent insulin-like growth factor binding protein complex. J Clin Endocrinol Metab, 1988. 67(2): p. 265–72. [DOI] [PubMed] [Google Scholar]
  • 64.Twigg SM and Baxter RC, Insulin-like growth factor (IGF)-binding protein 5 forms an alternative ternary complex with IGFs and the acid-labile subunit. J Biol Chem, 1998. 273(11): p. 6074–9. [DOI] [PubMed] [Google Scholar]
  • 65.Albiston AL and Herington AC, Tissue distribution and regulation of insulin-like growth factor (IGF)-binding protein-3 messenger ribonucleic acid (mRNA) in the rat: comparison with IGF-I mRNA expression. Endocrinology, 1992. 130(1): p. 497–502. [DOI] [PubMed] [Google Scholar]
  • 66.Olivecrona H, et al. , Acute and short-term effects of growth hormone on insulin-like growth factors and their binding proteins: serum levels and hepatic messenger ribonucleic acid responses in humans. J Clin Endocrinol Metab, 1999. 84(2): p. 553–60. [DOI] [PubMed] [Google Scholar]
  • 67.Tazuke SI, et al. , Hypoxia stimulates insulin-like growth factor binding protein 1 (IGFBP-1) gene expression in HepG2 cells: a possible model for IGFBP-1 expression in fetal hypoxia. Proc Natl Acad Sci U S A, 1998. 95(17): p. 10188–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Averous J, et al. , Induction of IGFBP-1 expression by amino acid deprivation of HepG2 human hepatoma cells involves both a transcriptional activation and an mRNA stabilization due to its 3’UTR. FEBS Lett, 2005. 579(12): p. 2609–14. [DOI] [PubMed] [Google Scholar]
  • 69.Cotterill AM, Holly JM, and Wass JA, The regulation of insulin-like growth factor binding protein (IGFBP)-1 during prolonged fasting. Clin Endocrinol (Oxf), 1993. 39(3): p. 357–62. [DOI] [PubMed] [Google Scholar]
  • 70.Robertson DG, et al. , Insulin and glucocorticoids regulate IGFBP-1 expression via a common promoter region. Biochem Biophys Res Commun, 1994. 200(1): p. 226–32. [DOI] [PubMed] [Google Scholar]
  • 71.Jones JI, et al. , Extracellular matrix contains insulin-like growth factor binding protein-5: potentiation of the effects of IGF-I. J Cell Biol, 1993. 121(3): p. 679–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Parker A, et al. , Binding of insulin-like growth factor (IGF)-binding protein-5 to smooth-muscle cell extracellular matrix is a major determinant of the cellular response to IGF-I. Mol Biol Cell, 1998. 9(9): p. 2383–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jones JI, et al. , Insulin-like growth factor binding protein 1 stimulates cell migration and binds to the alpha 5 beta 1 integrin by means of its Arg-Gly-Asp sequence. Proc Natl Acad Sci U S A, 1993. 90(22): p. 10553–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Abrass CK, Berfield AK, and Andress DL, Heparin binding domain of insulin-like growth factor binding protein-5 stimulates mesangial cell migration. Am J Physiol, 1997. 273(6): p. F899–906. [DOI] [PubMed] [Google Scholar]
  • 75.Zhang C, et al. , IGF binding protein-6 expression in vascular endothelial cells is induced by hypoxia and plays a negative role in tumor angiogenesis. Int J Cancer, 2012. 130(9): p. 2003–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Schedlich LJ, et al. , Nuclear import of insulin-like growth factor-binding protein-3 and −5 is mediated by the importin beta subunit. J Biol Chem, 2000. 275(31): p. 23462–70. [DOI] [PubMed] [Google Scholar]
  • 77.Schedlich LJ, et al. , Insulin-like growth factor-binding protein (IGFBP)-3 and IGFBP-5 share a common nuclear transport pathway in T47D human breast carcinoma cells. J Biol Chem, 1998. 273(29): p. 18347–52. [DOI] [PubMed] [Google Scholar]
  • 78.Xu Q, et al. , Evidence that IGF binding protein-5 functions as a ligand-independent transcriptional regulator in vascular smooth muscle cells. Circ Res, 2004. 94(5): p. E46–54. [DOI] [PubMed] [Google Scholar]
