Next Article in Journal
Comprehensive Discovery of the Accessible Primary Amino Group-Containing Segments from Cell Surface Proteins by Fine-Tuning a High-Throughput Biotinylation Method
Next Article in Special Issue
Cuproptosis-Related LncRNA-Based Prediction of the Prognosis and Immunotherapy Response in Papillary Renal Cell Carcinoma
Previous Article in Journal
Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial
Previous Article in Special Issue
Reclassification of TCGA Diffuse Glioma Profiles Linked to Transcriptomic, Epigenetic, Genomic and Clinical Data, According to the 2021 WHO CNS Tumor Classification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review

1
Department of Biosciences, Integral University, Dasauli, P.O. Basha, Kursi Road, Lucknow 226026, Uttar Pradesh, India
2
Department of Biochemistry, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
3
Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
4
Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
5
Department of Biology, College of Sciences, University of Hail, Hail P.O. Box 2240, Saudi Arabia
6
Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Hambakmoeiro 191, Yeonsu-gu, Incheon 21924, Republic of Korea
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 266; https://doi.org/10.3390/ijms24010266
Submission received: 17 September 2022 / Revised: 28 October 2022 / Accepted: 16 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Molecular Biomarkers in Cancer and Their Applications)

Abstract

:
The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin family that is overexpressed in several cancers. RAGE is highly expressed in the lung, and its expression increases proportionally at the site of inflammation. This receptor can bind a variety of ligands, including advanced glycation end products, high mobility group box 1, S100 proteins, adhesion molecules, complement components, advanced lipoxidation end products, lipopolysaccharides, and other molecules that mediate cellular responses related to acute and chronic inflammation. RAGE serves as an important node for the initiation and stimulation of cell stress and growth signaling mechanisms that promote carcinogenesis, tumor propagation, and metastatic potential. In this review, we discuss different aspects of RAGE and its prominent ligands implicated in cancer pathogenesis and describe current findings that provide insights into the significant role played by RAGE in cancer. Cancer development can be hindered by inhibiting the interaction of RAGE with its ligands, and this could provide an effective strategy for cancer treatment.

1. Introduction

Cancer is a leading cause of mortality globally—it is estimated to have accounted for 10 million deaths in 2020, which is approximately one-sixth of all deaths worldwide. In 2020, the most prevalent cancers were lung, breast, colorectal, prostate, skin (nonmelanoma), and stomach cancers, with 2.21, 2.26, 1.93, 1.41, 1.2, and 1.09 million cases, respectively. The most prevalent causes of cancer-related deaths were lung, colorectal, stomach, liver, and breast cancers, which accounted for 1.80, 0.935, 0.769, 0.83, and 0.685 million deaths, respectively [1].
Receptor for advanced glycation end products (RAGE) is a 45 kDa multiligand transmembrane receptor in the immunoglobulin (Ig) superfamily that plays an important role in pathophysiological activities and has attracted considerable attention for therapeutic applications [2,3]. RAGE expression is highly downregulated in lung cancers, whereas its ligands are widely overexpressed. In rat lungs, its expression is gradually increased from the fetal stage until birth and is high in the mature stage. However, RAGE is either not expressed in non-small cell lung cancer (NSCLC) tissues or its expression is significantly reduced compared with that in normal lungs, leading to reduced RAGE levels, which contribute to the impairment of cells, differentiation, as well as arrangement of the epithelial structure, with associated oncogenic transformation. RAGE is thought to be associated with the development and invasiveness of lung tumors, as well as angiogenesis, and is thus assessed as a diagnostic marker for lung malignancy. However, Hsieh et al. reported the extensive expression of RAGE and its ligand, S100A6, in human lung cancer, suggesting a possible role of RAGE-mediated signaling in the development of this cancer [4,5,6,7,8,9,10]. Recent findings confirm that RAGE levels are increased in various malignancies, including breast, pancreatic, prostate, colorectal, gastric, and liver cancer [2,11,12,13,14]. Because of its ability to discriminate between a wide range of structurally distinct endogenous and exogenous ligands, RAGE is classified as a pattern recognition receptor (PRR) and damaged-associated molecular pattern (DAMP) [15]. RAGE is present at moderate concentrations in most organs, except the lung, where it is highly expressed in alveolar (AT-1) epithelial cells [16]. RAGE can interact with multiple ligands, including AGEs, high-mobility group box1 (HMGB1), S100/Calgranulins family members, β-sheet fibrils, prions, adhesion molecules, complement components, advanced lipoxidation end products (ALEs), lipopolysaccharide, and others [17,18].
In identical tumor types, increased RAGE expression has been associated with increased tumor histological progression [10,13,19,20,21,22]. In vitro and in vivo experiments revealed that deletion of the short cytoplasmic domain of RAGE exerts a dominant-negative effect, impairing the signal transduction response to the RAGE ligand. The cytoplasmic RAGE domain is strongly charged and is critical for signaling and activation. Ligand–RAGE interaction stimulates cellular transport and many signal transduction cascades, including mitogen-activated protein kinases, phosphatidylinositol 3-kinase, Jak/STAT, and, most notably, the Rho GTPases, Rac-1 and Cdc-42 [11,23,24,25,26,27,28,29]. The expression of RAGE has been shown in a variety of human and mouse tumors, including pancreatic, breast, and colonic carcinomas, fibrosarcoma, and melanoma cell lines. A murine model with high RAGE expression has also been reported. RAGE and its ligand, HMGB1, were also found to be expressed in a murine model [30]. Recent studies suggest that RAGE–ligand interactions affect cellular responses, such as proliferation, survival, migration, and metastasis by activating MAP kinase (Erk1/2), Ras-extracellular signal-regulated kinase, Cdc42/Rac, stress-activated protein kinase/c-Jun-NH2–terminal kinase, and p38 mitogen-activated protein kinase [11,14,20,23,24,27,30,31]. RAGE also induces an NF-κB response. NF-κB is a transcription factor that protects against apoptosis, inducing antiapoptotic genes, such as A1, A20, XIAP, Bcl-2, and Bcl-XL, which are crucial for cell survival under constant cellular stimulation [32,33].
Full-length RAGE comprises a signal peptide (residues 1–22), an extracellular domain (residues 23–342) flanked by V, C1, and C2 domains, a transmembrane domain (residues 343–363), and a cytoplasmic domain (residues 364–404) as shown in Figure 1A,B. The C1 domain has basic amino acids on the surface, whereas the C2 domain is negatively charged, which offers the ability for dimerization of RAGE [34]. Among the RAGE domains, Gln119 in the V domain is implicated in beta-sheet H-bonding with Tyr150 on the C1 domain, exhibiting hydrophobicity between H-bonds. In addition, in the C1 domain, an H2O molecule connects to the amide nitrogen of Ile120 via the backbone atoms of Glu119 and Arg29 in the V domain. As hydrophobic interactions occur between the backbone atoms of Pro215 via the C1 F-G loop and Tyr118 via the C1 B-C loop and Ile91, the V and C1 domains of RAGE are consistently conserved and the VC1 domains have the same components [35,36]. The V and C1 domains, which form an integrated unit, VC1, are involved in the binding of RAGE with its ligands; however, the C1 domain also plays an important role in the identification of ligands [35,36,37,38]. Binding to RAGE involves homodimerization or oligomerization, characterized by different mechanisms that involve amino acid residues in the V and transmembrane domains [36,39]. Because RAGE monomer shows poor binding with monomeric ligands, binding to RAGE requires its oligomerization, which involves high-affinity binding between the receptor and ligand. Oligomerization of RAGE is mediated by the self-association of C1-C1 and C2-C2 domains [35,40,41].
Different isoforms of RAGE, including full-length RAGE, dominant-negative RAGE, N-truncated RAGE, and C-truncated secretory/soluble RAGE, have been reported. Excessive accumulation of RAGE on the cell membrane leads to multiple stimulatory and immunological responses [42,43,44]. RAGE expression is high in vascular and cancer cells. It binds to different ligands including calgranulin family proteins, HMGB1, amyloid β-peptide, β-sheet fibrils, advanced oxidation products (AOPs), Mac-1, and phosphatidylserine. The binding of these ligands to RAGE stimulates MAPK and NF-κB, which induces cellular proliferation as shown in Figure 2A [23,26,45,46,47,48,49]. Moreover, excessive RAGE expression prevents cell death under hypoxia and chemotherapy in cancer cells [27,50]. Studies on diabetes mellitus and hyperglycemia reveal a connection with breast cancer. RAGE, as a PRR, is implicated in inflammatory diseases, and its stimulation in concert with diabetes contributes to the progression of cancer [51,52,53].
Calgranulins are small cell signaling proteins with Ca-binding sites and EF-hand motifs. These S100 family members act as stimulatory ligands that ensure interaction between two or more monomers for intracellular and extracellular processes and are responsible for cell growth, differentiation, and mobility [2,54,55].
The shortage of oxygen in tumors necessitates the glycolytic metabolism of glucose to fulfill the energy needs and is associated with the increased formation of advanced glycation end products (AGEs). Carboxymethyllysine (CML) is a lysine glycation end product that has been identified as one of the most prominent AGEs of tissue proteins. RAGE is a receptor for AGEs in various human malignancies; nonetheless, the presence of AGEs in human malignancies has largely been unexplored [26,45,56,57].
Amphoterin (HMGB1) is an electrically neutral non-histone protein involved in transcription, DNA repair, differentiation, neural development, and extracellular signaling, and has a plausible connection with cancer. HMGB1 is a cytokine released by monocytes and macrophages or other cells, except for apoptotic cells. It is responsible for the pathogenesis of various inflammatory diseases and is involved in the secretion of inflammatory cytokines [10,22,58,59]. The interaction of RAGE/TLRs with HMGB1 is associated with the growth, progression, and metastasis of malignant tumors [60,61]. The binding of HMGB1 with RAGE promotes chemotaxis and the secretion of cytokines, which corresponds with the activation of NF-κB [62,63]. Because RAGE and its ligands are implicated in many malignancies, small molecules that target RAGE and its ligands may help treat various tumors.
In the present review, we discussed various aspects of RAGE and its prominent ligands, which are implicated in cancer pathogenesis, and summarized the recent findings that will provide novel insights into the significant role of RAGE and its ligands in cancer.

2. Immunotherapy for Cancer Treatment

Immunotherapy, in which immune cells are stimulated to destroy cancer cells, has gained significant attention among researchers and physicians as a valuable therapeutic option besides chemotherapy and targeted therapy. Exciting clinical remissions have been reported in incurably ill cancer patients with no other treatment options, which have laid the framework for many innovative cancer immunotherapies that are presently being investigated in clinical trials. However, the treatment outcomes of this remarkable approach are only relevant in 20–30% of all cancers [64,65,66]. Immunotherapy, often known as biological therapy, is a type of cancer treatment that involves the use of chemicals or molecules derived from living organisms (e.g., cancer vaccines, checkpoint inhibitors, immune system modulators, monoclonal antibodies, and T cells Lymphoma). Checkpoint inhibitors are increasingly being used in immunotherapy [67,68]. RAGE and its ligands are located on the surface of cells, including tumor cells and immune cells, providing avenues for cancer therapy using specific inhibitors.

3. Carbonyl Stress and RAGE Ligands in Cancer

3.1. Advanced Glycation End Products (AGEs)

AGEs are products of nonenzymatic glycation, in which the bases of DNA react with reducing sugars, such as glucose, fructose, or deoxyribose, to form a Schiff’s base, which can lead to the formation of an Amadori product through an enamine intermediate. Furthermore, AGEs can also be formed through a variety of different processes, such as the oxidation of sugars, lipids, and amino acids, which produce active aldehydes that eventually become AGEs. The Amadori product undergoes dehydration, cyclization, condensation, and isomerization, culminating in the production of AGEs [69]. The formation of AGEs is typically associated with increased free radical activity. AGEs can modify cell membranes and account for gene alterations leading to the malignant transformation of cells [70]. N-carboxyethyllysine (CEL), N-carboxymethyllysine (CML), methylglyoxal lysine dimer (MOLD), glyoxal lysine dimer (GOLD), and pentosidine are some of the AGEs that have been identified. Regardless of their different nature or the state of cross-links, different AGEs are known to be important in the development of a variety of diseases, including cancer [71]. Diabetes is defined by elevated oxidative stress conditions. Experimental data have demonstrated an increased risk for numerous types of cancers in diabetic patients. Hyperglycemia promotes oxidative stress in various cells through numerous metabolic pathways and causes oxidative DNA damage, which is a precursor to carcinogenesis.
Accumulating evidence implicates AGEs in aging, diabetes, and cancer. The interaction of RAGE with AGEs causes oxidative stress, which in turn promotes the production of AGEs and increases their interaction with RAGE [72]. RAGE–AGE interaction promotes the activation of the PI3K, oncogenic Ras, PKC, and Rho/GTPase (Cdc42 and Rac-1) signaling pathways shown in Figure 2, which leads to cell growth, stress responses, apoptosis, the release of growth factors and proinflammatory cytokines, and motility via changes in cellular properties [73,74]. Clinical evidence shows that individuals with oral cancer who also have diabetes mellitus have a higher risk of cancer spread and have lower survival rates [75]. The binding of RAGE with AGEs in human oral cancer cells is associated with metastasis and angiogenesis [75]. The interaction results in oxidative and glycation stress and its products may have life-threatening repercussions in the lung, oral, prostate, and breast cancers. Pathways, such as p38 MAPK, NF-κB, TNF-α, and AGE–RAGE interactions, may play a vital role in cell growth, propagation, apoptosis, and metastasis [76]. Treatment options remain insufficient for many tumors. We perceived certain important pathways that may play a key role as therapeutic targets in diverse types of malignancies.

