Skip to main content

Role of metastasis-induced protein S100A4 in human non-tumor pathophysiologies

Abstract

S100A4, an important member of the S100 family of proteins, is best known for its significant role in promoting cancer progression and metastasis. In addition to its expression in tumors, upregulation of S100A4 expression has been associated with various non-tumor pathophysiology processes. However, the mechanisms underlying the role of S100A4 remain unclear. Activated “host” cells (fibroblasts, immunocytes, vascular cells, among others) secrete S100A4 into the extracellular space in various non-tumor human disorders, where it executes its biological functions by interacting with intracellular target proteins. However, the exact molecular mechanisms underlying these interactions in different non-tumor pathophysiologies vary, and S100A4 is likely one of the cross-linking factors that acts as common intrinsic constituents of biological mechanisms. Numerous studies have indicated that the S100A4-mediated epithelial–mesenchymal transition plays a vital role in the occurrence and development of various non-tumor pathophysiologies. Epithelial–mesenchymal transition can be categorized into three general subtypes based on the phenotype and function of the output cells. S100A4 regulates tissue fibrosis associated with the type II epithelial–mesenchymal transition via various signaling pathways. Additionally, S100A4 stimulates fibroblasts to secrete fibronectin and collagen, thus forming the structural components of the extracellular matrix (ECM) and stimulating their deposition in tissues, contributing to the formation of a pro-inflammatory niche. Simultaneously, S100A4 enhances the motility of macrophages, neutrophils, and leukocytes and promotes the recruitment and chemotaxis of these inflammatory cells to regulate inflammation and immune functions. S100A4 also exerts a neuroprotective pro-survival effect on neurons by rescuing them from brain injury and participates in angiogenesis by interacting with other target molecules. In this review, we summarize the role of S100A4 in fibrosis, inflammation, immune response, neuroprotection, angiogenesis, and some common non-tumor diseases as well as its possible involvement in molecular pathways and potential clinical value.

Background

In humans, the S100 protein family contains 21 members that exhibit a high degree of sequence and structural similarity. These protein members are not functionally interchangeable and thus are involved in a wide range of biological processes, such as cell division, differentiation, apoptosis, proliferation, migration, and invasion; cytoskeletal organization; and enzyme activity [1,2,3]. The first member of the S100 protein family was documented in 1965, and the term S100 was used to refer to the solubility of this protein with a molecular mass of approximately 10 kDa in saturated ammonium sulfate solution [4]. S100 proteins have a high degree of homology with similar amino acid sequences and 3D structures. These structures are characterized by a symmetrical homodimeric fold containing two EF-hand helix-loop-helix Ca2+-binding domains in each monomer [5]. The carboxy-terminal EF-hand loop is a canonical Ca2+-coordinating site consisting of 12 amino acid residues. In contrast, the amino-terminal pseudo-EF-hand loop comprising 14 amino acid residues is considered a unique feature of S100 proteins [6, 7]. Most S100 proteins can act as Ca2+ sensors based on their role in signal transduction and also contribute to fluctuations in intracellular Ca2+ levels to cellular responses; this sensory capability is a result of changes in the S100 protein conformation following Ca2+ binding [2, 8]. Despite the high sequence similarity among S100 proteins, individual family members show unique characteristics, which are determined by cell- and tissue-specific expression as well as their varying biochemical properties, including affinity for lipid and divalent metal ions, conformation after oligomerization or heterodimerization, and post-translational modifications [3, 9].

S100A4 is categorized as a metastasis-associated protein and is also known as metastasin, pEL-98, 18A2, p9Ka, CAPL, calvasculin, and fibroblast-specific protein [10,11,12]. The human S100A4 protein is encoded by S100A4, which is located within a frequently rearranged gene cluster on chromosome 1q21. The gene contains four exons encoding the two EF-hands of the Ca2+-binding regions of S100A4 and its 101 amino acids with a molecular mass of approximately 12 kDa [13]. Binding of Ca2+ ions causes a conformational change in S100A4 to form a hydrophobic pocket for the recognition and combination of target molecules [14]. The original cloning efforts reported S100A4 to be a highly expressed transcript in cells with growth-related transformation [15], metastatic tumor cells [10], and in cells undergoing conversion from an epithelial to mesenchymal phenotype [16]. In addition, S100A4 is expressed in several highly motile cell types, such as T-lymphocytes, neutrophils, and macrophages, as well as in tissues, including the spleen, thymus, and bone marrow [17]. Numerous intracellular target proteins, including non-muscle myosin IIA, p53, and liprin-β1, combine with S100A4 to form complexes that regulate cell motility [18,19,20]. Moreover, S100A4 and several other S100 proteins were reported to be secreted into the extracellular space by various cells involved in the process of neurite outgrowth [21], angiogenesis [22], and chemokine and cytokine-like activities [23, 24].

A growing number of studies have demonstrated the direct involvement of S100A4 in cancer progression and metastasis. The expression of S100A4 in tumor cells is strongly correlated with an aggressive metastatic phenotype [10]. In humans, upregulation of S100A4, both in tumor and stroma cells, was associated with poor prognosis and low survival of patients with cancer [25, 26]. In its role as a metastasis regulating factor, expression of S100A4 has been reported to be affected by methylation, β-catenin, and extracellular factors including epidermal growth factor, tumor necrosis factor alpha (TNF-α), etc. [27]. In addition to the role of S100A4 in cancer progression and metastasis, this protein was recently found to be involved in some vital non-tumor pathophysiologies in humans, which has attracted much attention [28, 29]. Here, we summarize the role of S100A4 in non-tumor pathophysiologies, including fibrosis, inflammation, immune responses, neuroprotection, and angiogenesis, and some common non-tumor diseases as well as the possible molecular pathways in which S100A4 is involved and its potential clinical value.

Underlying mechanisms of S100A4 in non-tumor pathophysiologies

The protein S100A4 is closely associated with both non-tumor and tumors. In addition to being expressed in tumor cells, S100A4 is expressed in many normal cells, including fibroblasts, macrophages, lymphocytes, neutrophils, vascular cells, and bone marrow-derived cells [17, 30, 31]. Several recent publications have demonstrated an association between S100A4 and non-tumor pathophysiologies, such as tissue fibrosis, inflammation, immune reaction, neuroprotection, and cardiovascular events. However, the underlying mechanisms by which S100A4 is involved in these pathophysiologies remain unclear.

A study has indicated that the S100A4-mediated epithelial–mesenchymal transition (EMT) program plays a vital role in the occurrence and development of various diseases, including tumor and non-tumor diseases [32]. The EMT is a complex of molecular programs that occurs during embryogenesis, inflammation, tissue fibrosis, and cancer progression and metastasis. It can be categorized into three  general subtypes as described in an EMT meeting at Cold Spring Harbor Laboratory in 2008 [33]. The three types of EMT are recognized depending on the phenotype of the output cells. Type I EMT (normal tissue development) involves the transition of primordial epithelial cells into motile mesenchymal cells that eventually form the basic body of gastrulation and induce neural crest cell migration. These mesenchymal cells are then re-induced as secondary epithelial cells in mesodermal and endodermal organs after undergoing mesenchymal–epithelial transition. Type II EMT (pathological conditions) involves the transition of secondary epithelial or endothelial cells to resident or inflammation-induced fibroblasts, which occurs in response to persistent inflammation and fibrosis and is regulated by the high expression of S100A4 via various signaling pathways. Type III EMT is part of the metastatic process in which epithelial carcinoma cells in their primary nodule forms migrate to a distant site via blood circulation to reform as a secondary tumor nodule [34].