  • 79.Hwa V, Oh Y, and Rosenfeld RG, The insulin-like growth factor-binding protein (IGFBP) superfamily. Endocr Rev, 1999. 20(6): p. 761–87. [DOI] [PubMed] [Google Scholar]
  • 80.Rajah R, et al. , Insulin-like growth factor binding protein (IGFBP) proteases: Functional regulators of cell growth. Progress in Growth Factor Research, 1995. 6(2): p. 273–284. [DOI] [PubMed] [Google Scholar]
  • 81.Wetterau LA, et al. , Novel Aspects of the Insulin-like Growth Factor Binding Proteins. Molecular Genetics and Metabolism, 1999. 68(2): p. 161–181. [DOI] [PubMed] [Google Scholar]
  • 82.Besnard N, et al. , Proteolytic Activity Degrading Insulin-like Growth Factor-Binding Protein−2;2, −3, −4, and −5 in Healthy Growing and Atretic Follicles in the Pig Ovary1. Biology of Reproduction, 1997. 56(4): p. 1050–1058. [DOI] [PubMed] [Google Scholar]
  • 83.Busby WH, et al. , The Complement Component C1s Is the Protease That Accounts for Cleavage of Insulin-like Growth Factor-binding Protein-5 in Fibroblast Medium. Journal of Biological Chemistry, 2000. 275(48): p. 37638–37644. [DOI] [PubMed] [Google Scholar]
  • 84.Shi Z, et al. , ADAM 12, a Disintegrin Metalloprotease, Interacts with Insulin-like Growth Factor-binding Protein-3. Journal of Biological Chemistry, 2000. 275(24): p. 18574–18580. [DOI] [PubMed] [Google Scholar]
  • 85.Clay Bunn R and Fowlkes JL, Insulin-like growth factor binding protein proteolysis. Trends in Endocrinology & Metabolism, 2003. 14(4): p. 176–181. [DOI] [PubMed] [Google Scholar]
  • 86.Monget P and Oxvig C, PAPP-A and the IGF system. Annales d’Endocrinologie, 2016. 77(2): p. 90–96. [DOI] [PubMed] [Google Scholar]
  • 87.BOLDT HB, et al. , Mutational analysis of the proteolytic domain of pregnancy-associated plasma protein-A (PAPP-A): classification as a metzincin. Biochemical Journal, 2001. 358(2): p. 359–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lawrence JB, et al. , The insulin-like growth factor (IGF)-dependent IGF binding protein-4 protease secreted by human fibroblasts is pregnancy-associated plasma protein-A. Proceedings of the National Academy of Sciences, 1999. 96(6): p. 3149–3153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Conover CA, et al. , Metalloproteinase pregnancy-associated plasma protein A is a critical growth regulatory factor during fetal development. Development, 2004. 131(5): p. 1187–1194. [DOI] [PubMed] [Google Scholar]
  • 90.Laursen LS, et al. , Pregnancy-associated plasma protein-A (PAPP-A) cleaves insulin-like growth factor binding protein (IGFBP)-5 independent of IGF: implications for the mechanism of IGFBP-4 proteolysis by PAPP-A. FEBS Letters, 2001. 504(1–2): p. 36–40. [DOI] [PubMed] [Google Scholar]
  • 91.Monget P, et al. , Pregnancy-Associated Plasma Protein-A Is Involved in Insulin-Like Growth Factor Binding Protein-2 (IGFBP-2) Proteolytic Degradation in Bovine and Porcine Preovulatory Follicles: Identification of Cleavage Site and Characterization of IGFBP-2 Degradation1. Biology of Reproduction, 2003. 68(1): p. 77–86. [DOI] [PubMed] [Google Scholar]
  • 92.Overgaard MT, et al. , Pregnancy-associated Plasma Protein-A2 (PAPP-A2), a Novel Insulin-like Growth Factor-binding Protein-5 Proteinase. Journal of Biological Chemistry, 2001. 276(24): p. 21849–21853. [DOI] [PubMed] [Google Scholar]
  • 93.Lin T-M, et al. , Characterization of four human pregnancy-associated plasma proteins. American Journal of Obstetrics and Gynecology, 1974. 118(2): p. 223–236. [DOI] [PubMed] [Google Scholar]
  • 94.Overgaard MT, et al. , Messenger Ribonucleic Acid Levels of Pregnancy-Associated Plasma Protein-A and the Proform of Eosinophil Major Basic Protein: Expression in Human Reproductive and Nonreproductive Tissues1. Biology of Reproduction, 1999. 61(4): p. 1083–1089. [DOI] [PubMed] [Google Scholar]