3.2. High-Mobility Group Box1

HMGB1 is made up of an N-terminal A segment (residues 1–83), a B segment (residues 88–164), and a C-terminal acidic tail [77]. Receptors for HMGB1—RAGE and Toll-like receptors (TLRs)—exist on the surface of white blood cells as well as on endothelial cells; RAGE and TLRs activate the expression of NF-κB, which is responsible for the activation of genes encoding adhesion proteins as well as inflammatory cytokines and proangiogenic factors. HMGB1 influences the maturation and progression of liver, breast, colon, and gastrointestinal cancers [50,78,79,80,81,82,83]. It regulates the MAPK pathway in gastric, renal, colon, and liver carcinogenesis in mice. HMGB1 is primarily found in the nucleus, although it can translocate to the cytoplasm in response to cellular stress and injury. Through interaction with Bcl-1 and dissociation from Bcl-2, it regulates autophagy and apoptosis [21,79,84,85,86,87].
In vitro and in vivo studies revealed that HMBG-1 promotes PGDF activity and expression of the vascular endothelial factor. The expression of HMGB1 involves various autocrine/paracrine factors, which are responsible for the positive feedback mechanism to its receptors. HMGB1 stimulates and promotes the expression of proangiogenic genes in endothelial cells. It may bind various receptors, including RAGE, TLRs, and syndecan-1 (CD138), which activate NF-κB and ERK1/2 (Figure 2C). It can also bind lipid molecules, such as sulfatide and phosphatidylserine lipids, and can react with lipopolysaccharides and modify CD14 via TLR4-mediated signaling.
Extracellular HMGB1 interacts with RAGE to increase the release of proinflammatory cytokines via NF-κB activation, which is responsible for harmful inflammatory responses [88,89,90,91,92].
The binding of HMGB1 with RAGE through its carboxyl-terminal tail can stimulate RAGE, a criterion for cell motility; its binding to RAGE has a synergistic effect with a dominant inhibitor in stopping the progression of cancer [93]. HMGB1 is characterized by two homologous HMG-boxes, box A and box B, which are linked through linker proteins, approximately 30 and 20 amino acids long. HMGB1 acts as a chaperone to facilitate the incredibly quick binding of transcription factors to DNA [23,94,95]. It plays a very important role in the binding of the tumor suppressor protein, p53, to DNA. HMGB1 decreases p53 expression, which causes DNA damage [94,96,97]. HMGB1 binds to the V domain of RAGE, driving dynamic protein concatenation as well as cell migration via GTPases, such as Cdc42 and Rac; the RAGE binding site is in the HMGB1 box B at amino acid residues 150–183 [98]. In vitro, the binding of HMGB1 with RAGE and its effects through NF-κB have been documented in numerous types of cancers, including human pancreatic cancer (BxPC-3), human hepatocarcinoma (HUH7, H22, HCC), human fibrosarcoma (HT1080), human NSCLC, human bladder carcinoma (5637, BIU (Colo320 and WiDr)), human renal cell carcinoma (CCRCC), murine lung cancer (Lewis cells), mice hepatocarcinoma, and human gastric carcinoma (BGC-823, SGC-7901, MKN-28, and MKN-45); its effects mediated through Bcl-2 have been reported in mouse neuroblastoma (Neuro2a), human neuroblastoma (SH-SY5Y), and human nasopharynges (MCF-7) [11,78,80,84,85,86,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113].

3.3. S100/Calgranulin Family of Proteins

Ca2+ is a second messenger that regulates nerve conduction, muscle contraction, cell motility, gene expression, cell mortality, and necrosis. Ca2+ signaling is controlled by calcium-binding proteins, which regulate Ca2+ concentration in the cytoplasm [87,114,115,116]. S100 proteins are a family of calcium-binding proteins, which are involved in calcium homeostasis [117]. In addition, S100 proteins also bind Zn2+. S100 proteins, such as S100B, S100A6, S100A7, S100A8, S100A9, S100A10, and S100A12, are EF-hand calcium-binding proteins that form homo- and heterodimers. Cysteine and histidine are major constituent amino acids in these proteins. These proteins undergo a structural change upon calcium-binding, which is responsible for protein–protein interaction. The protein–protein interaction site of S100B contains hydrophobic and polar residues required for high binding affinity. S100B functions extracellularly in a cytokine-like manner and shows unique interaction with RAGE [55,118,119,120].

3.3.1. S100A4/Calvasculin

S100A4, also known as calvasculin/metastatin/placental calcium-binding protein/protein Mts1, is abundantly expressed in various cancers. Its expression is associated with malignant growth. It affects various cellular processes, such as angiogenesis, metastasis, and cell growth, and also increases the expression of matrix metalloproteinases [121,122,123,124]. The binding of S100A4 with RAGE increases MAPK/ERK and hypoxia signaling [123,124,125]. In vitro, the activation of NF-κB upon the interaction of S100A4 with RAGE has been reported in various cancer cells, including human osteosarcoma (II-11b), human melanoma (A375, B16-F10), and human pancreatic cancer (BxPC-3) cells [110,126,127,128,129]. The expression of S100A4 is increased in breast cancer in humans, rats, and mice and metastatic cells compared with that in nonmetastatic cells [130,131]. Stimulation of II-11b cells by extracellular S100A4 results in increased NF-κB and JNK activation. S100A4 was shown to stimulate the expression of NF-κB, ERK/2, p38MAP kinase, and JNK in other cell systems. The binding of S100A4 with RAGE was reported to stimulate NF-κB, resulting in an increase in MMP-13 expression in chondrocytes. Also, through its NF-κB promoting activity, S100A4 activates IKK, following its binding to RAGE. S100A4 can promote angiogenesis via annexin II, and through interactions with heparan sulfate proteoglycans, it promotes neurite outgrowth. S100A4 can interact with various receptors to transmit varied biological information [124,132,133,134,135,136,137].
S100A4 has a 12-amino acid calcium-binding domain at the C-terminus and an N-terminal pseudo-EF-hand with 14 residues [127,138]. It is a homodimer with noncovalent interactions between helices 1 and 4, and is responsible for the X-type four-helix bundle; both subunits have two EF-hand calcium-binding domains that have an N-terminal pseudo-EF hand and a C-terminal canonical characteristic EF-hand with a small two-stranded antiparallel β-sheet [139,140,141]. S100A4 undergoes a significant conformational change in the canonical EF-hand upon calcium binding, resulting in the exposure of the hydrophobic binding pocket composed of helices 3, and 4, the hinge region (loop 2), and the C-terminal loop region. In two crystal structures of calcium-bound S100A4, the hydrophobic cleft of each subunit is occupied by the C terminus of an attached dimer, which is responsible for the formation of S100A4 oligomers that were detected extracellularly [142]. The calcium-binding site of S100A4 is located at the C-terminal end of helix 1, and Ca2+ is coordinated via the backbone carboxyl group at Ser20, Glu23, Asp25, Lys28, the side-chain carbonyl of Glu33, and an H2O molecule. The two binding sites are contiguous in space and involve hydrophobic interactions between Lys28 from site 1 and Glu69 from site 2 [125,135,140,141,142].

3.3.2. S100A6/Calcyclin

Homodimeric S100A6 (10–11 kDa) is a calcium-binding protein that is believed to also interact with Zn2+. S100A6 belongs to the calgranulin family and is involved in the propagation of calcium signals. It is conspicuously located in the cytoplasm although it has also been detected in the extracellular matrix and various body fluids. It is highly expressed in the muscles, lungs, kidneys, and brain. The signaling pathway is aided by the extracellular binding of S100A6 with RAGE. Calcium binding induces a conformational change in S100A6, which increases its hydrophobicity and allows for interaction with target proteins. Elevated levels of S100A6 have been observed in epithelial cells, fibroblasts, and several types of tumor cells. The function of S100A6 is unknown; however, it is thought to be involved in cell propagation, cytoskeletal dynamics, and tumorigenesis [25,143,144,145,146].
S100A6 functions as a homodimer formed by noncovalent interactions and binds two Ca2+ per monomer, each through an EF-hand structure composed of two helices connected by a short loop region. S100A6 can also bind two Zn2+ per monomer and can serve as a Zn2+ chelator mostly in the cytoplasm, but also outside the cell. Under conditions of increased Ca2+ concentration, S100A6 can associate with the nuclear envelope and plasma membrane [147,148]. The role of S100A6 signaling in breast cancer has previously been reported; it promotes the function of cacy/SIP, which is related to tumor invasion and metastasis by increasing β-catenin levels. In contrast, S100A6 levels are decreased in human breast cancer cell lines [143,149,150]. The binding of S100A6 with the V and C2 domains of recombinant RAGE has been reported. Studies have shown that the C1 and C2 segments, as well as their associated linker, configure a binding site for S100A6. Isothermal titration calorimetry for the binding of mutant S100A6 with the V domain of RAGE revealed the involvement of basic charged amino acids, Lys44, Arg48, Arg98, Arg104, and Lys107, and Met102, and Gly106, which form a hydrophobic region. Gly47, Leu49, Glu50, Gln100, Asn103, and Asn105 interact with mutant S100A6, allowing proximity of the basic amino acids to adjacent surfaces on the V domain of RAGE. Gln24, Thr28, Arg62, Asn63, Lys64, Gln66, Asn69, and Phe70 are the S100A6 amino acid residues in the protein–protein interface as shown in Figure 3A,B [151,152]. S100A6 is overexpressed in breast, stomach, and pancreatic cancers, as well as in thyroid carcinoma, clear cell renal cell carcinoma, and mixed lineage leukemia. Its expression is low in prostate and oral cancer, and it is used as a diagnostic or predictive marker in pancreatic, gastric, and prostate cancers, melanoma, NSCLC, and hepatocellular carcinoma [54,153,154,155,156,157,158].

3.3.3. S100A7/Psoriasin

S100A7/psoriasin is a 12 kDa EF-hand calcium-binding protein in the S100 family that promotes cell migration through RAGE in a zinc-dependent manner. Extracellular expression of S100A7 is primarily associated with squamous cell cancer subtypes, namely lung cancer, head and neck cancer, cervical cancer, and bladder cancer, as well as with nonsquamous cancer subtypes, such as melanoma, breast cancer, and gastric cancer. S100A7, like S100A6, occurs in a homodimeric form with two Ca2+ EF-hand motifs and two Zn2+ ions at the binding site that span the dimer interface. S100A7 has been postulated to be involved in cell propagation, relocation, invasion, and tumor metastasis. Its expression is higher in cervical cancer compared with that in normal cervical tissues. The binding of S100A7 with RAGE activates the ERK signaling cascade, promotes cell mesenchymal competence, and induces epithelial–mesenchymal transition. Human S100A7 induces IL-1α expression in keratinocytes through RAGE-p38 MAPK-calpain-1 signaling (Figure 1A,B), and psoriasis-associated cytokines, such as IL-17a, IL-22, and IL-36, may upregulate S100A7 expression in keratinocytes [159,160,161,162,163,164,165,166,167,168,169,170]. Each monomer of the homodimer begins with helix I (Gln4 to Lys18), which leads through the first loop (Tyr19 to Asp27) into helix II (Lys28 to Phe39) where Pro40 results in a breach in the systematic helical pattern and a small turn linker helix II′ (Asn41 to Lys48) ensues in a different direction; this is followed by helix III (Tyr53 to Lys61), the calcium-binding loop (Asp62 to Asp72), and helix IV (Phe71 to Gln88). The zinc-binding site on S100A7 is mostly comprised of two histidines, His86 and His90, in the C terminus, and is located next to the end of helix IV [170].