In addition, S100A4 enhances the motility of macrophages, neutrophils, and leukocytes and promotes the recruitment and chemotaxis of these inflammatory cells to regulate inflammation and immune [35]. S100A4 also exerts a neuroprotective pro-survival effect on neurons by rescuing them from brain injury [36] and participates in angiogenesis via its interaction with other target molecules [37]. Intracellularly, S100A4 binds to several target molecules, which leads to changes in cytoskeletal dynamics and promotion of cell motility and proliferation [38]. Moreover, various types of cells, including fibroblasts, macrophages, and lymphocytes, express and release S100A4 into their extracellular space as an active extracellular factor; thus, S100A4 has a great tendency to regulate gene expression associated with proteolytic activity, angiogenesis, cell survival, and motility by modulating the signaling pathways of mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinase, p38, Jun N-terminal kinases, nuclear factor-kappa B (NF-κB), and p53 [39,40,41]. The role of S100A4 in human non-tumor pathophysiologies is summarized in Fig. 1.

Fig. 1
figure 1

Extra-S100A4 can be released into the extracellular space by fibroblasts, macrophages, lymphocytes, neutrophils, vascular cells, and other bone marrow derived cells. The S100A4 interacted with IL-10R exerts a neuron pro-survival effect under various injuries via JAK/STAT pathway partially. Besides, the expression of extra-S100A4 leads to increasing phosphorylation of Pyk-2, MAPKs, and activating NF-κB through the RAGE-dependent regulation associated with cell migratory abilities and chemotaxis. On the other hand, the intracellular S100A4 can combine with numerous target molecules, such as NMIIA, tropomyosin, P53, and actin, to form the complexes, facilitating the remodeling of microtubes and microfilaments to enhance cell motility and chemotaxis, contributing to the infiltration of fibroblasts, immune and vascular cells into the affected region releasing inflammatory factors. In addition, S100A4 colocalizes with P53 promoting cell proliferation and collagen expression via MAPK activation and phosphorylation of ERK. The TGF-β-mediated process induces the up-regulation of S100A4 promoting the generation of extracellular matrix, collagen, elastin and others underlying the basis for the course of inflammation tissue fibrosis. Moreover, the intracellular S100A4 regulates its upstream and/or downstream gene expression involved in proteolytic activity, angiogenesis and cell survival by modulating signal pathways of MAPKs, ERK, p38, JNK, NF-κB, and p53

S100A4 promotes tissue fibrosis

S100A4 is considered a specific fibroblast maker, and thus is frequently used to monitor or predict the mechanism of tissue fibrosis [42]. As a filament-associated protein, it is expressed in kidney fibrosis cells with fibroblast phenotypes, where it aids in conducting type II EMT [16]. All fibroblasts  observed to express S100A4 through genetic screens are likely to undergo type II EMT; thus, the protein is involved in adult epithelial or endothelial cells’ transition into fibroblasts [16]. Studies based on cell lineage-tracing have provided evidence for this transition during the formation of fibroblasts in the renal tissue [43] as well as in liver [44], lung [45], and heart [46] tissues. As in epithelial cells, endothelial cells are also sources of new fibroblasts in adult tissue, which we reported after observing that S100A4-positive fibroblasts underwent type II EMT in the heart  [46] and kidney [47]. The elevated expression of S100A4 in tubular epithelial cells contributes to their transformation to interstitial fibroblasts during kidney injury [46] and fibrosis [43]. Along with other marker proteins, such as vimentin, α-smooth muscle actin, and matrix metalloproteinases (MMPs), S100A4 is now accepted as an important hallmark of the EMT process. The expression of intracellular S100A4 is upregulated during the TGF-β-mediated transition of epithelial cells associated with the EMT program [48, 49]. Extracellular S100A4 changes the differentiated state of adjacent cells through damage-associated signaling, which regulates numerous cellular processes crucial to fibrotic progression depending on the location of S100A4 [28]. The initiation of S100A4 expression in epithelial cells that have undergone EMT promotes the generation of extracellular matrix (ECM) components, such as collagen, elastin, and other proteins, providing a basis for the onset of tissue fibrosis [50]. Fibroblast transcription site-1 exists in the promoters of EMT-associated genes, such as Twist, Snail, and β-catenin. It regulates the expression of S100A4 in fibroblast-regulated intestinal and pulmonary tissue fibrosis [51, 52]. Research indicates that S100A4 confers idiopathic pulmonary fibrosis lungs harboring fibrogenic mesenchymal progenitor cells with fibrogenicity [53]. Hypertrophic scars in tissues, followed by fibrosis induced by TNF-α, lead to an increase in MMP, S100A4, and vimentin expression, which can be antagonized by bone morphogenetic protein 2/4 [54].

Furthermore, S100A4 stimulates fibroblasts to secrete fibronectin and collagen, thus forming the structural components of the ECM and stimulating their deposition in tissues, contributing to the formation of a pro-inflammatory niche [55, 56]. The high production of another group of ECM regulators, the MMPs, is induced by both extra- and intracellular S100A4, with the extracellular S100A4-mediated stimulation of MMP being the major path of action [40]. Different MMPs not only promote degradation of the ECM, but also together with other active factors that are stimulated by S100A4 facilitate remodeling of the ECM, inducing a pro-inflammatory microenvironment.

S100A4 enhances cell migration and chemotaxis

Research has suggested that S100A4 is a potent trigger in pro-inflammatory pathways. High expression of S100A4 enhances the physical motility of macrophages and neutrophils and promotes the recruitment and chemotaxis of these inflammatory cells; this occurs in response to the secretion of inflammatory cytokines [57]. The intra- and extracellular biological functions of S100A4 commonly induce a pathological inflammation-like process in various inflammatory disorders. By interacting with intracellular cytoskeleton–associated target molecules, S100A4 facilitates remodeling of acto-myosin filaments, thus enhancing cell motility and chemotaxis [13]. This contributes to the aggravation of pathological processes via the infiltration of fibroblasts as well as immune and vascular cells into the affected regions, releasing inflammatory factors [58]. Under various pathological stimuli, numerous inflammatory cells upregulate the expression of S100A4 and release it in the form of plasma membrane-derived microvesicles into the extracellular space [59]. The inflammation-associated signal transduction by S100A4 is associated with numerous receptors, such as receptor for advanced glycation end products (RAGE), Toll-like receptor 4, and interleukin (IL)-10 receptor [60,61,62]. Extracellular S100A4 activates a major pro-inflammatory pathway in the cell, i.e., the MAPK pathway, which triggers the recruitment of cells involved in inflammation and the self-amplifying pro-inflammatory cycle by upregulating several pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Other than cytokines, acute phase reactants, granulocyte colony-stimulating factor, and the well-known inflammation-associated S100 family members, S100A8 and S100A9, are also upregulated, establishing an inflammatory milieu [63]. Additionally, extracellular S100A4 exerts a strong influence on the activation of another major pro-inflammatory transcription factor, NF-κB [61]. Moreover, remodeling of the ECM and expression of aberrant ECM molecules can be stimulated by S100A4, which, in turn, plays a substantial role in supporting the chronic inflammatory response at the affected site [64]. S100A4 also promotes colitis development via S100A4-mediated host inflammatory responses by increasing adhesion and colonization of Citrobacter rodentium [29].