  • 95.Wald NJ, Watt HC, and Hackshaw AK, Integrated Screening for Down’s Syndrome Based on Tests Performed during the First and Second Trimesters. New England Journal of Medicine, 1999. 341(7): p. 461–467. [DOI] [PubMed] [Google Scholar]
  • 96.Conover CA, Kiefer MC, and Zapf J, Posttranslational regulation of insulin-like growth factor binding protein-4 in normal and transformed human fibroblasts. Insulin-like growth factor dependence and biological studies. The Journal of Clinical Investigation, 1993. 91(3): p. 1129–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Durham SK, et al. , Regulation of insulin-like growth factor binding protein 4 by a specific insulin-like growth factor binding protein 4 proteinase in normal human osteoblast-like cells: Implications in bone cell physiology. Journal of Bone and Mineral Research, 1994. 9(1): p. 111–117. [DOI] [PubMed] [Google Scholar]
  • 98.Parker A, et al. , Properties of an insulin-like growth factor-binding protein-4 protease that is secreted by smooth muscle cells. Endocrinology, 1995. 136(6): p. 2470–2476. [DOI] [PubMed] [Google Scholar]
  • 99.Mazerbourg S, et al. , Pregnancy-Associated Plasma Protein-A (PAPP-A) in Ovine, Bovine, Porcine, and Equine Ovarian Follicles: Involvement in IGF Binding Protein-4 Proteolytic Degradation and mRNA Expression During Follicular Development. Endocrinology, 2001. 142(12): p. 5243–5253. [DOI] [PubMed] [Google Scholar]
  • 100.Byun D, et al. , Pregnancy-associated plasma protein-A accounts for the insulin-like growth factor (IGF)-binding protein-4 (IGFBP-4) proteolytic activity in human pregnancy serum and enhances the mitogenic activity of IGF by degrading IGFBP-4 in vitro. Journal of Clinical Endocrinology and Metabolism, 2001. 86(2): p. 847–854. [DOI] [PubMed] [Google Scholar]
  • 101.Bayes-Genis A, et al. , Insulin-like growth factor binding protein-4 protease produced by smooth muscle cells increases in the coronary artery after angioplasty. Arteriosclerosis, Thrombosis, and Vascular Biology, 2001. 21(3): p. 335–341. [DOI] [PubMed] [Google Scholar]
  • 102.Conover CA, et al. , Evidence That the Insulin-Like Growth Factor Binding Protein-4 Protease in Human Ovarian Follicular Fluid Is Pregnancy Associated Plasma Protein-A. The Journal of Clinical Endocrinology & Metabolism, 1999. 84(12): p. 4742–4745. [DOI] [PubMed] [Google Scholar]
  • 103.Giudice LC, et al. , Identification and regulation of the IGFBP-4 protease and its physiological inhibitor in human trophoblasts and endometrial stroma: Evidence for paracrine regulation of IGF-II bioavailability in the placental bed during human implantation. Journal of Clinical Endocrinology and Metabolism, 2002. 87(5): p. 2359–2366. [DOI] [PubMed] [Google Scholar]
  • 104.Boldt HB and Conover CA, Pregnancy-associated plasma protein-A (PAPP-A): A local regulator of IGF bioavailability through cleavage of IGFBPs. Growth Hormone & IGF Research, 2007. 17(1): p. 10–18. [DOI] [PubMed] [Google Scholar]
  • 105.Oxvig C, The role of PAPP-A in the IGF system: location, location, location. Journal of Cell Communication and Signaling, 2015. 9(2): p. 177–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Conover CA, Key questions and answers about pregnancy-associated plasma protein-A. Trends in Endocrinology & Metabolism, 2012. 23(5): p. 242–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Oxvig C, et al. , Circulating human pregnancy-associated plasma protein-A is disulfide-bridged to the proform of eosinophil major basic protein. Journal of Biological Chemistry, 1993. 268(17): p. 12243–6. [PubMed] [Google Scholar]