3.3.4. S100A8/Calgranulin-A and S100A9/Calgranulin-B

Cytosolic calcium regulates various cellular functions. Notably, calcium-binding proteins are important molecules in signal transduction, differentiation, and cell cycle regulation. S100A8/CACA/CFAG/MRP8 (10.8 kDa) and S100A9/MRP14 (13.2 kDa) are calcium-binding proteins expressed by neutrophils and activated monocytes. At basal concentrations, S100A8 and S100A9 display growth-promoting effects upon binding RAGE. Their binding to RAGE is implicated in breast cancer metastasis and stimulates the phosphorylation of LIN-11, IsL-1, and MEC-3 protein domain kinase, as well as cofilin. The binding of S100A8 and S100A9 with RAGE stabilizes Snail via NF-κB, enhances lung metastasis, and improves mesenchymal properties [171,172,173]. In vitro and in vivo data reveal homodimerization of S100A8 and S100A9, forming calprotectin; the resulting isoform may play a variety of roles in phagocyte physiology. S100A8 and S100A9 are found in the cytoplasm of neutrophils, monocyte membranes, plasma, and other body fluids. Calprotectin expression is associated with rheumatoid arthritis, autoimmune diseases, cystic fibrosis, chronic bronchitis, acute allograft rejection, gut irritation, inflammatory dermatoses, and abscesses, and it is a relevant marker for medical diagnosis [174,175,176,177,178]. RAGE is a key receptor of S100A8 and S100A9 in tumor cells, whereas TLR4 is a major receptor for both these ligands in macrophages.
Carboxylated N-glycans are expressed on RAGE, for example, on immune and tumor cells, which mediate S100A8/A9 and RAGE binding, promoting receptor-mediated signaling and pathogenesis. In colon cancer, S100A8 and S100A9 induce RAGE- and carboxylated glycan-dependent phosphorylation of ERK1/2 and SAPK/JNK, but in prostate and breast cancer, they promote p38 phosphorylation. The binding of S100A8/S100A9 on RAGE and carboxylated glycan in colon cancer activates the NF-κB pathways, resulting in a life-threatening link between inflammation and cancer. S100A8 and S100A9 are recognized as novel target genes in hepatic carcinoma cells during inflammation-mediated liver carcinogenesis [179,180,181,182,183,184,185]. Individually, S100A8 and S100A9 domains for the calcium-binding loop are fully conserved, and Ca2+ binds to the EF-hand I, which exhibits additional varied sequences correspondingly, as in the crystal structures with specific ligands. In the homodimers of S100A8 and S100A9 with calcium-binding sites at the N terminus occupied, each monomer is held together by a hydrophobic core of conserved sequences. Homodimers are stabilized by interactions of hydrophobic sidechains among these helices. S100A8 homodimer interaction takes place at the surface of each monomer between Leu72 and Ile76, whereas S100A9 interaction takes place on S100A8 at the site of Ile76 in a single chain that connects two helices. S100A9/S100A8 connection provides extra balance to the S100A9 subunit and promotes transition for a stretch of 6–14 amino acid residues at the C-terminal end, α-helix customs a disorganized state to α-helical configuration through increased electron compactness. S100A8 and S100A9 heterodimers have disulfide bridges between S100A9 at Asp30, and in S100A8 at His83 and His87, these amino acids participate in the binding of two Zn+ ions [186,187,188,189].

3.3.5. S100A14/S100 Calcium-Binding Protein A14

S100A14 is an 11.6 kDa protein belonging to the S100 family. It has an N-glycosylation site, a protein kinase phosphorylation site, and an N-myristoylation motif [190]. S100A14 is highly expressed in some types of cancers, including ovarian, breast, and uterine cancers, and at low levels in the kidney, colon, and rectal cancers [191,192]. The binding of extracellular S100A14 with RAGE has been shown to control proliferation and apoptosis in esophageal cancer cells, and cell proliferation and invasion in oral squamous cell carcinoma [193,194,195]. Although the expression level and living purposes retain a tissue or malignancy character, S100A14 has been reported to remain contrary expressed in several human melanomas responsible for diverse life progression including propagation, apoptosis, cell motility keratinocytes variation, and motion transduction. S100A14 triggers the MAPK pathway, which causes lung adenocarcinoma, epithelial-mesenchymal transition cervical cancer cells, and metastatic breast cancer cells to proliferate. The interaction of RAGE and S100A14 is associated with increased cancer growth. However, S100A14 has been found to hinder gastric malignancy metastasis by hindering Ca2+ entry by limiting the FAK signaling pathway and MMP expression. In vivo and in vitro data demonstrate that S10014 inhibits NPC cellular motility via the NF-κB signaling pathway; moreover, it has been discovered that overexpression of S100A14 lowers the risk of NPC resistance to cisplatin [192,196,197,198,199,200]. Although the involvement of S100A14 in the translocation from the cytosol to the plasma membrane in breast cancer is well known, S100A14 has also been discovered to play a dynamic role in bladder cancer and growth. S100A14-related genes may be useful for detecting cancer in the peripheral blood of patients with advanced cancer. In vivo data reveal that the S100A14 gene is regulated by p53, which is associated with esophageal squamous cell cancer. Exogenous S100A14 ligation with RAGE on ESCC cell lines reported that a low dosage of exogenous S100A14 activates ERK1/2 and NF-κB signaling, stimulating cell proliferation or increasing cell endurance, as shown in Figure 1A.
On other hand, a higher amount of S100A14, can cause apoptosis and increase the generation of reactive oxygen species. Exogenous S100A14 stimulates cell proliferation or promotes apoptosis at different doses based on in vitro findings [201,202,203,204,205,206]. An S100A14 monomer interacts with aromatic amino acids in helix I at position Phe29 and in helix IV at positions Phe80, and Phe84, and additionally Trp85, which aids in helical orientation and maintains contact between two subunits. Numerous hydrophobic residues make the monomer–monomer on the helix I connection through helix I0 and helix IV0, and the various connection link residues in helices IV and IV0 become constant. The association between two monomers was intended to be enhanced by residues 53–57 in the hinge loop, which was recognized by residues at the N-terminal of helix I0 and the C-terminal segment of helix IV. As a result, it is said that S100A14 takes a conformation distinguished from that of the canonical apo arrangement, and that the Ca2+ loaded from helices II and III are almost antiparallel, employing the canonical apo arrangement of S100 proteins. However, the superimposition of the current structure of S100A13 with the structures of the apo and holo forms of S100A13 reveals a well-structured fold that used the last N-terminal of S100A14 with 13 residues for the Ca2+-binding loop, and another Ca2+-binding loop reported at the end C-terminal of S100A14 [190,207,208,209,210,211,212].

3.3.6. S100A16/S100 Calcium-Binding Protein A16

S100A16/S100F is a 10–12 kDa protein with the EF-hand Ca2+ binding motif significantly regulated in lung, ovarian, prostate, and breast cancers [140,213,214]. S100A16 mRNA has been found in a variety of tissues, including the brain, as astrocyte-specific glioblastoma cells, where it has been shown to accumulate in nucleoli and translocate to the cytosol in response to Ca2+ stimulation, implying that it may perform a role in ribonucleoprotein complex processing, gene silencing, or cell cycle development. The identified nuclear localization signal is absent in S100A16, as it is in other members of the S100 family. Phosphorylated S100A16 protein has been discovered in the nucleoli of HeLa cells, indicating that phosphorylation of S100A16 may play a role in nuclear translocation. S100A16 binds a single Ca2+ monomer at each atom, and it also binds Zn2+. However, the Ca2+ and Zn2+ binding sites are at distinct sites [215,216,217,218]. S100A16 expressions, mutually with S100A14, have been linked to a dangerous condition in patients with breast cancer; both proteins, in cooperation, can increase the aggressive activity of breast cancer cells via cytoskeleton functions. In MCF-7, high S100A16 accumulation supports epithelial–mesenchymal transition via the Notch-1 pathway, whereas low accumulation in oral squamous cell carcinoma patients is associated with deficient progenesis. It has been shown that the expression S100A16 in various malignancies is highly correlated with high prognostic outcomes. S100A16 in comparison to S100A14, in single cooperation in oral squamous cell carcinoma, indicated that S100A16 may serve the same purpose as S100A14. S100A14 is a prominent ligand of RAGE signaling pathways, and S100A16 is the most associated protein, implying that S100A16 has a role in tumor invasion and development [218,219,220,221,222]. Two common cell signaling pathways through serine/threonine kinases show S100A16 in prostate malignancy: MAPK/ERK and PI3K/AKT (Figure 1A) [223,224]. Consequently, with the suppression of p53 expression, S100A16 has been shown to promote metastasis. The high expression of S100A16, assumed in the relocation, invasion, and proliferation in human prostate cancer cells, was discovered in vitro [225].
Interestingly, the S100 family proteins have two sites for Ca+ binding, one at the EF-hand C-terminal and the other at the S100 short N-terminal. S100 family proteins have highly conserved Ca+ ligand amino acids at 1, 3, 5, 7, and 12 locations, which generate an enhanced attraction for metals. The backbone oxygen atoms of the residues at the positions of 1, 4, 6, and 9, as well as two side-chain oxygen atoms of the residues at the 14 locations, which is generally glutamate, are placed at the position of the 14th residue. However, due to the absence of glutamate at the comparable location S100A16, the Ca+ binding affinity is reduced by the S100A16 shortage of glutamate by the side of this site. In addition, the N-terminal has 15 amino acids rather than 14, which is unique to S100A16. Dimerization of Apo S100A16 with Ca+-loaded S100A16 generally occurs across communications between helices I, I0, IV, and IV0, which is responsible for forming an X-type helix package. Trp80 and Ile83, two hydrophobic amino acids in helix IV0, form many contacts with Leu8, Val12, and Leu15 in helix I0, as well as Trp80 and Ile83 in helix IV0 of the other subunit. Amino acids at positions Glu45, Leu46, His48, and Met49, in the hinge loop between helices II and III, form connections with amino acids towards the N-terminus of the subsequent subunit of helix I0. The connections in a dimeric arrangement of S100A16 align helices I and IV in opposing orders to helices I0 and IV0, respectively [226].

3.3.7. S100B/S100 Calcium-Binding Protein B

S100B is a 10.7 kDa protein, and its expression has been reported in many disorders, including neurological diseases and cancer. High S100B absorptions continue to be used as a biomarker for malignant melanoma, with the increasing amount of S100B indicating cancer progression. High expression of S100B is still linked to unfavorable liver metastases and early deterioration in colorectal cancer. On other hand, high expression of S100B reveals ovarian malignancy-associated utilizing primary cancers. By activating NK-кB, S100B ligation with RAGE improves cell survival. S100B, a RAGE ligand that promotes tumorigenesis by promoting IL-6 expression and STAT-3 activation, is abundantly expressed in gliomas [227,228,229,230,231,232,233,234,235]. Based on the cell nature, RAGE, in conjugation with S100B and S100B, can activate STAT-3 through phosphorylation. S100B ligation with RAGE, a primary controller designed for downstream Akt-1 and STAT-3 activation, has been observed (Figure 1A). The results indicated that the C6 glioma-conditioned medium can promote the malignant transformation of mesenchymal stem cells and that this transformation is mediated by S1001B interaction with RAGE [120,236,237]. S100B has received much attention because of its binding to p53, a tumor suppressor gene in cancer that lowers the p53 protein level, perhaps hindering wild-type p53 malignant development [238,239,240,241,242,243]. S100B interaction with p53 causes Ca+-dependent progression; nevertheless, S100B ligation with Ca+ causes conformational rearrangement, causing several hydrophobic amino acids to become solvent-exposed. As a result, an interaction flanked by two associate proteins has been identified in rat S100B in multipart, using the C-terminal negative regulating domain, which comprises the amino acids in helices III and IV, and the S100B hinge loop [244,245]. Ca2+ binding reveals a significant hydrophobic grove near helix III on S100B. Homodimeric S100B binds two Ca2+, one at the position of the hinge-connected C-terminal and another at the site of the N-terminal EF-hand, indicating that many ligands result in RAGE-mediated signal transduction [38,39,246,247]. S100B can bind to Zn2+, Cu2+, and Ca2+; therefore, Zn2+ interaction with S100B increases the attraction, as mentioned above, to Ca2+, particularly in the peptide and protein function [38,39,246,247,248,249]. S100B ligation, in combination with RAGE activation, promotes oxidative stress through the activation of ERK1/2, p38, and JNK MAP kinase pathways [143,250,251]. S100B has a 12 amino-acid loop with acidic amino acids required for Ca2+ organization at Asp61 on the X-axis, Asp65 on the Y-axis, and Glu72 on the Z-axis, which has many homologous sequences in the S100 family Ca2+ binding EF-hand ligand. Helix III, which closely follows in the loop in human S100B, causes a conformational change in the C-terminal loop with Ca2+ binding, resulting in a redirection of helix III involving helices II and IV, as seen in rat apo S100B [210,211,252].