S100A4 has been reported to be associated with leukocyte migration, which stimulates cytokine production, particularly, that of granulocyte colony-stimulating factor and eotaxin-2, by T lymphocytes involved in allergic inflammation [65, 66]. The chemo-attracting activity is mediated by extracellular S100A4 via its interactions with specific receptors, such as RAGE [40]. In this manner, S100A4 acts as an important modulator of T cell migration during immune responses, specifically in T-cells [67, 68]. The absence of intracellular S100A4 leads to over-assembly of acto-myosin complexes, resulting in inhibition of chemotactic motility in cultured bone marrow-derived macrophages [57]. Recently, researchers observed that only memory T-cells expressed S100A4. Furthermore, sub-populations of effector memory T-cells showed the highest expression of S100A4. Interestingly, S100A4-deficient memory T-cells also migrated towards chemokines, facilitating autoimmune inflammatory disorders; this is in sharp contrast to the observation that loss of S100A4 expression decreased chemokine-stimulated motility [68]. S100A4 expression is also known to be involved in the pathogenesis of several autoimmune diseases, such as rheumatoid arthritis, systemic sclerosis, and psoriasis. It is markedly upregulated in the joint synovial tissue and also exists in the plasma, while maintaining a bioactive multimeric conformation, in rheumatoid arthritis patients [39]. The levels of the bioactive form of S100A4 are correlated with its disease activity, and thus S100A4 is successfully decreased by blocking therapy in rheumatoid arthritis patients [69].

S100A4 facilitates neuroprotection

S100A4 plays a pro-survival role in the brain; because of this role, the protein is also considered a neuroprotective factor that protects many types of cells during brain injury through several signaling pathways [21, 60]. The expression of S100A4 in astrocytes not only contributes to the normal process of myelination, but also modulates post-traumatic events in the nervous system. Its expression has been detected in white matter astrocytes at brain trauma sites [70] and was also observed to be markedly upregulated in the hippocampus after excitotoxic injury [60]. In contrast, grey matter astrocytes were observed to be consistently S100A4-negative even after injury [71]. Moreover, S100A4 was found to be clearly overexpressed in astrocytes of the spinal cord adjacent to the injury site [72]. In vitro, S100A4 is secreted by astrocytes to exert its extracellular effects associated with neurite outgrowth and survival of primary hippocampal and cerebellar neurons [60, 73].

S100A4 exerts a neuroprotective pro-survival effect on neurons by rescuing them from brain injury via the Janus kinase/signal transducer and activator of transcription pathway in part by interacting with IL-10R [60]. In a previous study, mimicking S100A4-induced neuroprotection in vivo increased neuronal loss after traumatic brain injury and helped identify two neurotrophic motifs in the S100A4 sequence: H3 and H6 [60]. Thus, S100A4 stimulated the neuritogenesis and survival of cultured neurons. Furthermore, S100A4 protects neurons from an injured central nervous system by neuritogenic and pro-survival effects and likely by promoting glial–axonal interactions [60]. In the peripheral nervous system, the expression of S100A4 increases in the myelinated Schwann cells and unmyelinated Remak bundles at the injury site following a dorsal root or peripheral nerve injury. Its expression also increases in a sub-population of neurons (mainly sensory and autonomic) [74]. Similar to the central nervous system, S100A4 exerts pro-regenerative and pro-survival effects in an injured peripheral nervous system, and mimetic peptides derived from neurotrophic motifs of S100A4 contribute to axonal sprouting and survival [75].

S100A4 regulates angiogenesis

Recent studies linked S100A4 to several metastatic diseases. One of its metastasis-promoting mechanisms is its function as a potent stimulator of angiogenesis [76]. Angiogenesis is a multi-step process of degradation of the basement membranes and ECM by MMPs, followed by migration of endothelial cells towards the angiogenic stimuli. Next, cell proliferation takes place, followed by formation of a tubular structure that sprouts from pre-existing blood vessels [77]. Research has shown that S100A4 is a pro-angiogenic factor that participates in angiogenesis, endothelial cell migration and invasion via its interaction with annexin 2, formation of plasmin, and stimulation of MMP production [78, 79]. S100A4-mediated angiogenesis also functions synergistically with the vascular endothelial growth factor, which is an important angiogenic factor that promotes neo-vascularization and vascular leakage through an upstream regulator, the brain-derived growth factor [53, 78].

In vivo studies have shown that aging S100A4-transgenic mice have higher chances of acquiring hemangioma compared to non-transgenic mice, and the implantation of S100A4-containing micropellets induce neovascularization in the mouse cornea [22]. In vitro experiments have indicated that S100A4 exogenously applied in cell culture can stimulate the motility of endothelial cells and does not influence the proliferation of these cells, demonstrating that S100A4 acts only in cooperation with other angiogenic factors such as vascular endothelial growth factor and brain-derived growth factor to achieve maximum angiogenesis. Moreover, S100A4 is thought to stimulate endothelial cell motility in a cell-specific manner only because of its inefficacy in promoting the motility of mouse fibroblasts. This indicates that S100A4 identifies a specific receptor on the surface of endothelial cells to help them move [22]. Recently, RAGE located on the surface of human endothelial cells was reported to be utilized by S100A4 to enhance endothelial cell migration and neovascularization. This helps the cells associated with other angiogenic factors and achieve angiogenesis-related responses [53, 80].

S100A4 is involved in several common non-tumor diseases and the potential clinical application

The promotion of tissue fibrosis by S100A4 has been reported in chronic obstructive pulmonary disease (COPD), pulmonary arterial hypertension (PAH), and cardiac hypertrophy. A recent study by Reimann and colleagues showed that S100A4 expression was increased in the vasculature tissues of COPD patients and in tissues of murine lungs with vascular remodeling. However, there is no clear evidence regarding how S100A4 contributes to vascular remodeling in COPD patients [81]. The complex programs of EMT and endothelial–mesenchymal transition (EndMT) likely contribute to the pathology of COPD because they actively accompany the increase in S100A4 expression in COPD lungs [82]. S100A4 is not only widely reported to be upregulated in fibrosis, but also a key protein regulating the EMT and EndMT processes [83,84,85,86,87]. Increased expression of S100A4 during EndMT in COPD patients may be the mechanism by which primary arterial smooth muscle cells contribute to pulmonary vascular remodeling [88]; this association was made after determining that transitioning endothelial cells stained positively for S100A4.