  • 108.Glerup S, et al. , Cell Surface Detachment of Pregnancy-associated Plasma Protein-A Requires the Formation of Intermolecular Proteinase-Inhibitor Disulfide Bonds and Glycosaminoglycan Covalently Bound to the Inhibitor. Journal of Biological Chemistry, 2007. 282(3): p. 1769–1778. [DOI] [PubMed] [Google Scholar]
  • 109.Laursen LS, et al. , Regulation of Insulin-Like Growth Factor (IGF) Bioactivity by Sequential Proteolytic Cleavage of IGF Binding Protein-4 and −5. Molecular Endocrinology, 2007. 21(5): p. 1246–1257. [DOI] [PubMed] [Google Scholar]
  • 110.Rehage M, et al. , Transgenic Overexpression of Pregnancy-Associated Plasma Protein-A Increases the Somatic Growth and Skeletal Muscle Mass in Mice. Endocrinology, 2007. 148(12): p. 6176–6185. [DOI] [PubMed] [Google Scholar]
  • 111.Qin X, et al. , Pregnancy-Associated Plasma Protein-A Increases Osteoblast Proliferation in Vitro and Bone Formation in Vivo. Endocrinology, 2006. 147(12): p. 5653–5661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Phang D, et al. , Inactivation of insulin-like-growth factors diminished the anabolic effects of pregnancy-associated plasma protein-A (PAPP-A) on bone in mice. Growth Hormone & IGF Research, 2010. 20(3): p. 192–200. [DOI] [PubMed] [Google Scholar]
  • 113.Jepsen MR, et al. , Stanniocalcin-2 Inhibits Mammalian Growth by Proteolytic Inhibition of the Insulin-like Growth Factor Axis. Journal of Biological Chemistry, 2015. 290(6): p. 3430–3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Resch ZT, et al. , Pregnancy-Associated Plasma Protein A Gene Expression as a Target of Inflammatory Cytokines. Endocrinology, 2004. 145(3): p. 1124–1129. [DOI] [PubMed] [Google Scholar]
  • 115.Conover CA, Harrington SC, and Bale LK, Differential regulation of pregnancy associated plasma protein-A in human coronary artery endothelial cells and smooth muscle cells. Growth Hormone & IGF Research, 2008. 18(3): p. 213–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Folkersen J, et al. , Pregnancy-associated plasma protein A: Circulating levels during normal pregnancy. American Journal of Obstetrics and Gynecology, 1981. 139(8): p. 910–914. [DOI] [PubMed] [Google Scholar]
  • 117.Han VK, et al. , The expression of insulin-like growth factor (IGF) and IGF-binding protein (IGFBP) genes in the human placenta and membranes: evidence for IGF-IGFBP interactions at the feto-maternal interface. The Journal of Clinical Endocrinology & Metabolism, 1996. 81(7): p. 2680–2693. [DOI] [PubMed] [Google Scholar]
  • 118.Takeda Y and Iwashita M, Role of growth factors on fetal growth and maturation. Annals of the Academy of Medicine, Singapore, 1993. 22(2): p. 134–141. [PubMed] [Google Scholar]
  • 119.Irwin JC, et al. , Role of the IGF system in trophoblast invasion and pre-eclampsia. Hum Reprod, 1999. 14 Suppl 2: p. 90–6. [DOI] [PubMed] [Google Scholar]
  • 120.Smith GCS, et al. , Early Pregnancy Levels of Pregnancy-Associated Plasma Protein A and the Risk of Intrauterine Growth Restriction, Premature Birth, Preeclampsia, and Stillbirth. The Journal of Clinical Endocrinology & Metabolism, 2002. 87(4): p. 1762–1767. [DOI] [PubMed] [Google Scholar]
  • 121.Poon LCY, et al. , First-trimester maternal serum pregnancy-associated plasma protein-A and pre-eclampsia. Ultrasound in Obstetrics & Gynecology, 2009. 33(1): p. 23–33. [DOI] [PubMed] [Google Scholar]
  • 122.Giudice I, et al. , Correlation of neonatal weight with maternal serum levels of pregnancy-associated plasma protein-A during the first trimester of pregnancy: a retrospective study. 2015. 43(2): p. 227. [DOI] [PubMed] [Google Scholar]