3.3.8. S100P/S100 Calcium-Binding Protein P

S100P, a RAGE ligand, has been found in prostate, pancreatic, lung, breast, and colon cancers [253,254,255,256,257]. The interaction of RAGE with S100P stimulates major signaling pathways, including ERK1/2, NF-κB, and JAK/STAT, which increased S100P levels in colon cancers linked to metastasis (Figure 1A) [30,256,258,259,260]. S100P is highly expressed in pancreatic tumors, where it regulates multiple intracellular and extracellular processes, including cell proliferation, survival, treatment resistance, and diversity. S100P, on the other hand, includes the genesis of cancer and plays an important role in cancer progression. In vitro findings illustrate that the invasiveness is reduced by silencing S100P by employing siRNA, and in vivo findings illustrate a reduction in metastasis by silencing the S100P by using siRNA [31,256]. In vitro studies show that S100P is expressed in human colon cancer and stimulates cell growth, cell relocation, ERK phosphorylation, and NF-κB activation; these activities were mediated through RAGE [211]. S100P includes multiple α-helical concerns, with four systematic α-helical segments inserted at Glu3 to Ser19, Lys30 to Glu40, Ala53 to Ala63, Glu71 to His86, and another α-helical segment introduced in the linker region at Phe44 to Gly48. The Ca2+-binding loop I, on the other hand, attaches to helices I, one at Glu3 to Ser19 and the other at Lys30 to Glu40, forming the N-terminal EF-hand, whereas helices III at Ala53 to Ala63 and helices IV at Phe71 to His86 are linked via the Ca2+-binding loop II, forming the C-terminal EF-hand. The Ca2+-binding loop forms the N-terminal EF-Hand I attaching to helices I, one at Glu3 to Ser19 and the other at Lys30 to Glu40, whereas the C-terminal EF-hand is formed by the Ca2+-binding loop II connecting helices III at Ala53 to Ala63 and helices IV at Phe71 to His86. There appears to be no change in preceding Ca2+ binding in the S100 family, but there is a change in the relative position of loop I to helices I and II. Phe44 to Gly48 participate in a short α-helix that connects with two Ca2+ binding domains at Leu41 to Asp52 [261,262]. The hydrophobic surface of S100P at Tyr88 and Phe89 shows a substantial feature in the recognition of a target protein compared to other S100 family proteins, which play a crucial part in the acknowledgment of target proteins. S100P reveals the central linker region at sites 43 to 49, the amino acid contained at sites 83 to 94 in helix IV of the first monomer, and the 2nd to 14 amino acid of helix I, which is another monomer that indicates a prolonged surface binding directed at the RAGE V-segment. S100P hydrophobic residues are nonpolar amino acids of S100P, such as Ala92, Gly93, Phe89, Tyr88, Gly9, Ile12, Met8, Met1, Phe44, Gly43, Pro42, Leu41, and Gly48, that bind on the V-segment of the RAGE. The charge–charge interaction involving the acidic amino acids at the position of Glu5 and Asp13 represents the V-segment of RAGE to S100P, which may contribute to the maintenance of the complexes. Thr6, Thr2, Ser47, Gln46, and Cys85, as well as neutral and polar amino acids, make non-hydrophobic interactions with the RAGE V-segment. Amino acids on the RAGE V-segment that interact with the S100P ligand include the basic amino acids Arg48, Lys52, Lys62, Arg98, Arg104, and Lys110, which are responsible for the inter-spread of basic patches, as well as amino acids such as Leu53, Trp61, Val63, Pro66, Gly68, Gly56, Met102, and Pro71 as shown in Figure 4A,B [263,264].

3.4. Adhesion Molecules

Cell adhesion molecules are life-threatening in tumor growth and are classified into several functions and classifications. Cell adhesion molecules of various classes and roles can collaborate and temper the signaling purpose of receptor tyrosine kinases and soluble proteins in the extracellular matrix, accompanied by cadherins, integrins, immunoglobulin family CAMs, and CD44 [265]. They play a crucial role in cancer growth and metastasis in pancreatic cancer, tumorigenic cancer stem cells, melanoma, breast, lung, and oral cancer. [266,267,268,269]. Although RAGE has been suggested to maintain lung homeostasis by arbitrating cell adhesion molecules, the ERM protein family (ezrin, radixin, and moesin) has a strong cross-link between the cytoskeleton and the plasma membrane. It can interact with transmembrane proteins and the cytoskeleton, forming structural linkages that help to improve the cell cortex and control signal transmission pathways. According to a new study, the interaction of RAGE with ERM appears to be essential in regulating EMT-related structural alterations in alveolar epithelial cells. In pro-inflammatory cytokines that support EMT of human alveolar epithelial cells, ERM interaction with RAGE shows important stages in the rearrangement of the actin cytoskeleton [270]. RAGE shares substantial structural features with adhesion molecules such as ALCAM (CD166), BCAM (CD239), and MCAM (CD146, Mel-Cam, Muc18, and S-Endo1), according to recent findings [271]. MCAM has been linked to advancing melanoma, prostate cancer, and breast cancer. Members of the IgSF (immunoglobulin superfamily) association have been identified as biomarkers for cancer development.
Similarly, IgSF associates such as L1CAM, NCAM, PECAM-1, ALCAM, and ICAM-1 are associated with metastatic development in melanoma, glioma, breast, ovarian, endometrial, prostate, and colon cancer [272,273,274,275,276]. In colorectal cancer, cell adhesion molecules play an important role in determining the prognosis of this common disease. However, studies on adhesion molecule interactions with the RAGE axis may play a role in cancer progression and metastasis, and the machinery that causes the most significant reorganizations of the submembrane cytoskeleton is poorly understood, which could be a therapeutic target for cancer.

3.5. Complement Component

The complement component is a danger-sensing organization for external pathogens and endogenous modified characters. Although there is no conclusive proof, the complement component has a function in tumor resistance, according to available studies. The complement component, on the other hand, appears to be more effectively induced in tumor cells and their non-malignant specimens [277]. Even though each tumor has its unique profile of antigens and complements, the fascinating categorization of membrane components responsible for complement activation, and their pathways, is unclear. Tumors employ a variety of inhibitory mechanisms, including the articulation of CD35, CD46, CD55, and CD59 (membrane-bound proteins) with factor H or factor-H similar proteins, factor I, and C4b-binding protein, to alter complement activation (soluble regulatory proteins) [278,279,280,281]. Both complement stimulation results, anaphylatoxins C3a and C5a, can maintain chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and increase cancer cell motility and spreading potential [282]. The complement component, which functions as a crucial character in both adaptive and innate immune systems, can work with the activator of immunoglobulin and non-immunoglobulin complement systems. By collaborating with a wide range of surface molecules, C1q plays a supervisory function in cellular processes [283]. C1q also helps other dangerous immune functions, such as allowing the apoptotic to be activated and modulating cellular activities within adaptive immune responses, and an integrin b2, also known as MAC-1, can bind to the collagen domain of C1q, while RAGE, as a receptor for Mac-1, may generate a complex structure that can enhance the binding affinity [284,285,286,287]. RAGE acts as a receptor for the globular domain complement component C1q, which improves C1q-mediated phagocytosis by allowing for critical interactions in response to RAGE and C1q [288].

3.6. Advanced Lipoxidation end Products (ALEs)

Advanced lipoxidation end products (ALEs) are a collection of adducts and cross-links formed by the nonenzymatic response of reactive carbonyl species (RCS) produced by lipid peroxidation, lipid breakdown by nucleophilic sites of proteins (primarily Cys, Lys, His, and Arg residues), amino phospholipids, and DNA [289]. Lipoxidation of a protein is a nonenzymatic post-translational modification that occurs when reactive lipid molecules are covalently incorporated into proteins. Studies have correlated protein lipoxidation to cancer, neurodegeneration, and atherosclerosis [290].
Lipoxidation is a well-known process between electrophilic carbonyl species and certain proteins, which is characterized by the oxidation of lipids and, in some situations, the alteration of protein characteristics. Studies on lipoxidation have revealed the impact of cancer growth on tumor cells, the change of immune elements, and subsequent immune response stress. Protein adducts disrupt a variety of proteins in cancer that can activate various types of machinery, such as cell division, differentiation, and death. In in vivo findings on the human colon, adenocarcinoma showws a reduced appearance of TGFb-1 and a scarcity of 4-HNE protein adducts, implying that neoplastic evolution is healthier under these conditions. Meanwhile, TGFb-1 and 4-HNE collaborate to promote colon cell apoptosis by activating the JNK pathway [291,292]. At normal conditions, the adducted residues are in their neutral configuration, a proportional change in their ionization state that weakens the ionic bridges and makes nearby anionic residues more approachable and accessible to relieve ion pairs with the RAGE-positive residues. The ability of ALEs to interact with RAGE protein is mostly determined by the type of the adduct and the modification site [293].

3.7. Lipopolysaccharide

Lipopolysaccharide is a bacterial antigen that plays a vital role in cancer progression by encouraging the release of IL-8 and TGF-1, which promotes the growth and invasion of liver cancer cells, colorectal cancer cells, and leukemia cells [294,295,296,297]. TLR4 is a major LPS receptor, and RAGE can interact with LPS and cause pro-inflammatory signaling [298]. TLR4 is an important LPS receptor, and RAGE can interact with LPS and cause pro-inflammatory signaling [298,299]. According to the latest in vitro studies in breast cancer cells, LPS increases the expression of S100A7, which inhibits TLR4 and promotes RAGE accumulation in breast carcinogenesis. In vitro, treatment of LPS on an S100A7 overexcited in breast cancer cell line (MDA-MB-231), and in vivo treatment of MVT-1 (mouse mammary tumor cell line) with mS100A7/A15 revealed enhanced RAGE manifestation in a dose-dependent and time-dependent manner [75,300]. Another study found that LPS-induced inflammation of normal cervical epithelial cells promotes the malignant transformation of the cell. Additionally, the LPS may promote the proliferation and invasion of normal epithelial cells; the accumulation of IL-1, IL-6, and TNF were promoted in cervical epithelial cell lines, whereas RAGE inhibitors altered the LPS [301].

4. RAGE-Specific Signaling in Cancer Progression

RAGE–AGE interaction enhances the activation of the PI3K, oncogenic Ras, PKC, and Rho/GTPase partners (Cdc-42 and Rac-1) signaling pathways, which is mandatory for cell survival, stress responses, apoptosis, the release of growth factors, proinflammatory cytokines, and motility (Table 1) [21,207,301,302]. A protein initiates the release of IL-1, called the nucleotide-binding oligomerization domain-like receptor. Whereas IL-1, on the other hand, stimulates the nuclear translocation of HMGB-1 to the cytoplasm. Extracellularly, HMGB-1 can interact with RAGE and TLRs to trigger the release of proinflammatory cytokines, such as NF-κB, and elicit physiological responses. HMGB-1 tempted signaling through surface receptors, such as receptors for advanced glycation end-product and toll-like receptors, can discharge pro-inflammatory cytokines and chemokines, which include detrimental inflammatory responses, according to in vitro and in vivo investigations [19,50,91,92,106,111,303,304]. Intracellular signaling of HMGB-1 via RAGE and TLR activates many signaling pathways, including MAP kinases and JNK, as well as NF-κB translocation and inflammation [126,305]. In CRC cell lines, S100A4 interacts extracellularly with RAGE and impacts cell motility and metastasis, priming MAPK/ERK hyperactivation and hypoxia signaling (Figure 2E) [127]. Previously, in vitro studies suggested that S100A4 interaction with RAGE are involved in various cancers and activate several signaling pathways, including NF-κB upregulation in human osteosarcoma, human melanoma, and human pancreatic cancer, and ERK or Cdc42 upregulation in human colorectal carcinoma and thyroid cancer [127,203,299,305,306]. The combination of S100A6 and RAGE in vivo has been shown to produce nasopharyngeal cancer by upregulating p38 [170]. RAGE and S100A7 interaction were discovered to be involved in breast cancer and human cervical cancer through upregulation of ERK, MMP9, and NF-κB in vivo and in vitro investigations (Figure 2E) [171,182,307]. The upregulation of NF-κB, p38, ERK1/2, SAPK/JNK and the upregulation of p38, p-erk1/2 in human hepatocellular carcinoma can cause human breast cancer, prostate cancer, colon tumor, oral, esophageal tumor, and squamous carcinoma.
In vitro and in vivo findings of S100A8/9 interaction with RAGE can cause human breast cancer, human prostate cancer, colon tumor, and oral and esophageal tumors [225,308,309,310,311]. The overexpression of ERK1/2, NF-κB, and Akt, as well as the downregulation of p21 and p27, have been found to have a role in diverse malignancies, such as esophageal squamous cell carcinoma and human prostate cancer in vitro [235,312]. In vitro data on the interaction of RAGE with S100B/P suggest a role in ovarian cancer, glioma (C6), and human colon adenocarcinoma by downregulating p53 and upregulating Akt1, STAT3, and in nasopharyngeal carcinoma, colon cancer, and pancreatic cancer by upregulating NF-κB, ERK1/2, MMP2, and MMP9 [256,298,299,313,314,315,316]. According to recent in vitro studies in breast cancer cells, LPS increases the expression of S100A7, which inhibits TLR4 and promotes RAGE accumulation in breast carcinogenesis. In invasive breast cancer cells, RAGE interaction with S100A7 mediates oncogenic response; in vitro, LPS treatment of S100A7 rose in a breast cancer cell line (MDA-MB-231). In vivo treatment of the mouse mammary tumor cell line (MVT-1) with mS100A7/A15 revealed enhanced RAGE expression in a dose- and time-dependent manner [23,112].