S100A4 was found to be involved in the pathogenesis of both human and experimental PAH, which is a progressive disease related to pulmonary vasculature construction and remodeling. Here, S100A4 expression was observed to increase in the adventitia and neointima of early occlusive and plexiform lesions in PAH patients [89]. Similarly, in another study, overexpression of S100A4 in mice was reported to increase right ventricular systolic pressure [90], while some mice underwent pulmonary arterial remodeling similar to occlusive and plexogenic lesions that are typically formed in humans [89]. In a recent report, female S100A4-positive mice readily developed PAH, which may be attributable to the increase in S100A4 expression in these female mice [91]. The researchers considered 17β-estradiol/S100A4 RAGE to be responsible for the development of PAH and gender bias in this disease. This conclusion was based on the fact that in conjunction with RAGE activation, 17β-estradiol upregulates S100A4 expression and proliferation of human pulmonary artery smooth muscle cells [91].

S100A4 has also been reported to be upregulated in cardiac hypertrophy [92], which is a complex process involved in numerous cellular events, such as cytoskeletal and/or ECM reorganization, energy metabolism, signal transduction, gene expression, and cardiomyocyte apoptosis [93]. Expression of S100A4 increases in various cardiac cell types, such as cardiomyocytes, cardiac fibroblasts, and immune cells. In cardiac fibroblasts, S100A4 co-localizes with p53 and regulates its target genes, which are associated with cell proliferation and collagen expression. In addition, co-expression of S100A4 and p53 promotes an overload of cardiac pressure that induces cardiac fibrosis; under this condition, S100A4 likely plays a major role in the pro-fibrotic proliferation and maturation of cardiac fibroblasts [94]. In vitro, S100A4 induces cardiomyocyte hypertrophy and increases cell survival, which inhibit apoptosis via MAPK activation and phosphorylation of extracellular signal-regulated kinase 1/2 [41]. In humans, S100A4 is infrequently detected in coronary arteries, while it is markedly expressed in smooth muscle cells of patients suffering from coronary atherosclerosis and coronary restenosis, suggesting that the expression level of S100A4 in smooth muscle cells has significant potential to aid in the risk assessment of coronary atherosclerosis and coronary restenosis [95].

S100A4 is likewise up-regulated in the skin lesions of systemic sclerosis patients. Amelioration of fibrosis symptoms was observed in S100A4-deficient mice from amongst different experimental fibrosis mouse models [56]. Furthermore, S100A4 is released from the upper dermal compartments of psoriatic skin, while S100A4-specific antibodies reduce vascularization, keratinocyte proliferation, and thickness of psoriatic skin [96]. Significant upregulation of S100A4 has been detected in other autoimmune diseases, such as idiopathic inflammatory myopathies [63] and fibrostenosing Crohn’s disease [97]. Moreover, elevated S100A4 expression in human articular chondrocytes during rheumatism and osteoarthritis leads to increased phosphorylation of protein tyrosine kinase-2 and MAPKs; it further activates NF-κB, increasing secreted MMP13 levels [40]. These effects are regulated partly through RAGE, and thus inhibition of RAGE would negatively impact S100A4-dependent signaling pathways [40]. Analysis of bone tissue from osteoarthritis patients using microarrays and quantitative PCR indicated elevated expression of S100A4 as well as that of other wingless-type mouse mammary tumor virus integration site family-related proteins [98].

Elevated S100A4 expression leads to the development and progression of many non-tumor diseases, indicating that targeting of S100A4 expression or activities is a novel strategy for treating non-tumor pathologies. Because of the increase in S100A4 expression in remodeled intrapulmonary arteries, neutralizing antibodies of S100A4 may prevent vascular remodeling of lung fibrosis [37, 81]. The expression of S100A4 is low in the adult normal myocardium, but significantly enhanced in myocardial infarction, and the detection of plasma-S100A4 serves as a novel biomarker for acute myocardial infarction [99]. Cardiac myocyte-specific overexpression of S100A4 after myocardial infarction may protect the infarcted myocardium against myocardial ischemia, while deletion of S100A4 increases cardiac damage [36]. Extracellular S100A4 regulates bone formation in inflammatory bone disease, increasing the risk of fractures and delaying bone healing, and treating primary calvarial osteoblasts with recombinant S100A4 reduces matrix mineralization [100]. Additionally, S100A4 can be a target for the treatment of periodontitis, which inhibits osteogenic differentiation and enhances matrix degradation [35]. The study showing that silencing of the mouse S100A4 gene ameliorates retinal neovascularization in a mouse model indicates that knockdown of S100A4 may be an effective treatment for ocular neovascularization diseases [53]. Furthermore, therapy targeting S100A4 can suppress the progression of rheumatoid arthritis and persistent high S100A4 expression will predict poor treatment outcome of rheumatoid arthritis [101].

Future perspectives

The roles of S100A4 in cancer progression and metastasis have been demonstrated in numerous studies; this review highlights the participation of S100A4 in the pathophysiologies of several human disorders. The diverse cellular functions of S100A4 and how it exerts its intra- and extracellular influence in non-tumor pathologies remain unclear. As an important cross-linking factor that participates in different signaling pathways, S100A4 participates in mechanisms that are common under different pathological conditions, such as type II EMT in inflammation and fibrosis. S100A4-mediated type II EMT explains several non-tumor biological diseases and the mechanisms by which disease progression is promoted. However, a better understanding of the molecular mechanisms by which S100A4 promotes the progression of these disorders is required. A priority area in studies of S100A4 is to develop novel treatment strategies targeting S100A4 through anti-S100A4 compounds for the therapy of non-tumor diseases.

Conclusions

In addition to malignant tumors, S100A4 plays an important role in many kinds of pathological process and non-tumor diseases including fibrosis, inflammation, immune response, neuroprotection, and angiogenesis.

Abbreviations

BDNF:

brain derived growth factor

BMP:

bone morphogenetic protein

CNS:

central nervous system

COPD:

chronic obstructive pulmonary disease

ECM:

extracellular matrix

EMT:

epithelial–mesenchymal transition

EndMT:

endothelial–mesenchymal transition

ERK:

extracellular signal-regulated kinase

G-CSF:

granulocyte colony stimulating factor

JAK:

Janus kinase

JNK:

jun n-terminal kinase

FSP1:

fibroblast-specific protein 1

hPASMCs:

human pulmonary artery smooth muscle cells

IL-10R:

interleukin-10 receptor

MAPKs:

mitogen-activated protein kinases

MET:

mesenchymal–epithelial transition

MMP:

matrix metalloproteinase

Mts1:

metastasin NF-κB: nuclear factor-kappa B

NMIIA:

non-muscle myosin

IIA PAH:

pulmonary arterial hypertension

PNS:

peripheral nervous system

Pyk-2:

protein tyrosine kinase-2

RAGE:

receptor of advanced glycation end products

RVSP:

right ventricular systolic pressure

STAT:

signal transducer and activator of transcription

TLR4:

toll-like receptor 4

TNF-α:

tumor necrosis factor-alpha

VEGF:

vascular endothelial growth factor

Wnt:

wingless-type mouse mammary tumor virus integration site family

References

  1. Leclerc E, Heizmann CW. The importance of Ca2+/Zn2+ signaling S100 proteins and RAGE in translational medicine. Front Biosci. 2011;3:1232–62.