  • 123.Resch ZT, Simari RD, and Conover CA, Targeted Disruption of the Pregnancy-Associated Plasma Protein-A Gene Is Associated with Diminished Smooth Muscle Cell Response to Insulin-like Growth Factor-I and Resistance to Neointimal Hyperplasia after Vascular Injury. Endocrinology, 2006. 147(12): p. 5634–5640. [DOI] [PubMed] [Google Scholar]
  • 124.Chen B-K, et al. , Localization and Regulation of Pregnancy-Associated Plasma Protein A Expression in Healing Human Skin. The Journal of Clinical Endocrinology & Metabolism, 2003. 88(9): p. 4465–4471. [DOI] [PubMed] [Google Scholar]
  • 125.Hourvitz A, et al. , Pregnancy-Associated Plasma Protein-A Gene Expression in Human Ovaries Is Restricted to Healthy Follicles and Corpora Lutea. The Journal of Clinical Endocrinology & Metabolism, 2000. 85(12): p. 4916–4920. [DOI] [PubMed] [Google Scholar]
  • 126.Hourvitz A, et al. , The Regulated Expression of the Pregnancy-Associated Plasma Protein-A in the Rodent Ovary: A Proposed Role in the Development of Dominant Follicles and of Corpora Lutea. Endocrinology, 2002. 143(5): p. 1833–1844. [DOI] [PubMed] [Google Scholar]
  • 127.Bayes-Genis A, et al. , Pregnancy-Associated Plasma Protein A as a Marker of Acute Coronary Syndromes. New England Journal of Medicine, 2001. 345(14): p. 1022–1029. [DOI] [PubMed] [Google Scholar]
  • 128.Bulut I, et al. , Relationship Between Pregnancy-Associated Plasma Protein-A and Lung Cancer. The American Journal of the Medical Sciences, 2009. 337(4): p. 241–244. [DOI] [PubMed] [Google Scholar]
  • 129.Conover CA, Role of PAPP-A in Aging and Age-related Disease. Experimental gerontology, 2013. 48(7): p. 612–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Cheryl AC and Claus O, 40 YEARS OF IGF1: PAPP-A and cancer. Journal of Molecular Endocrinology, 2018. 61(1): p. T1–T10. [DOI] [PubMed] [Google Scholar]
  • 131.Harrington SC, Simari RD, and Conover CA, Genetic Deletion of Pregnancy-Associated Plasma Protein-A Is Associated With Resistance to Atherosclerotic Lesion Development in Apolipoprotein E–Deficient Mice Challenged With a High-Fat Diet. Circulation Research, 2007. 100(12): p. 1696–1702. [DOI] [PubMed] [Google Scholar]
  • 132.Conover CA, et al. , Transgenic overexpression of pregnancy-associated plasma protein-A in murine arterial smooth muscle accelerates atherosclerotic lesion development. American Journal of Physiology-Heart and Circulatory Physiology, 2010. 299(2): p. H284–H291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Li X, et al. , PAPP-A: A possible pathogenic link to the instability of atherosclerotic plaque. Medical Hypotheses, 2008. 70(3): p. 597–599. [DOI] [PubMed] [Google Scholar]
  • 134.Etter C, et al. , Pregnancy-associated plasma protein-A is an independent short-time predictor of mortality in patients on maintenance haemodialysis. European Heart Journal, 2009. 31(3): p. 354–359. [DOI] [PubMed] [Google Scholar]
  • 135.Kalousová M, et al. , Increased levels of pregnancy-associated plasma protein A are associated with mortality in hemodialysis patients: Preliminary results. Blood Purification, 2004. 22(3): p. 298–300. [DOI] [PubMed] [Google Scholar]
  • 136.Aso Y, et al. , Elevated Pregnancy-Associated Plasma Protein-A in Sera from Type 2 Diabetic Patients with Hypercholesterolemia: Associations with Carotid Atherosclerosis and Toe-Brachial Index. The Journal of Clinical Endocrinology & Metabolism, 2004. 89(11): p. 5713–5717. [DOI] [PubMed] [Google Scholar]
  • 137.Fialová L, et al. , Pregnancy-associated plasma protein-A in patients with cerebrovascular diseases--a pilot study. Prague Med Rep, 2006. 107(1): p. 37–45. [PubMed] [Google Scholar]
  • 138.Veeken J.v.d., et al. , Crosstalk Between Epidermal Growth Factor Receptor- and Insulin-Like Growth Factor-1 Receptor Signaling: Implications for Cancer Therapy. Current Cancer Drug Targets, 2009. 9(6): p. 748–760. [DOI] [PubMed] [Google Scholar]