5. Anti-RAGE Therapeutics in Cancer Management

In vitro experiments with S100 proteins and HMGB-1 in mutual ligation with RAGE stimulate HMGB-1 ligation on RAGE. Likewise, via S100P, the first 40 amino acids of HMGB-1, the HMGB-1 derivative peptide, can prevent the ligation and activation of RAGE. RAGE inhibitor peptide sequences occur at residues 32–42 on S100P in the same region as many other S100 family members. RAGE inhibitor peptides can disrupt the connection between S100P, HMGB-1, and S100A4 on RAGE as shown in Figure 5 (Table 2) [31,317,318]. The impact of RAGE inhibitors on various cancers is described further down. RAGE inhibitors have been investigated for their potential to inhibit cancer progression. Some of these are described below.

5.1. siRNA Inhibitor

RAGE has been identified as an oncogenic protein that promotes the development and spread of various malignancies. However, the migratory and invasive abilities of NSCLC cells are reduced when siRNA inhibits RAGE. RAGE is also involved in EMT, reducing RAGE and initiating transmutation from mesenchymal to epithelial signals in cell lines. Moreover, we found RAGE to be a factor in epithelial–mesenchymal transition (EMT), with RAGE deficiency resulting in the transmutation of mesenchymal to epithelial markers in cell lines. According to in vivo data, siRNA can decrease RAGE and tumor growth and proliferation [320,321,322]. In NSCLC cells, siRNA-suppressed RAGE inhibits PI3K/AKT activity, impacts downstream mTOR/p70S6K signaling, and reduces the phosphorylation initiation of KRAS and RAF-1 at the same time. As a result, RAGE may have a role in NSCLC growth, metastasis, and other malignancies [322] and siRNA targeting of RAGE might be useful as a therapeutic approach for NSCLC.

5.2. shRNA Inhibitor

RAGE accumulation is associated with increased microvessel efficiency in colorectal tissue samples, and the familiar method can reduce VEGF expression by silencing RAGE expression, implying that RAGE plays a role in VEGF expression and blood vessel formation in colorectal cancer. Furthermore, the VEGF gene promoter’s convenient domain has significant GC-rich motifs that may be modulated by transcriptional factor explicitness (SP1) [323,324]. According to the findings, silencing RAGE affects a variety of stages of colorectal cancer angiogenesis, including an SP1 decrease and VEGF articulation [325,326].

5.3. RBGO1 and ADC

In contrast to RAGE, RGBO1 is a monoclonal antibody. KLH (keyhole limpet hemocyanin) linked RAGE was injected into mice, and the KLH coupled peptides corresponding to amino acid residues 198–217 or 327–344 of the RAGE protein. Replicas were selected based on a positive ELISA screen utilizing BSA (bovine serum albumin) linked peptides. RGBO1 is a clone of the full-length RAGE protein [145]. In vivo efficacy was determined by injecting RBGO1 ADC (3 mg/kg) or MCF (45 g/kg) into female nude mice with 5mm HEC1A xenograft tumors on a two-time weekly basis for four weeks. As a result, whereas RGBO1 therapy alone had no significant effect on cell survival, RGBO1 treatment on MCF was determined to be effective.
Furthermore, RBGO1 ADC is effective in vivo at a 7.4 µg/ml concentration. The RAGE importance on RBGO1 ADC was significantly more potent than the same ADC focus on HER2. RAGE-directed efficacy of ADCs is based on V-domain binding, and RGBO1 ADC is 200-fold more effective than cytotoxic drug therapy alone [13]. These findings suggest that using RAGE-targeting RGBO1 ADCs as a treatment for endometrial cancer is highly effective.

5.4. Sorafenib

Sorafenib, a kinase inhibitor, has been approved to treat early-stage kidney cancer, advanced liver cancer, and thyroid carcinoma. RAGE is overexpressed in both tumor and stromal cells; however, RAGE accumulation is more prominent in tumors, and RAGE targets in HCC cell lines can enhance autophagy [327]. RAGE activation by the AMPK/mTOR pathway triggers autophagy. Furthermore, sorafenib promoted apoptosis by modifying AMPK activity and preventing RAGE. By regulating autophagy, RAGE promotes HCC proliferation and sorafenib resistance. An in vitro investigation identified HCC and sorafenib resistance as probable causes [327].

5.5. Duloxetine

Duloxetine, commonly known as serotonin/noradrenaline, is a reuptake inhibitor (RI) used to treat depression and neuropathic pain. It has also been employed for the treatment of chemotherapy-induced acentric neuropathy [321,325]. Gao et al. examined GL261 cell lines of mouse glioma cells using duloxetine in a dose-dependent manner (30 mg/kg) by way of oral feeding for 14 days and concluded that duloxetine has the potential to suppress S100B production as well as the inhibition of GL261 glioma cell proliferation within the cranium [113].

5.6. Heparin

In various studies, an anti-thrombotic medicine, achieving the perfect anti-coagulant, was critically employed for modern hematology and oncology with the affected patient, who was at the greatest risk of bleeding and circulatory thromboembolism through RAGE of an NF-κB dependent on luciferase reporter assay and the human fibrosarcoma cell line (HT1080); heparin decreased the HMGB1 level and encouraged the activation of NF-κB (Figure 5). Heparin, but not HT1080mock or HT1080dnRAGE cells, suppressed the relocation, invasion, tumor formation, and lung metastasis of the human fibrosarcoma cell line (HT1080RAGE cells) [23]. According to reported data, heparin had therapeutic potential in individuals with specific types of malignant tumors, such as chondroitin sulfate and heparan sulfate-directed RAGE, suggesting that lung metastasis was reduced [110,326].

5.7. S100P-Derived RAGE Inhibitor

The HMGB-1 antagonist peptide sequence was compared to the S100P sequence and a resemblance was found in amino acid sequence and pattern between the first 40 residues of S100P and the 33 residues of RAGE-obligatory peptide derived from HMGB-1. The discovery of this likeness resulted in the modeling of three peptides from the S100P sequence, each of which was 95% pure. The peptides were designed with the amino terminus by amidation, and the carboxyl terminus by acetylation to improve their consistency. A RAGE inhibitor derived from S100P can prevent the adherence of various RAGE ligands involved in inflammation and cancer. In vivo data revealed that the S100P-derived inhibitor may have therapeutic potential against glioma and pancreatic cancer and may inhibit a variety of RAGE ligands, implying that it could be useful in additional RAGE-related diseases [3,85].

5.8. FPS-ZM1 and LY294002

RAGE overexpression stimulates cervical cancer cell proliferation and increases the appearance of proliferating cell nuclear antigens. Meanwhile, RAGE overexpression inhibited cells, resulting in apoptosis suppression, a reduction in Bax/Bcl-2 levels, and activation of PI3K/AKT. SiHa cell capability and PCNA appearance were decreased by FPS-ZM1, a RAGE inhibitor, which also increased cell death and Bax/Bcl-2 levels. The PI3K inhibitor LY294002 decreased PI3K and AKT activation and RAGE accumulation, promoting cell proliferation and death [3]. It has a wide range of RAGE ligands, suggesting that it may be effective in other RAGE-related disorders.

5.9. Gefitinib

HMGB1 has a key function in autophagy stimulation in vital carcinogenesis, and it is mostly found in the cytoplasm of human gastric cancer cells with cell death and survival supervisory body. Gefitinib is an EGFR inhibitor, and HMGB1 release from tumor cells treated with gefitinib promotes autophagy, interacts with RAGE, and induces ERK1/2 signaling in gastric cancer cells. HMGB1 inhibitors, such as gefitinib, and RAGE inhibitors might play an essential role in preventing cancer regrowth, as suggested by HMGB1-related autophagy in chemotherapy [41].

5.10. Metformin

Metformin use has been linked to duction in the occurrence and mortality of breast cancer in diabetics patient. AGEs appear to have accelerated MCF-7 cell proliferation, which was inhibited by 0.01mM or 0.1mM metformin or anti-RAGE antibodies. Metformin, at a dosage of 0.01 mM, also suppressed the AGE-induced upregulation of RAGE and VEGF mRNA levels in MCF-7 cells. In AGEs-exposed MCF-7 cells, compound C, an AMP-triggered protein, appeared to block the growth inhibition of metformin, RAGE, and VEGF effects. As reported, metformin may limit AGE-induced growth and VEGF expression in MCF-7 breast cancer cells by RAGE expression via the AMP-triggered protein kinase pathway. As a result, metformin may protect diabetic individuals against breast cancer expansion by inhibiting the AGEs–RAGE axis [328,329,330,331,332,333].

5.11. RAP (RAGE Antagonist Peptide)

In vivo and in vitro research reveals that RAP inhibitor inhibits the ligation of S100A4, S100P, and HMGB-1 with RAGE at micromolar concentrations. The efficiency of the ligands to promote RAGE activation of NF-κB in cancer cells is hampered by the presence of a RAP inhibitor. Systemic in vivo study of RAP significantly reduced the growth and spread of pancreatic and glioma tumors. The RAP has the potential to explore RAGE issues and cure RAGE-related disorders in vivo [304].

5.12. Cromolyn

Cromolyn is an antiallergen, and its study revealed that it inhibits S100P. Cromolyn is a RAGE inhibitor that reduces cancer growth, endurance, and invasiveness in vitro by inhibiting RAGE interaction with S100P. Cromolyn inhibited the NF-κB pathway in pancreatic cancer with endogenic S100P in vitro and in vivo. In mouse models, studies show that the processes clarify the pragmatic suppression of tumor development and cromolyn’s capacity to increase gemcitabine’s efficacy in eradicating pancreatic cancer cells [315].

5.13. Ethyl Pyruvate

An ethyl ester of pyruvic acid effectively inhibits HMGB1 in treating inflammatory diseases and malignancies [334,335,336]. The effectiveness of ethyl pyruvate on human malignant mesothelioma cells and tumor formation has been demonstrated in vivo investigations. Because ethyl pyruvate successfully decreased RAGE accumulation and the activation of NF-κB, research has discovered that it has a significant role in reducing HMGB1 release in malignant mesothelioma cells. Furthermore, ethyl pyruvate reduced the HMGB1 serum scale in vivo and suppressed the growth of malignant mesothelioma xenografts [336]. Moreover, an in vitro study found that ethyl pyruvate can inhibit non-small cell lung cancer growth and progression by inducing apoptosis via the HMGB1-RAGE axis and the NF-κB/STAT3 pathway (Figure 5) [337]. Consequently, ethyl pyruvate has been identified as a promising treatment for malignant mesothelioma, with the ability to reduce the progression of NSCLC.