    Google Scholar 

  2. Santamaria-Kisiel L, Rintala-Dempsey AC, Shaw GS. Calcium-dependent and -independent interactions of the S100 protein family. Biochem J. 2006;396(2):201–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Donato R, Cannon BR, Sorci G, Riuzzi F, Hsu K, Weber DJ, Geczy CL. Functions of S100 proteins. Curr Mol Med. 2013;13(1):24–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Moore BW. A soluble protein characteristic of the nervous system. Biochem Biophys Res Commun. 1965;19(6):739–44.

    Article  CAS  PubMed  Google Scholar 

  5. Zimmer DB, Eubanks JO, Ramakrishnan D, Criscitiello MF. Evolution of the S100 family of calcium sensor proteins. Cell Calcium. 2013;53(3):170–9.

    Article  CAS  PubMed  Google Scholar 

  6. Kligman D, Hilt DC. The S100 protein family. Trends Biochem Sci. 1988;13(11):437–43.

    Article  CAS  PubMed  Google Scholar 

  7. Strynadka NC, James MN. Crystal structures of the helix-loop-helix calcium-binding proteins. Annu Rev Biochem. 1989;58:951–98.

    Article  CAS  PubMed  Google Scholar 

  8. Yap KL, Ames JB, Swindells MB, Ikura M. Diversity of conformational states and changes within the EF-hand protein superfamily. Proteins. 1999;37(3):499–507.

    Article  CAS  PubMed  Google Scholar 

  9. Heizmann CW, Cox JA. New perspectives on S100 proteins: a multi-functional Ca(2+)-, Zn(2+)- and Cu(2+)-binding protein family. Biometals. 1998;11(4):383–97.

    Article  CAS  PubMed  Google Scholar 

  10. Ebralidze A, Tulchinsky E, Grigorian M, Afanasyeva A, Senin V, Revazova E, Lukanidin E. Isolation and characterization of a gene specifically expressed in different metastatic cells and whose deduced gene product has a high degree of homology to a Ca2+-binding protein family. Genes Dev. 1989;3(7):1086–93.

    Article  CAS  PubMed  Google Scholar 

  11. Masiakowski P, Shooter EM. Nerve growth factor induces the genes for two proteins related to a family of calcium-binding proteins in PC12 cells. Proc Natl Acad Sci USA. 1988;85(4):1277–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Watanabe Y, Usuda N, Tsugane S, Kobayashi R, Hidaka H. Calvasculin, an encoded protein from mRNA termed pEL-98, 18A2, 42A, or p9Ka, is secreted by smooth muscle cells in culture and exhibits Ca(2+)-dependent binding to 36-kDa microfibril-associated glycoprotein. J Biol Chem. 1992;267(24):17136–40.

    CAS  PubMed  Google Scholar 

  13. Garrett SC, Varney KM, Weber DJ, Bresnick AR. S100A4, a mediator of metastasis. J Biol Chem. 2006;281(2):677–80.

    Article  CAS  PubMed  Google Scholar 

  14. Malashkevich VN, Varney KM, Garrett SC, Wilder PT, Knight D, Charpentier TH, Ramagopal UA, Almo SC, Weber DJ, Bresnick AR. Structure of Ca2+-bound S100A4 and its interaction with peptides derived from nonmuscle myosin-IIA. Biochemistry. 2008;47(18):5111–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Goto K, Endo H, Fujiyoshi T. Cloning of the sequences expressed abundantly in established cell lines: identification of a cDNA clone highly homologous to S-100, a calcium binding protein. J Biochem. 1988;103(1):48–53.

    Article  CAS  PubMed  Google Scholar 

  16. Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Tomaszewski JE, Neilson EG. Identification and characterization of a fibroblast marker: FSP1. J Cell Biol. 1995;130(2):393–405.

    Article  CAS  PubMed  Google Scholar 

  17. Takenaga K, Nakamura Y, Sakiyama S. Cellular localization of pEL98 protein, an S100-related calcium binding protein, in fibroblasts and its tissue distribution analyzed by monoclonal antibodies. Cell Struct Funct. 1994;19(3):133–41.

    Article  CAS  PubMed  Google Scholar 

  18. Kriajevska MV, Cardenas MN, Grigorian MS, Ambartsumian NS, Georgiev GP, Lukanidin EM. Non-muscle myosin heavy chain as a possible target for protein encoded by metastasis-related mts-1 gene. J Biol Chem. 1994;269(31):19679–82.

    CAS  PubMed  Google Scholar 

  19. Grigorian M, Andresen S, Tulchinsky E, Kriajevska M, Carlberg C, Kruse C, Cohn M, Ambartsumian N, Christensen A, Selivanova G, et al. Tumor suppressor p53 protein is a new target for the metastasis-associated Mts1/S100A4 protein: functional consequences of their interaction. J Biol Chem. 2001;276(25):22699–708.

    Article  CAS  PubMed  Google Scholar 

  20. Kriajevska M, Fischer-Larsen M, Moertz E, Vorm O, Tulchinsky E, Grigorian M, Ambartsumian N, Lukanidin E. Liprin beta 1, a member of the family of LAR transmembrane tyrosine phosphatase-interacting proteins, is a new target for the metastasis-associated protein S100A4 (Mts1). J Biol Chem. 2002;277(7):5229–35.

    Article  CAS  PubMed  Google Scholar 

  21. Kiryushko D, Novitskaya V, Soroka V, Klingelhofer J, Lukanidin E, Berezin V, Bock E. Molecular mechanisms of Ca(2+) signaling in neurons induced by the S100A4 protein. Mol Cell Biol. 2006;26(9):3625–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ambartsumian N, Klingelhofer J, Grigorian M, Christensen C, Kriajevska M, Tulchinsky E, Georgiev G, Berezin V, Bock E, Rygaard J, et al. The metastasis-associated Mts1(S100A4) protein could act as an angiogenic factor. Oncogene. 2001;20(34):4685–95.

    Article  CAS  PubMed  Google Scholar 

  23. Mohan SK, Rani SG, Kumar SM, Yu C. S100A13-C2A binary complex structure—a key component in the acidic fibroblast growth factor for the non-classical pathway. Biochem Biophys Res Commun. 2009;380(3):514–9.

    Article  CAS  PubMed  Google Scholar 

  24. Ryckman C, Vandal K, Rouleau P, Talbot M, Tessier PA. Proinflammatory activities of S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion. J Immunol. 2003;170(6):3233–42.

    Article  CAS  PubMed  Google Scholar 

  25. Rudland PS, Platt-Higgins A, Renshaw C, West CR, Winstanley JH, Robertson L, Barraclough R. Prognostic significance of the metastasis-inducing protein S100A4 (p9Ka) in human breast cancer. Cancer Res. 2000;60(6):1595–603.

    CAS  PubMed  Google Scholar 

  26. Missiaglia E, Blaveri E, Terris B, Wang YH, Costello E, Neoptolemos JP, Crnogorac-Jurcevic T, Lemoine NR. Analysis of gene expression in cancer cell lines identifies candidate markers for pancreatic tumorigenesis and metastasis. Int J Cancer. 2004;112(1):100–12.