  • 139.Roudabush FL, et al. , Transactivation of the EGF Receptor Mediates IGF-1-stimulated Shc Phosphorylation and ERK1/2 Activation in COS-7 Cells. Journal of Biological Chemistry, 2000. 275(29): p. 22583–22589. [DOI] [PubMed] [Google Scholar]
  • 140.Yee D, Insulin-like Growth Factor Receptor Inhibitors: Baby or the Bathwater? JNCI: Journal of the National Cancer Institute, 2012. 104(13): p. 975–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Beckwith H and Yee D, Minireview: Were the IGF Signaling Inhibitors All Bad? Molecular Endocrinology, 2015. 29(11): p. 1549–1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Heidegger I, et al. , Targeting the insulin-like growth factor network in cancer therapy. Cancer Biology & Therapy, 2011. 11(8): p. 701–707. [DOI] [PubMed] [Google Scholar]
  • 143.Mikkelsen JH, et al. , Inhibition of the Proteolytic Activity of Pregnancy-associated Plasma Protein-A by Targeting Substrate Exosite Binding. Journal of Biological Chemistry, 2008. 283(24): p. 16772–16780. [DOI] [PubMed] [Google Scholar]
  • 144.Mikkelsen JH, et al. , Indirect targeting of IGF receptor signaling in vivo by substrate-selective inhibition of PAPP-A proteolytic activity. Oncotarget, 2014. 5(4): p. 1014–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Becker MA, et al. , A Novel Neutralizing Antibody Targeting Pregnancy-Associated Plasma Protein-A Inhibits Ovarian Cancer Growth and Ascites Accumulation in Patient Mouse Tumorgrafts. Molecular Cancer Therapeutics, 2015. 14(4): p. 973–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Mohrin M, et al. , Pharmacological inhibition of longevity regulator PAPP-A restrains mesenchymal stromal cell activity. bioRxiv, 2020: p. 2020.02.05.936310. [Google Scholar]
  • 147.Tsumoto K, et al. , Future perspectives of therapeutic monoclonal antibodies. Immunotherapy, 2019. 11(2): p. 119–127. [DOI] [PubMed] [Google Scholar]
  • 148.Griffiths J, Mills MT, and Ong AC, Long-acting somatostatin analogue treatments in autosomal dominant polycystic kidney disease and polycystic liver disease: a systematic review and meta-analysis. BMJ Open, 2020. 10(1): p. e032620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Caroli A, et al. , Effect of longacting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): a randomised, placebo-controlled, multicentre trial. The Lancet, 2013. 382(9903): p. 1485–1495. [DOI] [PubMed] [Google Scholar]
  • 150.Meijer E, et al. , Effect of Lanreotide on Kidney Function in Patients With Autosomal Dominant Polycystic Kidney Disease: The DIPAK 1 Randomized Clinical Trial. JAMA, 2018. 320(19): p. 2010–2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Perico N, et al. , Octreotide-LAR in later-stage autosomal dominant polycystic kidney disease (ALADIN 2): A randomized, double-blind, placebo-controlled, multicenter trial. PLOS Medicine, 2019. 16(4): p. e1002777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Pyronnet S, et al. , Antitumor effects of somatostatin. Molecular and Cellular Endocrinology, 2008. 286(1): p. 230–237. [DOI] [PubMed] [Google Scholar]
  • 153.Grozinsky-Glasberg S, et al. , Somatostatin analogues in the control of neuroendocrine tumours: efficacy and mechanisms. 2008. 15(3): p. 701. [DOI] [PubMed] [Google Scholar]
  • 154.Cohen P, et al. , Prostate-specific antigen (PSA) is an insulin-like growth factor binding protein-3 protease found in seminal plasma. The Journal of Clinical Endocrinology & Metabolism, 1992. 75(4): p. 1046–1053. [DOI] [PubMed] [Google Scholar]
  • 155.Rajah R, et al. , 7S nerve growth factor is an insulin-like growth factor-binding protein protease. Endocrinology, 1996. 137(7): p. 2676–2682. [DOI] [PubMed] [Google Scholar]