5.14. Papaverine

A non-narcotic opium alkaloid derived from the plant Papaver somniferum. Contrary to multiple tumor cells, papaverine demonstrated discriminating anticancer effects [338,339]. An in vitro experiment was performed to test the anti-RAGE result of papaverine, improved using the structure-based pharmacological technique known as a conversion-to-small-molecules-via-optimized-peptide strategy (COSMOS), in human fibrosarcoma cell lines (HT1080). It was revealed utilizing RAGE or dnRAGE expressing (dominant-negative) HT1080 cell lines that papaverine inhibited RAGE-dependent human fibrosarcoma cell propagation, relocation, and invasion in a dose-dependent manner by significant suppression of RAGE-dependent NF-κB determined by HMGB1 (Figure 5) [339]. Furthermore, papaverine inhibited cell proliferation in human glioblastoma (U87MG and T98G) cell lines via downregulating HMGB1 and RAGE [339]. Therefore, papaverine may inhibit RAGE and can be safeguarded as an encouraging anticancer medication.

5.15. Hispidin and Ergothioneine

Hispidin is a polyphenol derived from the fungus Phellinus linteus that can be used as a major medication due to its biological applicability as an antioxidant and anticancer. Hispidin may be a novel gemcitabine chemosensitizer and a cooperative representative that can shift the gemcitabine therapeutic indication to pancreatic cancer treatment. Furthermore, hispidin induces apoptosis in colon cancer cells via a class of reactive oxygen species. Fungi and bacteria make ergothioneine, a thiol molecule. A mixture of 2 µM ergothioneine and hispidin reduces the development of AGEs, RAGE expression, and the activation of NF-κB in a PC12 cell line from rat pheochromocytomata (Figure 5) [340,341,342,343]. The AGEs–RAGE axis-associated stimulation of carcinogenesis was minimized in collaboration with ergothioneine and hispidin.

6. Conclusions

We reviewed RAGE and its ligands, as well as various inhibitors that can provide a platform for researchers and clinicians to develop a novel strategy for tackling numerous types of cancers, considering that RAGE is a cell surface receptor known to interact with a wide range of ligands. Multiple small RAGE inhibitors were explored in this review for their ability to inhibit RAGE in various malignancies. The current research shows that RAGE and its interaction with ligands increase the efficiency of antiapoptotic proteins while diminishing the effectiveness of proapoptotic proteins in promoting cancer cell proliferation, invasion, and angiogenesis. In the initial sections of this review, we described the role of RAGE ligands in cancer progression, particularly AGEs, HMGB1, S100 proteins, adhesion molecules, complement components, ALEs, and lipopolysaccharides, which play important roles in cancer biology in conjunction with RAGE. In the later sections, we focused on various anti-RAGE therapeutics, particularly mAbs, peptides, and small molecules, for managing cancer metastasis. The utilization of existing and newly discovered RAGE inhibitors for cancer prognosis might reduce the burden of cancer. Additionally, several existing RAGE inhibitors, acting as anticancer drugs, which could be beneficial in integrated treatment strategies, were also presented.

Author Contributions

T.F.; writing—original draft preparation, Z.R., I.H., E.-H.C. and M.S.; writing—review and editing, M.S.K., Y.A., S.K. and D.K.Y.: Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are thankful to the Basic Science Research Program of the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2021R1A6A1A03038785 and 2020R1I1A1A01073071).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors are thankful to the management of Integral University for providing the basic infrastructure for the research. The manuscript has manuscript id provided by Research and Development office, Integral University, Lucknow (IU/R&D/2022-MCN0001702).

Conflicts of Interest

The authors declare that there are no conflict of interest in this manuscript. On behalf of all authors.