    Article  CAS  PubMed  Google Scholar 

  27. Stein U, Arlt F, Walther W, Smith J, Waldman T, Harris ED, Mertins SD, Heizmann CW, Allard D, Birchmeier W, et al. The metastasis-associated gene S100A4 is a novel target of beta-catenin/T-cell factor signaling in colon cancer. Gastroenterology. 2006;131(5):1486–500.

    Article  CAS  PubMed  Google Scholar 

  28. Ellson CD, Dunmore R, Hogaboam CM, Sleeman MA, Murray LA. Danger-associated molecular patterns and danger signals in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2014;51(2):163–8.

    PubMed  Google Scholar 

  29. Zhang J, Jiao Y, Hou S, Tian T, Yuan Q, Hao H, Wu Z, Bao X. S100A4 contributes to colitis development by increasing the adherence of Citrobacter rodentium in intestinal epithelial cells. Sci Rep. 2017;7(1):12099.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Grigorian M, Tulchinsky E, Burrone O, Tarabykina S, Georgiev G, Lukanidin E. Modulation of mts1 expression in mouse and human normal and tumor cells. Electrophoresis. 1994;15(3–4):463–8.

    Article  CAS  PubMed  Google Scholar 

  31. Takenaga K, Nakamura Y, Sakiyama S. Expression of a calcium binding protein pEL98 (mts1) during differentiation of human promyelocytic leukemia HL-60 cells. Biochem Biophys Res Commun. 1994;202(1):94–101.

    Article  CAS  PubMed  Google Scholar 

  32. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401.

    Article  CAS  PubMed  Google Scholar 

  33. Kalluri R, Weinberg RA. The basics of epithelial–mesenchymal transition. J Clin Investig. 2009;119(6):1420–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zeisberg M, Neilson EG. Biomarkers for epithelial–mesenchymal transitions. J Clin Investig. 2009;119(6):1429–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhou M, Li ZQ, Wang ZL. S100A4 upregulation suppresses tissue ossification and enhances matrix degradation in experimental periodontitis models. Acta Pharmacol Sin. 2015;36(11):1388–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Doroudgar S, Quijada P, Konstandin M, Ilves K, Broughton K, Khalafalla FG, Casillas A, Nguyen K, Gude N, Toko H, et al. S100A4 protects the myocardium against ischemic stress. J Mol Cell Cardiol. 2016;100:54–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xia H, Gilbertsen A, Herrera J, Racila E, Smith K, Peterson M, Griffin T, Benyumov A, Yang L, Bitterman PB, et al. Calcium-binding protein S100A4 confers mesenchymal progenitor cell fibrogenicity in idiopathic pulmonary fibrosis. J Clin Investig. 2017;127(7):2586–97.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Boye K, Maelandsmo GM. S100A4 and metastasis: a small actor playing many roles. Am J Pathol. 2010;176(2):528–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Klingelhofer J, Senolt L, Baslund B, Nielsen GH, Skibshoj I, Pavelka K, Neidhart M, Gay S, Ambartsumian N, Hansen BS, et al. Up-regulation of metastasis-promoting S100A4 (Mts-1) in rheumatoid arthritis: putative involvement in the pathogenesis of rheumatoid arthritis. Arthritis Rheum. 2007;56(3):779–89.

    Article  CAS  PubMed  Google Scholar 

  40. Yammani RR, Carlson CS, Bresnick AR, Loeser RF. Increase in production of matrix metalloproteinase 13 by human articular chondrocytes due to stimulation with S100A4: role of the receptor for advanced glycation end products. Arthritis Rheum. 2006;54(9):2901–11.

    Article  CAS  PubMed  Google Scholar 

  41. Schneider M, Kostin S, Strom CC, Aplin M, Lyngbaek S, Theilade J, Grigorian M, Andersen CB, Lukanidin E, Lerche Hansen J, et al. S100A4 is upregulated in injured myocardium and promotes growth and survival of cardiac myocytes. Cardiovasc Res. 2007;75(1):40–50.

    Article  CAS  PubMed  Google Scholar 

  42. Smith BN, Bhowmick NA. Role of EMT in metastasis and therapy resistance. J Clin Med. 2016;5(2):17.

    Article  PubMed Central  Google Scholar 

  43. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Investig. 2002;110(3):341–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, Kalluri R. Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition. J Biol Chem. 2007;282(32):23337–47.

    Article  CAS  PubMed  Google Scholar 

  45. Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA. 2006;103(35):13180–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13(8):952–61.

    Article  CAS  PubMed  Google Scholar 

  47. Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol. 2008;19(12):2282–7.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Kalluri R, Neilson EG. Epithelial–mesenchymal transition and its implications for fibrosis. J Clin Investig. 2003;112(12):1776–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rygiel KA, Robertson H, Marshall HL, Pekalski M, Zhao L, Booth TA, Jones DE, Burt AD, Kirby JA. Epithelial–mesenchymal transition contributes to portal tract fibrogenesis during human chronic liver disease. Lab Investig. 2008;88(2):112–23.

    Article  CAS  PubMed  Google Scholar 

  50. Okada H, Danoff TM, Kalluri R, Neilson EG. Early role of Fsp1 in epithelial–mesenchymal transformation. Am J Physiol. 1997;273(4 Pt 2):F563–74.

    CAS  PubMed  Google Scholar 

  51. Alpers CE, Seifert RA, Hudkins K, Johnson RJ, Bowen-Pope DF. PDGF-receptor localizes to mesangial, parietal epithelial, and interstitial cells in human and primate kidneys. Kidney Int. 1993;43(2):286–94.

    Article  CAS  PubMed  Google Scholar 

  52. Liau G, Mudryj M, de Crombrugghe B. Identification of the promoter and first exon of the mouse alpha 1 (III) collagen gene. J Biol Chem. 1985;260(6):3773–7.

    CAS  PubMed  Google Scholar 

  53. Cheng G, He T, Xing Y. Silencing of S100A4, a metastasis-associated protein, inhibits retinal neovascularization via the downregulation of BDNF in oxygen-induced ischaemic retinopathy. Eye. 2016;30(6):877–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yan C, Grimm WA, Garner WL, Qin L, Travis T, Tan N, Han YP. Epithelial to mesenchymal transition in human skin wound healing is induced by tumor necrosis factor-alpha through bone morphogenic protein-2. Am J Pathol. 2010;176(5):2247–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Grum-Schwensen B, Klingelhofer J, Beck M, Bonefeld CM, Hamerlik P, Guldberg P, Grigorian M, Lukanidin E, Ambartsumian N. S100A4-neutralizing antibody suppresses spontaneous tumor progression, pre-metastatic niche formation and alters T-cell polarization balance. BMC Cancer. 2015;15:44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Tomcik M, Palumbo-Zerr K, Zerr P, Avouac J, Dees C, Sumova B, Distler A, Beyer C, Cerezo LA, Becvar R, et al. S100A4 amplifies TGF-beta-induced fibroblast activation in systemic sclerosis. Ann Rheum Dis. 2015;74(9):1748–55.