  • 156.Booth BA, Boes M, and Bar RS, IGFBP-3 proteolysis by plasmin, thrombin, serum: heparin binding, IGF binding, and structure of fragments. American Journal of Physiology-Endocrinology and Metabolism, 1996. 271(3): p. E465–E470. [DOI] [PubMed] [Google Scholar]
  • 157.Campbell PG and Andress DL, Insulin-like growth factor (IGF)-binding protein-5-(201—218) region regulates hydroxyapatite and IGF-I binding. American Journal of Physiology-Endocrinology and Metabolism, 1997. 273(5): p. E1005–E1013. [DOI] [PubMed] [Google Scholar]
  • 158.Zheng B, et al. , Insulin-Like Growth Factor-Binding Protein-5 Is Cleaved by Physiological Concentrations of Thrombin*. Endocrinology, 1998. 139(4): p. 1708–1714. [DOI] [PubMed] [Google Scholar]
  • 159.Byun D, et al. , Pregnancy-Associated Plasma Protein-A Accounts for the Insulin-Like Growth Factor (IGF)-Binding Protein-4 (IGFBP-4) Proteolytic Activity in Human Pregnancy Serum and Enhances the Mitogenic Activity of IGF by Degrading IGFBP-4 in Vitro1. The Journal of Clinical Endocrinology & Metabolism, 2001. 86(2): p. 847–854. [DOI] [PubMed] [Google Scholar]
  • 160.Loechel F, et al. , ADAM 12-S Cleaves IGFBP-3 and IGFBP-5 and Is Inhibited by TIMP-3. Biochemical and Biophysical Research Communications, 2000. 278(3): p. 511–515. [DOI] [PubMed] [Google Scholar]
  • 161.Mohan S, et al. , ADAM-9 Is an Insulin-like Growth Factor Binding Protein-5 Protease Produced and Secreted by Human Osteoblasts. Biochemistry, 2002. 41(51): p. 15394–15403. [DOI] [PubMed] [Google Scholar]
  • 162.Fowlkes JL, et al. , Proteolysis of insulin-like growth factor binding protein-3 during rat pregnancy: a role for matrix metalloproteinases. Endocrinology, 1994. 135(6): p. 2810–2813. [DOI] [PubMed] [Google Scholar]
  • 163.Fowlkes JL, et al. , Matrix metalloproteinases degrade insulin-like growth factor-binding protein-3 in dermal fibroblast cultures. Journal of Biological Chemistry, 1994. 269(41): p. 25742–6. [PubMed] [Google Scholar]
  • 164.Cohen P, et al. , Leukotriene D4 facilitates airway smooth muscle cell proliferation via modulation of the IGF axis. American Journal of Physiology-Lung Cellular and Molecular Physiology, 1995. 269(2): p. L151–L157. [DOI] [PubMed] [Google Scholar]
  • 165.Rajah R, et al. , Leukotriene D4 induces MMP-1, which functions as an IGFBP protease in human airway smooth muscle cells. American Journal of Physiology-Lung Cellular and Molecular Physiology, 1996. 271(6): p. L1014–L1022. [DOI] [PubMed] [Google Scholar]
  • 166.Conover CA and De Leon DD, Acid-activated insulin-like growth factor-binding protein-3 proteolysis in normal and transformed cells. Role of cathepsin D. Journal of Biological Chemistry, 1994. 269(10): p. 7076–7080. [PubMed] [Google Scholar]
  • 167.Conover CA, Perry JE, and Tindall DJ, Endogenous cathepsin D-mediated hydrolysis of insulin-like growth factor-binding proteins in cultured human prostatic carcinoma cells. The Journal of Clinical Endocrinology & Metabolism, 1995. 80(3): p. 987–993. [DOI] [PubMed] [Google Scholar]
  • 168.Nunn SE, Peehl DM, and Cohen P, Acid-activated insulin-like growth factor binding protein protease activity of Cathepsin D in normal and malignant prostatic epithelial cells and seminal plasma. Journal of Cellular Physiology, 1997. 171(2): p. 196–204. [DOI] [PubMed] [Google Scholar]
  • 169.Claussen M, et al. , Proteolysis of Insulin-Like Growth Factors (IGF) and IGF Binding Proteins by Cathepsin D*. Endocrinology, 1997. 138(9): p. 3797–3803. [DOI] [PubMed] [Google Scholar]
  • 170.Liu C, et al. , Rosiglitazone inhibits insulin-like growth factor-1-induced polycystic kidney disease cell growth and p70S6 kinase activation. Molecular Medicine Reports, 2013. 8(3): p. 861–4. [DOI] [PubMed] [Google Scholar]

RESOURCES