References

  1. Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 6 September 2022).
  2. Neeper, M.; Schmidt, A.M.; Brett, J.; Yan, S.D.; Wang, F.; Pan, Y.C.; Elliston, K.; Stern, D.; Shaw, A. Cloning and Expression of a Cell Surface Receptor for Advanced Glycosylation End Products of Proteins. J. Biol. Chem. 1992, 267, 14998–15004. [Google Scholar] [CrossRef] [PubMed]
  3. Li, R.; Song, Y.; Zhou, L.; Li, W.; Zhu, X. Downregulation of RAGE Inhibits Cell Proliferation and Induces Apoptosis via Regulation of PI3K/AKT Pathway in Cervical Squamous Cell Carcinoma. OncoTargets Ther. 2020, 13, 2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Buckley, S.T.; Ehrhardt, C. The Receptor for Advanced Glycation End Products (RAGE) and the Lung. J. Biomed. Biotechnol. 2010, 2010, 917108. [Google Scholar] [CrossRef] [Green Version]
  5. Lizotte, P.-P.; Hanford, L.E.; Enghild, J.J.; Nozik-Grayck, E.; Giles, B.-L.; Oury, T.D. Developmental Expression of the Receptor for Advanced Glycation End-Products (RAGE) and Its Response to Hyperoxia in the Neonatal Rat Lung. BMC Dev. Biol. 2007, 7, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Schraml, P.; Bendik, I.; Ludwig, C.U. Differential Messenger RNA and Protein Expression of the Receptor for Advanced Glycosylated End Products in Normal Lung and Non-Small Cell Lung Carcinoma. Cancer Res. 1997, 57, 3669–3671. [Google Scholar]
  7. Bartling, B.; Demling, N.; Silber, R.-E.; Simm, A. Proliferative Stimulus of Lung Fibroblasts on Lung Cancer Cells Is Impaired by the Receptor for Advanced Glycation End-Products. Am. J. Respir. Cell Mol. Biol. 2006, 34, 83–91. [Google Scholar] [CrossRef]
  8. Bartling, B.; Hofmann, H.-S.; Weigle, B.; Silber, R.-E.; Simm, A. Down-Regulation of the Receptor for Advanced Glycation End-Products (RAGE) Supports Non-Small Cell Lung Carcinoma. Carcinogenesis 2005, 26, 293–301. [Google Scholar] [CrossRef] [Green Version]
  9. Hofmann, H.-S.; Hansen, G.; Burdach, S.; Bartling, B.; Silber, R.-E.; Simm, A. Discrimination of Human Lung Neoplasm from Normal Lung by Two Target Genes. Am. J. Respir. Crit. Care Med. 2004, 170, 516–519. [Google Scholar] [CrossRef]
  10. Hsieh, H.-L.; Schäfer, B.W.; Sasaki, N.; Heizmann, C.W. Expression Analysis of S100 Proteins and RAGE in Human Tumors Using Tissue Microarrays. Biochem. Biophys. Res. Commun. 2003, 307, 375–381. [Google Scholar] [CrossRef]
  11. Taguchi, A.; Blood, D.C.; del Toro, G.; Canet, A.; Lee, D.C.; Qu, W.; Tanji, N.; Lu, Y.; Lalla, E.; Fu, C. Blockade of RAGE–Amphoterin Signalling Suppresses Tumour Growth and Metastases. Nature 2000, 405, 354–360. [Google Scholar] [CrossRef]
  12. Kwak, T.; Drews-Elger, K.; Ergonul, A.; Miller, P.C.; Braley, A.; Hwang, G.H.; Zhao, D.; Besser, A.; Yamamoto, Y.; Yamamoto, H. Targeting of RAGE-Ligand Signaling Impairs Breast Cancer Cell Invasion and Metastasis. Oncogene 2017, 36, 1559–1572. [Google Scholar] [CrossRef] [PubMed]
  13. Logsdon, C.D.; Fuentes, M.K.; Huang, E.H.; Arumugam, T. RAGE and RAGE Ligands in Cancer. Curr. Mol. Med. 2007, 7, 777–789. [Google Scholar] [CrossRef] [PubMed]
  14. Kang, R.; Tang, D.; Schapiro, N.E.; Livesey, K.M.; Farkas, A.; Loughran, P.; Bierhaus, A.; Lotze, M.T.; Zeh, H.J. The Receptor for Advanced Glycation End Products (RAGE) Sustains Autophagy and Limits Apoptosis, Promoting Pancreatic Tumor Cell Survival. Cell Death Differ. 2010, 17, 666–676. [Google Scholar] [CrossRef] [Green Version]
  15. Chavakis, T.; Bierhaus, A.; Al-Fakhri, N.; Schneider, D.; Witte, S.; Linn, T.; Nagashima, M.; Morser, J.; Arnold, B.; Preissner, K.T. The Pattern Recognition Receptor (RAGE) Is a Counterreceptor for Leukocyte Integrins a Novel Pathway for Inflammatory Cell Recruitment. J. Exp. Med. 2003, 198, 1507–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Fehrenbach, H.; Kasper, M.; Tschernig, T.; Shearman Ms, S.D.; Müller, M. Receptor Foradvanced Glycation Endproducts (Rage) Exillbits Idghly Differential Cellular and Subcellular Localisation in Rat and Human Lung. Cell. Mol. Biol. 1998, 44, 1147–1157. [Google Scholar] [PubMed]
  17. Bierhaus, A.; Humpert, P.M.; Morcos, M.; Wendt, T.; Chavakis, T.; Arnold, B.; Stern, D.M.; Nawroth, P.P. Understanding RAGE, the Receptor for Advanced Glycation End Products. J. Mol. Med. 2005, 83, 876–886. [Google Scholar] [CrossRef]
  18. Schmidt, A.M.; Du Yan, S.; Yan, S.F.; Stern, D.M. The Multiligand Receptor RAGE as a Progression Factor Amplifying Immune and Inflammatory Responses. J. Clin. Investig. 2001, 108, 949–955. [Google Scholar] [CrossRef]
  19. Ishiguro, H.; Nakaigawa, N.; Miyoshi, Y.; Fujinami, K.; Kubota, Y.; Uemura, H. Receptor for Advanced Glycation End Products (RAGE) and Its Ligand, Amphoterin Are Overexpressed and Associated with Prostate Cancer Development. Prostate 2005, 64, 92–100. [Google Scholar] [CrossRef]
  20. Kuniyasu, H.; Chihara, Y.; Takahashi, T. Co-Expression of Receptor for Advanced Glycation End Products and the Ligand Amphoterin Associates Closely with Metastasis of Colorectal Cancer. Oncol. Rep. 2003, 10, 445–448. [Google Scholar] [CrossRef]
  21. Kuniyasu, H.; Oue, N.; Wakikawa, A.; Shigeishi, H.; Matsutani, N.; Kuraoka, K.; Ito, R.; Yokozaki, H.; Yasui, W. Expression of Receptors for Advanced Glycation End-products (RAGE) Is Closely Associated with the Invasive and Metastatic Activity of Gastric Cancer. J. Pathol. 2002, 196, 163–170. [Google Scholar] [CrossRef]
  22. Lotze, M.T.; Tracey, K.J. High-Mobility Group Box 1 Protein (HMGB1): Nuclear Weapon in the Immune Arsenal. Nat. Rev. Immunol. 2005, 5, 331–342. [Google Scholar] [CrossRef] [PubMed]
  23. Huttunen, H.J.; Fages, C.; Kuja-Panula, J.; Ridley, A.J.; Rauvala, H. Receptor for Advanced Glycation End Products-Binding COOH-Terminal Motif of Amphoterin Inhibits Invasive Migration and Metastasis. Cancer Res. 2002, 62, 4805–4811. [Google Scholar] [PubMed]
  24. Sakaguchi, T.; Yan, S.F.; Du Yan, S.; Belov, D.; Rong, L.L.; Sousa, M.; Andrassy, M.; Marso, S.P.; Duda, S.; Arnold, B. Central Role of RAGE-Dependent Neointimal Expansion in Arterial Restenosis. J. Clin. Investig. 2003, 111, 959–972. [Google Scholar] [CrossRef] [Green Version]
  25. Hofmann, M.A.; Drury, S.; Fu, C.; Qu, W.; Taguchi, A.; Lu, Y.; Avila, C.; Kambham, N.; Bierhaus, A.; Nawroth, P. RAGE Mediates a Novel Proinflammatory Axis: A Central Cell Surface Receptor for S100/Calgranulin Polypeptides. Cell 1999, 97, 889–901. [Google Scholar] [CrossRef] [Green Version]
  26. Kislinger, T.; Fu, C.; Huber, B.; Qu, W.; Taguchi, A.; Du Yan, S.; Hofmann, M.; Yan, S.F.; Pischetsrieder, M.; Stern, D. N ε-(Carboxymethyl) Lysine Adducts of Proteins Are Ligands for Receptor for Advanced Glycation End Products That Activate Cell Signaling Pathways and Modulate Gene Expression. J. Biol. Chem. 1999, 274, 31740–31749. [Google Scholar] [CrossRef] [Green Version]
  27. Lander, H.M.; Tauras, J.M.; Ogiste, J.S.; Hori, O.; Moss, R.A.; Schmidt, A.M. Activation of the Receptor for Advanced Glycation End Products Triggers a P21 Ras-Dependent Mitogen-Activated Protein Kinase Pathway Regulated by Oxidant Stress. J. Biol. Chem. 1997, 272, 17810–17814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Huang, J.; Guh, J.; Chen, H.; Hung, W.; Lai, Y.; Chuang, L. Role of Receptor for Advanced Glycation End-product (RAGE) and the JAK/STAT-signaling Pathway in AGE-induced Collagen Production in NRK-49F Cells. J. Cell. Biochem. 2001, 81, 102–113. [Google Scholar] [CrossRef]
  29. Yeh, C.-H.; Sturgis, L.; Haidacher, J.; Zhang, X.-N.; Sherwood, S.J.; Bjercke, R.J.; Juhasz, O.; Crow, M.T.; Tilton, R.G.; Denner, L. Requirement for P38 and P44/P42 Mitogen-Activated Protein Kinases in RAGE-Mediated Nuclear Factor-ΚB Transcriptional Activation and Cytokine Secretion. Diabetes 2001, 50, 1495–1504. [Google Scholar] [CrossRef] [Green Version]
  30. Arumugam, T.; Simeone, D.M.; Schmidt, A.M.; Logsdon, C.D. S100P Stimulates Cell Proliferation and Survival via Receptor for Activated Glycation End Products (RAGE). J. Biol. Chem. 2004, 279, 5059–5065. [Google Scholar] [CrossRef] [Green Version]
  31. Arumugam, T.; Simeone, D.M.; Van Golen, K.; Logsdon, C.D. S100P Promotes Pancreatic Cancer Growth, Survival, and Invasion. Clin. Cancer Res. 2005, 11, 5356–5364. [Google Scholar] [CrossRef] [Green Version]
  32. Huttunen, H.J.; Fages, C.; Rauvala, H. Receptor for Advanced Glycation End Products (RAGE)-Mediated Neurite Outgrowth and Activation of NF-ΚB Require the Cytoplasmic Domain of the Receptor but Different Downstream Signaling Pathways. J. Biol. Chem. 1999, 274, 19919–19924. [Google Scholar] [CrossRef] [PubMed]
  33. Bierhaus, A.; Schiekofer, S.; Schwaninger, M.; Andrassy, M.; Humpert, P.M.; Chen, J.; Hong, M.; Luther, T.; Henle, T.; Klöting, I. Diabetes-Associated Sustained Activation of the Transcription Factor Nuclear Factor-ΚB. Diabetes 2001, 50, 2792–2808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Moysa, A.; Hammerschmid, D.; Szczepanowski, R.H.; Sobott, F.; Dadlez, M. Enhanced Oligomerization of Full-Length RAGE by Synergy of the Interaction of Its Domains. Sci. Rep. 2019, 9, 20332. [Google Scholar] [CrossRef] [Green Version]
  35. Fritz, G. RAGE: A Single Receptor Fits Multiple Ligands. Trends Biochem. Sci. 2011, 36, 625–632. [Google Scholar] [CrossRef] [PubMed]
  36. Dattilo, B.M.; Fritz, G.; Leclerc, E.; Vander Kooi, C.W.; Heizmann, C.W.; Chazin, W.J. The Extracellular Region of the Receptor for Advanced Glycation End Products Is Composed of Two Independent Structural Units. Biochemistry 2007, 46, 6957–6970. [Google Scholar] [CrossRef] [Green Version]
  37. Xue, J.; Rai, V.; Singer, D.; Chabierski, S.; Xie, J.; Reverdatto, S.; Burz, D.S.; Schmidt, A.M.; Hoffmann, R.; Shekhtman, A. Advanced Glycation End Product Recognition by the Receptor for AGEs. Structure 2011, 19, 722–732. [Google Scholar] [CrossRef] [Green Version]
  38. Leclerc, E.; Fritz, G.; Vetter, S.W.; Heizmann, C.W. Binding of S100 Proteins to RAGE: An Update. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2009, 1793, 993–1007. [Google Scholar] [CrossRef] [Green Version]
  39. Zong, H.; Madden, A.; Ward, M.; Mooney, M.H.; Elliott, C.T.; Stitt, A.W. Homodimerization Is Essential for the Receptor for Advanced Glycation End Products (RAGE)-Mediated Signal Transduction. J. Biol. Chem. 2010, 285, 23137–23146. [Google Scholar] [CrossRef] [Green Version]
  40. Yatime, L.; Andersen, G.R. Structural Insights into the Oligomerization Mode of the Human Receptor for Advanced Glycation End-products. FEBS J. 2013, 280, 6556–6568. [Google Scholar] [CrossRef] [Green Version]
  41. Kierdorf, K.; Fritz, G. RAGE Regulation and Signaling in Inflammation and Beyond. J. Leukoc. Biol. 2013, 94, 55–68. [Google Scholar] [CrossRef]
  42. Xie, J.; Burz, D.S.; He, W.; Bronstein, I.B.; Lednev, I.; Shekhtman, A. Hexameric Calgranulin C (S100A12) Binds to the Receptor for Advanced Glycated End Products (RAGE) Using Symmetric Hydrophobic Target-Binding Patches. J. Biol. Chem. 2007, 282, 4218–4231. [Google Scholar] [CrossRef] [PubMed]
  43. Iwamura, M.; Yamamoto, Y.; Kitayama, Y.; Higuchi, K.; Fujimura, T.; Hase, T.; Yamamoto, H. Epidermal Expression of Receptor for Advanced Glycation End Products (RAGE) Is Related to Inflammation and Apoptosis in Human Skin. Exp. Dermatol. 2016, 25, 235–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Hudson, B.I.; Carter, A.M.; Harja, E.; Kalea, A.Z.; Arriero, M.; Yang, H.; Grant, P.J.; Schmidt, A.M. Identification, Classification, and Expression of RAGE Gene Splice Variants. FASEB J. 2008, 22, 1572–1580. [Google Scholar] [CrossRef] [PubMed]
  45. Foell, D.; Wittkowski, H.; Vogl, T.; Roth, J. S100 Proteins Expressed in Phagocytes: A Novel Group of Damage-associated Molecular Pattern Molecules. J. Leukoc. Biol. 2007, 81, 28–37. [Google Scholar] [CrossRef] [Green Version]
  46. Ding, Q.; Keller, J.N. Evaluation of Rage Isoforms, Ligands, and Signaling in the Brain. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2005, 1746, 18–27. [Google Scholar] [CrossRef] [Green Version]
  47. Win, M.T.T.; Yamamoto, Y.; Munesue, S.; Saito, H.; Han, D.; Motoyoshi, S.; Kamal, T.; Ohara, T.; Watanabe, T.; Yamamoto, H. Regulation of RAGE for Attenuating Progression of Diabetic Vascular Complications. Exp. Diabetes Res. 2011, 2012, 894605. [Google Scholar] [CrossRef] [Green Version]
  48. Du Yan, S.; Chen, X.; Fu, J.; Chen, M.; Zhu, H.; Roher, A.; Slattery, T.; Zhao, L.; Nagashima, M.; Morser, J. RAGE and Amyloid-β Peptide Neurotoxicity in Alzheimer’s Disease. Nature 1996, 382, 685–691. [Google Scholar] [CrossRef]
  49. Tsang, D.K.; Crowe, D.L. The Mitogen Activated Protein Kinase Pathway Is Required for Proliferation but Not Invasion of Human Squamous Cell Carcinoma Lines. Int. J. Oncol. 1999, 15, 519–542. [Google Scholar] [CrossRef]
  50. Flohr, A.M.; Rogalla, P.; Meiboom, M.; Borrmann, L.; Krohn, M.; Thode-Halle, B.; Bullerdiek, J. Variation of HMGB1 Expression in Breast Cancer. Anticancer. Res. 2001, 21, 3881–3885. [Google Scholar]
  51. Reed, J.; Jurgensmeier, J.; Matsuyama, S. Bcl-2 Family Proteins and Mitochondria. Biochim. Biophys. Acta (BBA)-Bioenerg. 1998, 1366, 127–137. [Google Scholar] [CrossRef] [Green Version]