    Article  CAS  PubMed  Google Scholar 

  57. Li ZH, Dulyaninova NG, House RP, Almo SC, Bresnick AR. S100A4 regulates macrophage chemotaxis. Mol Biol Cell. 2010;21(15):2598–610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Helfman DM, Kim EJ, Lukanidin E, Grigorian M. The metastasis associated protein S100A4: role in tumour progression and metastasis. Br J Cancer. 2005;92(11):1955–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ambartsumian N, Grigorian M. S100A4, a link between metastasis and inflammation. Mol Biol. 2016;50(4):577–88.

    Article  CAS  Google Scholar 

  60. Dmytriyeva O, Pankratova S, Owczarek S, Sonn K, Soroka V, Ridley CM, Marsolais A, Lopez-Hoyos M, Ambartsumian N, Lukanidin E, et al. The metastasis-promoting S100A4 protein confers neuroprotection in brain injury. Nat Commun. 2012;3:1197.

    Article  PubMed  CAS  Google Scholar 

  61. Grotterod I, Maelandsmo GM, Boye K. Signal transduction mechanisms involved in S100A4-induced activation of the transcription factor NF-kappaB. BMC Cancer. 2010;10:241.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Hansen MT, Forst B, Cremers N, Quagliata L, Ambartsumian N, Grum-Schwensen B, Klingelhofer J, Abdul-Al A, Herrmann P, Osterland M, et al. A link between inflammation and metastasis: serum amyloid A1 and A3 induce metastasis, and are targets of metastasis-inducing S100A4. Oncogene. 2015;34(4):424–35.

    Article  CAS  PubMed  Google Scholar 

  63. Cerezo LA, Kuncova K, Mann H, Tomcik M, Zamecnik J, Lukanidin E, Neidhart M, Gay S, Grigorian M, Vencovsky J, et al. The metastasis promoting protein S100A4 is increased in idiopathic inflammatory myopathies. Rheumatology. 2011;50(10):1766–72.

    Article  PubMed  CAS  Google Scholar 

  64. Sorokin L. The impact of the extracellular matrix on inflammation. Nat Rev Immunol. 2010;10(10):712–23.

    Article  CAS  PubMed  Google Scholar 

  65. Grum-Schwensen B, Klingelhofer J, Grigorian M, Almholt K, Nielsen BS, Lukanidin E, Ambartsumian N. Lung metastasis fails in MMTV-PyMT oncomice lacking S100A4 due to a T-cell deficiency in primary tumors. Cancer Res. 2010;70(3):936–47.

    Article  CAS  PubMed  Google Scholar 

  66. Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, Wherry EJ. Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity. 2014;40(2):289–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Haining WN, Ebert BL, Subrmanian A, Wherry EJ, Eichbaum Q, Evans JW, Mak R, Rivoli S, Pretz J, Angelosanto J, et al. Identification of an evolutionarily conserved transcriptional signature of CD8 memory differentiation that is shared by T and B cells. J Immunol. 2008;181(3):1859–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Weatherly K, Bettonville M, Torres D, Kohler A, Goriely S, Braun MY. Functional profile of S100A4-deficient T cells. Immun Inflamm Dis. 2015;3(4):431–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Oslejskova L, Grigorian M, Hulejova H, Vencovsky J, Pavelka K, Klingelhofer J, Gay S, Neidhart M, Brabcova H, Suchy D, et al. Metastasis-inducing S100A4 protein is associated with the disease activity of rheumatoid arthritis. Rheumatology. 2009;48(12):1590–4.

    Article  CAS  PubMed  Google Scholar 

  70. Kozlova EN, Lukanidin E. Mts1 protein expression in the central nervous system after injury. Glia. 2002;37(4):337–48.

    Article  PubMed  Google Scholar 

  71. Kozlova EN, Lukanidin E. Metastasis-associated mts1 (S100A4) protein is selectively expressed in white matter astrocytes and is up-regulated after peripheral nerve or dorsal root injury. Glia. 1999;27(3):249–58.

    Article  CAS  PubMed  Google Scholar 

  72. Zhang KH, Xiao HS, Lu PH, Shi J, Li GD, Wang YT, Han S, Zhang FX, Lu YJ, Zhang X, et al. Upregulation of S100A4 after spinal cord transection in adult rats. Acta Pharmacol Sin. 2004;25(8):1007–12.

    CAS  PubMed  Google Scholar 

  73. Yammani RR, Long D, Loeser RF. Interleukin-7 stimulates secretion of S100A4 by activating the JAK/STAT signaling pathway in human articular chondrocytes. Arthritis Rheum. 2009;60(3):792–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sandelin M, Zabihi S, Liu L, Wicher G, Kozlova EN. Metastasis-associated S100A4 (Mts1) protein is expressed in subpopulations of sensory and autonomic neurons and in Schwann cells of the adult rat. J Comp Neurol. 2004;473(2):233–43.

    Article  CAS  PubMed  Google Scholar 

  75. Moldovan M, Pinchenko V, Dmytriyeva O, Pankratova S, Fugleholm K, Klingelhofer J, Bock E, Berezin V, Krarup C, Kiryushko D. Peptide mimetic of the S100A4 protein modulates peripheral nerve regeneration and attenuates the progression of neuropathy in myelin protein P0 null mice. Mol Med. 2013;19:43–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. de Silva Rudland S, Martin L, Roshanlall C, Winstanley J, Leinster S, Platt-Higgins A, Carroll J, West C, Barraclough R, Rudland P. Association of S100A4 and osteopontin with specific prognostic factors and survival of patients with minimally invasive breast cancer. Clin Cancer Res. 2006;12(4):1192–200.

    Article  PubMed  CAS  Google Scholar 

  77. Deryugina EI, Quigley JP. Pleiotropic roles of matrix metalloproteinases in tumor angiogenesis: contrasting, overlapping and compensatory functions. Biochim Biophys Acta. 2010;1803(1):103–20.

    Article  CAS  PubMed  Google Scholar 

  78. Schmidt-Hansen B, Ornas D, Grigorian M, Klingelhofer J, Tulchinsky E, Lukanidin E, Ambartsumian N. Extracellular S100A4(mts1) stimulates invasive growth of mouse endothelial cells and modulates MMP-13 matrix metalloproteinase activity. Oncogene. 2004;23(32):5487–95.

    Article  CAS  PubMed  Google Scholar 

  79. Semov A, Moreno MJ, Onichtchenko A, Abulrob A, Ball M, Ekiel I, Pietrzynski G, Stanimirovic D, Alakhov V. Metastasis-associated protein S100A4 induces angiogenesis through interaction with Annexin II and accelerated plasmin formation. J Biol Chem. 2005;280(21):20833–41.

    Article  CAS  PubMed  Google Scholar 

  80. Abu El-Asrar AM, Nawaz MI, De Hertogh G, Alam K, Siddiquei MM, Van den Eynde K, Mousa A, Mohammad G, Geboes K, Opdenakker G. S100A4 is upregulated in proliferative diabetic retinopathy and correlates with markers of angiogenesis and fibrogenesis. Mol Vis. 2014;20:1209–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Reimann S, Fink L, Wilhelm J, Hoffmann J, Bednorz M, Seimetz M, Dessureault I, Troesser R, Ghanim B, Klepetko W, et al. Increased S100A4 expression in the vasculature of human COPD lungs and murine model of smoke-induced emphysema. Respir Res. 2015;16:127.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Sohal SS. Endothelial to mesenchymal transition (EndMT): an active process in chronic obstructive pulmonary disease (COPD)? Respir Res. 2016;17:20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Mahmood MQ, Sohal SS, Shukla SD, Ward C, Hardikar A, Noor WD, Muller HK, Knight DA, Walters EH. Epithelial mesenchymal transition in smokers: large versus small airways and relation to airflow obstruction. Int J Chron Obstruct Pulmon Dis. 2015;10:1515–24.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Sohal SS, Reid D, Soltani A, Ward C, Weston S, Muller HK, Wood-Baker R, Walters EH. Reticular basement membrane fragmentation and potential epithelial mesenchymal transition is exaggerated in the airways of smokers with chronic obstructive pulmonary disease. Respirology. 2010;15(6):930–8.

    Article  PubMed  Google Scholar 

  85. Sohal SS, Reid D, Soltani A, Ward C, Weston S, Muller HK, Wood-Baker R, Walters EH. Evaluation of epithelial mesenchymal transition in patients with chronic obstructive pulmonary disease. Respir Res. 2011;12:130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Bjornland K, Winberg JO, Odegaard OT, Hovig E, Loennechen T, Aasen AO, Fodstad O, Maelandsmo GM. S100A4 involvement in metastasis: deregulation of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in osteosarcoma cells transfected with an anti-S100A4 ribozyme. Cancer Res. 1999;59(18):4702–8.

    CAS  PubMed  Google Scholar 

  87. Schneider M, Hansen JL, Sheikh SP. S100A4: a common mediator of epithelial–mesenchymal transition, fibrosis and regeneration in diseases? J Mol Med. 2008;86(5):507–22.

    Article  CAS  PubMed  Google Scholar 

  88. Coll-Bonfill N, Musri MM, Ivo V, Barbera JA, Tura-Ceide O. Transdifferentiation of endothelial cells to smooth muscle cells play an important role in vascular remodelling. Am J Stem Cells. 2015;4(1):13–21.

    PubMed  PubMed Central  Google Scholar 

  89. Greenway S, van Suylen RJ, Sarvaas GDM, Kwan E, Ambartsumian N, Lukanidin E, Rabinovitch M. S100A4/Mts1 produces murine pulmonary artery changes resembling plexogenic arteriopathy and is increased in human plexogenic arteriopathy. Am J Pathol. 2004;164(1):253–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Merklinger SL, Wagner RA, Spiekerkoetter E, Hinek A, Knutsen RH, Kabir MG, Desai K, Hacker S, Wang L, Cann GM, et al. Increased fibulin-5 and elastin in S100A4/Mts1 mice with pulmonary hypertension. Circ Res. 2005;97(6):596–604.

    Article  CAS  PubMed  Google Scholar 

  91. Dempsie Y, Nilsen M, White K, Mair KM, Loughlin L, Ambartsumian N, Rabinovitch M, Maclean MR. Development of pulmonary arterial hypertension in mice over-expressing S100A4/Mts1 is specific to females. Respir Res. 2011;12:159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Strom CC, Kruhoffer M, Knudsen S, Stensgaard-Hansen F, Jonassen TE, Orntoft TF, Haunso S, Sheikh SP. Identification of a core set of genes that signifies pathways underlying cardiac hypertrophy. Comp Funct Genom. 2004;5(6–7):459–70.

    Article  CAS  Google Scholar 

  93. Gupta S, Das B, Sen S. Cardiac hypertrophy: mechanisms and therapeutic opportunities. Antioxid Redox Signal. 2007;9(6):623–52.

    Article  CAS  PubMed  Google Scholar 

  94. Tamaki Y, Iwanaga Y, Niizuma S, Kawashima T, Kato T, Inuzuka Y, Horie T, Morooka H, Takase T, Akahashi Y, et al. Metastasis-associated protein, S100A4 mediates cardiac fibrosis potentially through the modulation of p53 in cardiac fibroblasts. J Mol Cell Cardiol. 2013;57:72–81.

    Article  CAS  PubMed  Google Scholar 

  95. Brisset AC, Hao H, Camenzind E, Bacchetta M, Geinoz A, Sanchez JC, Chaponnier C, Gabbiani G, Bochaton-Piallat ML. Intimal smooth muscle cells of porcine and human coronary artery express S100A4, a marker of the rhomboid phenotype in vitro. Circ Res. 2007;100(7):1055–62.

    Article  CAS  PubMed  Google Scholar 

  96. Zibert JR, Skov L, Thyssen JP, Jacobsen GK, Grigorian M. Significance of the S100A4 protein in psoriasis. J Investig Dermatol. 2010;130(1):150–60.

    Article  CAS  PubMed  Google Scholar 

  97. Cunningham MF, Docherty NG, Burke JP, O’Connell PR. S100A4 expression is increased in stricture fibroblasts from patients with fibrostenosing Crohn’s disease and promotes intestinal fibroblast migration. Am J Physiol Gastrointest Liver Physiol. 2010;299(2):G457–66.

    Article  CAS  PubMed  Google Scholar 

  98. Hopwood B, Tsykin A, Findlay DM, Fazzalari NL. Microarray gene expression profiling of osteoarthritic bone suggests altered bone remodelling, WNT and transforming growth factor-beta/bone morphogenic protein signalling. Arthritis Res Ther. 2007;9(5):R100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Gong XJ, Song XY, Wei H, Wang J, Niu M. Serum S100A4 levels as a novel biomarker for detection of acute myocardial infarction. Eur Rev Med Pharmacol Sci. 2015;19(12):2221–5.

    PubMed  Google Scholar 

  100. Kim H, Lee YD, Kim MK, Kwon JO, Song MK, Lee ZH, Kim HH. Extracellular S100A4 negatively regulates osteoblast function by activating the NF-κB pathway. BMB Rep. 2017;50(2):97–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Senolt L, Cerezo LA, Sumova B, Pecha O, Plestilova L, Forejtova S, Ruzickova O, Husakova M, Zavada J, Pavelka K, et al. High levels of metastasis-inducing S100A4 protein and treatment outcome in early rheumatoid arthritis: data from the PERAC cohort. Biomarkers. 2015;20(1):47–51.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

All authors have contributed to manuscript writing. All authors read and approved the final manuscript.

Acknowledgements

This work was supported in part by grants from the National Science Foundation of China (#81472729 and #81672426), the Foundation of Tianjin Health Bureau (#2014KY29, #2015KY28, and #15KG112), the Ph.D. Candidate Research Innovation Fund of Nankai University School of Medicine.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

Funding

National Natural Science Foundation of China (Grant Numbers: 81472729 and 81672426).

Consent for publication

Consent for publication has been made online.

Ethics approval and consent to participate

This is a review article, there are no issues relevant to ethics approval and consent to participant.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shiwu Zhang.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fei, F., Qu, J., Li, C. et al. Role of metastasis-induced protein S100A4 in human non-tumor pathophysiologies. Cell Biosci 7, 64 (2017). https://doi.org/10.1186/s13578-017-0191-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13578-017-0191-1

Keywords