  52. Liao, S.; Li, J.; Wei, W.; Wang, L.; Zhang, Y.; Li, J.; Wang, C.; Sun, S. Association between Diabetes Mellitus and Breast Cancer Risk: A Meta-Analysis of the Literature. Asian Pac. J. Cancer Prev. 2011, 12, 1061–1065. [Google Scholar] [PubMed]
  53. Larsson, S.C.; Mantzoros, C.S.; Wolk, A. Diabetes Mellitus and Risk of Breast Cancer: A Meta-analysis. Int. J. Cancer 2007, 121, 856–862. [Google Scholar] [CrossRef] [PubMed]
  54. Marenholz, I.; Heizmann, C.W.; Fritz, G. S100 Proteins in Mouse and Man: From Evolution to Function and Pathology (Including an Update of the Nomenclature). Biochem. Biophys. Res. Commun. 2004, 322, 1111–1122. [Google Scholar] [CrossRef] [PubMed]
  55. Donato, R. S100: A Multigenic Family of Calcium-Modulated Proteins of the EF-Hand Type with Intracellular and Extracellular Functional Roles. Int. J. Biochem. Cell Biol. 2001, 33, 637–668. [Google Scholar] [CrossRef] [PubMed]
  56. Shinohara, M.; Thornalley, P.J.; Giardino, I.; Beisswenger, P.; Thorpe, S.R.; Onorato, J.; Brownlee, M. Overexpression of Glyoxalase-I in Bovine Endothelial Cells Inhibits Intracellular Advanced Glycation Endproduct Formation and Prevents Hyperglycemia-Induced Increases in Macromolecular Endocytosis. J. Clin. Investig. 1998, 101, 1142–1147. [Google Scholar] [CrossRef] [PubMed]
  57. Reddy, S.; Bichler, J.; Wells-Knecht, K.J.; Thorpe, S.R.; Baynes, J.W.N. Epsilon.-(Carboxymethyl) Lysine Is a Dominant Advanced Glycation End Product (AGE) Antigen in Tissue Proteins. Biochemistry 1995, 34, 10872–10878. [Google Scholar] [CrossRef]
  58. Thomas, J.O.; Travers, A.A. HMG1 and 2, and Related ‘Architectural’DNA-Binding Proteins. Trends Biochem. Sci. 2001, 26, 167–174. [Google Scholar] [CrossRef]
  59. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef]
  60. Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of Chromatin Protein HMGB1 by Necrotic Cells Triggers Inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef]
  61. Klune, J.R.; Dhupar, R.; Cardinal, J.; Billiar, T.R.; Tsung, A. HMGB1: Endogenous Danger Signaling. Mol. Med. 2008, 14, 476–484. [Google Scholar] [CrossRef]
  62. Ulloa, L.; Messmer, D. High-Mobility Group Box 1 (HMGB1) Protein: Friend and Foe. Cytokine Growth Factor Rev. 2006, 17, 189–201. [Google Scholar] [CrossRef] [PubMed]
  63. Palumbo, R.; Galvez, B.G.; Pusterla, T.; De Marchis, F.; Cossu, G.; Marcu, K.B.; Bianchi, M.E. Cells Migrating to Sites of Tissue Damage in Response to the Danger Signal HMGB1 Require NF-ΚB Activation. J. Cell Biol. 2007, 179, 33–40. [Google Scholar] [CrossRef] [PubMed]
  64. Sahin, U.; Türeci, Ö. Personalized Vaccines for Cancer Immunotherapy. Science 2018, 359, 1355–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T Cell Immunotherapy for Human Cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Restifo, N.P.; Smyth, M.J.; Snyder, A. Acquired Resistance to Immunotherapy and Future Challenges. Nat. Rev. Cancer 2016, 16, 121–126. [Google Scholar] [CrossRef] [PubMed]
  67. Zhao, X.; Subramanian, S. Intrinsic Resistance of Solid Tumors to Immune Checkpoint Blockade Therapy. Cancer Res. 2017, 77, 817–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Pardoll, D.M. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  69. Ahmad, S.; Khan, H.; Siddiqui, Z.; Khan, M.Y.; Rehman, S.; Shahab, U.; Godovikova, T.; Silnikov, V. AGEs, RAGEs and s-RAGE; Friend or Foe for Cancer. Semin. Cancer Biol. 2018, 49, 44–55. [Google Scholar] [CrossRef]
  70. Gangemi, S.; Allegra, A.; Aguennouz, M.; Alonci, A.; Speciale, A.; Cannavò, A.; Cristani, M.; Russo, S.; Spatari, G.; Alibrandi, A. Relationship between Advanced Oxidation Protein Products, Advanced Glycation End Products, and S-Nitrosylated Proteins with Biological Risk and MDR-1 Polymorphisms in Patients Affected by B-Chronic Lymphocytic Leukemia. Cancer Investig. 2012, 30, 20–26. [Google Scholar] [CrossRef]
  71. Chhipa, A.S.; Borse, S.P.; Baksi, R.; Lalotra, S.; Nivsarkar, M. Targeting Receptors of Advanced Glycation End Products (RAGE): Preventing Diabetes Induced Cancer and Diabetic Complications. Pathol.-Res. Pract. 2019, 215, 152643. [Google Scholar] [CrossRef]
  72. Abe, R.; Yamagishi, S. AGE-RAGE System and Carcinogenesis. Curr. Pharm. Des. 2008, 14, 940–945. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, J.Y.; Park, H.K.; Yoon, J.S.; Kim, S.J.; Kim, E.S.; Ahn, K.S.; Kim, D.-S.; Yoon, S.S.; Kim, B.K.; Lee, Y.Y. Advanced Glycation End Product (AGE)-Induced Proliferation of HEL Cells via Receptor for AGE-Related Signal Pathways. Int. J. Oncol. 2008, 33, 493–501. [Google Scholar] [CrossRef] [PubMed]
  74. Riehl, A.; Németh, J.; Angel, P.; Hess, J. The Receptor RAGE: Bridging Inflammation and Cancer. Cell Commun. Signal. 2009, 7, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ko, S.-Y.; Ko, H.-A.; Shieh, T.-M.; Chang, W.-C.; Chen, H.-I.; Chang, S.-S.; Lin, I.-H. Cell Migration Is Regulated by AGE-RAGE Interaction in Human Oral Cancer Cells in Vitro. PLoS ONE 2014, 9, e110542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ahmad, S.; Khan, M.Y.; Rafi, Z.; Khan, H.; Siddiqui, Z.; Rehman, S.; Shahab, U.; Khan, M.S.; Saeed, M.; Alouffi, S. Oxidation, Glycation and Glycoxidation—The Vicious Cycle and Lung Cancer. Semin. Cancer Biol. 2018, 49, 29–36. [Google Scholar] [CrossRef]
  77. Park, J.S.; Arcaroli, J.; Yum, H.-K.; Yang, H.; Wang, H.; Yang, K.-Y.; Choe, K.-H.; Strassheim, D.; Pitts, T.M.; Tracey, K.J. Activation of Gene Expression in Human Neutrophils by High Mobility Group Box 1 Protein. Am. J. Physiol.-Cell Physiol. 2003, 284, C870–C879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Rowell, J.P.; Simpson, K.L.; Stott, K.; Watson, M.; Thomas, J.O. HMGB1-Facilitated P53 DNA Binding Occurs via HMG-Box/P53 Transactivation Domain Interaction, Regulated by the Acidic Tail. Structure 2012, 20, 2014–2024. [Google Scholar] [CrossRef] [Green Version]
  79. Smolarczyk, R.; Cichoń, T.; Jarosz, M.; Szala, S. HMGB1-Rola w Progresji i Terapii Przeciwnowotworowej. Adv. Hyg. Exp. Med. /Postep. Hig. Med. Dosw. 2012, 66, 913–920. [Google Scholar] [CrossRef]
  80. Zhang, Q.-B.; Jia, Q.; Wang, H.; Hu, C.-X.; Sun, D.; Jiang, R.-D.; Zhang, Z.-L. High-Mobility Group Protein Box1 Expression Correlates with Peritumoral Macrophage Infiltration and Unfavorable Prognosis in Patients with Hepatocellular Carcinoma and Cirrhosis. BMC Cancer 2016, 16, 880. [Google Scholar] [CrossRef] [Green Version]
  81. Choi, Y.R.; Kim, H.; Kang, H.J.; Kim, N.-G.; Kim, J.J.; Park, K.-S.; Paik, Y.-K.; Kim, H.O.; Kim, H. Overexpression of High Mobility Group Box 1 in Gastrointestinal Stromal Tumors with KIT Mutation. Cancer Res. 2003, 63, 2188–2193. [Google Scholar]
  82. Völp, K.; Brezniceanu, M.-L.; Bösser, S.; Brabletz, T.; Kirchner, T.; Göttel, D.; Joos, S.; Zörnig, M. Increased Expression of High Mobility Group Box 1 (HMGB1) Is Associated with an Elevated Level of the Antiapoptotic c-IAP2 Protein in Human Colon Carcinomas. Gut 2006, 55, 234–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yan, W.; Chang, Y.; Liang, X.; Cardinal, J.S.; Huang, H.; Thorne, S.H.; Monga, S.P.S.; Geller, D.A.; Lotze, M.T.; Tsung, A. High-mobility Group Box 1 Activates Caspase-1 and Promotes Hepatocellular Carcinoma Invasiveness and Metastases. Hepatology 2012, 55, 1863–1875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lin, L.; Zhong, K.; Sun, Z.; Wu, G.; Ding, G. Receptor for Advanced Glycation End Products (RAGE) Partially Mediates HMGB1-ERKs Activation in Clear Cell Renal Cell Carcinoma. J. Cancer Res. Clin. Oncol. 2012, 138, 11–22. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, Q.-Y.; Wu, L.-Q.; Zhang, T.; Han, Y.-F.; Lin, X. Autophagy-Mediated HMGB1 Release Promotes Gastric Cancer Cell Survival via RAGE Activation of Extracellular Signal-Regulated Kinases 1/2. Oncol. Rep. 2015, 33, 1630–1638. [Google Scholar] [CrossRef] [Green Version]
  86. Pusterla, T.; Nèmeth, J.; Stein, I.; Wiechert, L.; Knigin, D.; Marhenke, S.; Longerich, T.; Kumar, V.; Arnold, B.; Vogel, A. Receptor for Advanced Glycation Endproducts (RAGE) Is a Key Regulator of Oval Cell Activation and Inflammation-associated Liver Carcinogenesis in Mice. Hepatology 2013, 58, 363–373. [Google Scholar] [CrossRef]
  87. Sharma, S.; Evans, A.; Hemers, E. Mesenchymal-Epithelial Signalling in Tumour Microenvironment: Role of High-Mobility Group Box 1. Cell Tissue Res. 2016, 365, 357–366. [Google Scholar] [CrossRef] [Green Version]
  88. Van Beijnum, J.R.; Nowak-Sliwinska, P.; Van den Boezem, E.; Hautvast, P.; Buurman, W.A.; Griffioen, A.W. Tumor Angiogenesis Is Enforced by Autocrine Regulation of High-Mobility Group Box 1. Oncogene 2013, 32, 363–374. [Google Scholar] [CrossRef] [Green Version]
  89. Tang, D.; Kang, R.; Livesey, K.M.; Cheh, C.-W.; Farkas, A.; Loughran, P.; Hoppe, G.; Bianchi, M.E.; Tracey, K.J.; Zeh III, H.J. Endogenous HMGB1 Regulates Autophagy. J. Cell Biol. 2010, 190, 881–892. [Google Scholar] [CrossRef]
  90. Salmivirta, M.; Rauvala, H.; Elenius, K.; Jalkanen, M. Neurite Growth-Promoting Protein (Amphoterin, P30) Binds Syndecan. Exp. Cell Res. 1992, 200, 444–451. [Google Scholar] [CrossRef]
  91. Park, J.S.; Svetkauskaite, D.; He, Q.; Kim, J.-Y.; Strassheim, D.; Ishizaka, A.; Abraham, E. Involvement of Toll-like Receptors 2 and 4 in Cellular Activation by High Mobility Group Box 1 Protein. J. Biol. Chem. 2004, 279, 7370–7377. [Google Scholar] [CrossRef] [Green Version]
  92. Hori, O.; Brett, J.; Slattery, T.; Cao, R.; Zhang, J.; Chen, J.X.; Nagashima, M.; Lundh, E.R.; Vijay, S.; Nitecki, D. The Receptor for Advanced Glycation End Products (RAGE) Is a Cellular Binding Site for Amphoterin: Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system (∗). J. Biol. Chem. 1995, 270, 25752–25761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Youn, J.H.; Oh, Y.J.; Kim, E.S.; Choi, J.E.; Shin, J.-S. High Mobility Group Box 1 Protein Binding to Lipopolysaccharide Facilitates Transfer of Lipopolysaccharide to CD14 and Enhances Lipopolysaccharide-Mediated TNF-α Production in Human Monocytes. J. Immunol. 2008, 180, 5067–5074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Štros, M.; Muselíková-Polanská, E.; Pospíšilová, Š.; Strauss, F. High-Affinity Binding of Tumor-Suppressor Protein P53 and HMGB1 to Hemicatenated DNA Loops. Biochemistry 2004, 43, 7215–7225. [Google Scholar] [CrossRef] [PubMed]
  95. Sheflin, L.G.; Fucile, N.W.; Spaulding, S.W. The Specific Interactions of HMG 1 and 2 with Negatively Supercoiled DNA Are Modulated by Their Acidic C-Terminal Domains and Involve Cysteine Residues in Their HMG 1/2 Boxes. Biochemistry 1993, 32, 3238–3248. [Google Scholar] [CrossRef]
  96. Catez, F.; Yang, H.; Tracey, K.J.; Reeves, R.; Misteli, T.; Bustin, M. Network of Dynamic Interactions between Histone H1 and High-Mobility-Group Proteins in Chromatin. Mol. Cell. Biol. 2004, 24, 4321–4328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Liang, S.; Clarke, M.F. Regulation of P53 Localization. Eur. J. Biochem. 2001, 268, 2779–2783. [Google Scholar] [CrossRef] [PubMed]
  98. McKinney, K.; Prives, C. Efficient Specific DNA Binding by P53 Requires Both Its Central and C-Terminal Domains as Revealed by Studies with High-Mobility Group 1 Protein. Mol. Cell. Biol. 2002, 22, 6797–6808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Wu, X.; Mi, Y.; Yang, H.; Hu, A.; Zhang, Q.; Shang, C. The Activation of HMGB1 as a Progression Factor on Inflammation Response in Normal Human Bronchial Epithelial Cells through RAGE/JNK/NF-ΚB Pathway. Mol. Cell. Biochem. 2013, 380, 249–257. [Google Scholar] [CrossRef]
  100. Chen, R.-C.; Yi, P.-P.; Zhou, R.-R.; Xiao, M.-F.; Huang, Z.-B.; Tang, D.-L.; Huang, Y.; Fan, X.-G. The Role of HMGB1-RAGE Axis in Migration and Invasion of Hepatocellular Carcinoma Cell Lines. Mol. Cell. Biochem. 2014, 390, 271–280. [Google Scholar] [CrossRef] [Green Version]
  101. Dhumale, S.S.; Waghela, B.N.; Pathak, C. Quercetin Protects Necrotic Insult and Promotes Apoptosis by Attenuating the Expression of RAGE and Its Ligand HMGB1 in Human Breast Adenocarcinoma Cells. IUBMB Life 2015, 67, 361–373. [Google Scholar] [CrossRef]
  102. Huang, Z.; Zhong, Z.; Zhang, L.; Wang, X.; Xu, R.; Zhu, L.; Wang, Z.; Hu, S.; Zhao, X. Down-Regulation of HMGB1 Expression by ShRNA Constructs Inhibits the Bioactivity of Urothelial Carcinoma Cell Lines via the NF-ΚB Pathway. Sci. Rep. 2015, 5, 12807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zhu, L.; Li, X.; Chen, Y.; Fang, J.; Ge, Z. High-Mobility Group Box 1: A Novel Inducer of the Epithelial–Mesenchymal Transition in Colorectal Carcinoma. Cancer Lett. 2015, 357, 527–534. [Google Scholar] [CrossRef] [PubMed]
  104. Pang, X.; Zhang, Y.; Zhang, S. High-Mobility Group Box 1 Is Overexpressed in Cervical Carcinoma and Promotes Cell Invasion and Migration in Vitro. Oncol. Rep. 2017, 37, 831–840. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, M.; Geng, Y.; Deng, Q.; Li, R.; Shao, X.; Zhang, Z.; Xu, W.; Wu, Y.; Ma, Q. Translationally Controlled Tumor Protein Affects Colorectal Cancer Metastasis through the High Mobility Group Box 1-Dependent Pathway. Int. J. Oncol. 2018, 53, 1481–1492. [Google Scholar] [CrossRef]
  106. Kuniyasu, H.; Chihara, Y.; Kondo, H. Differential Effects between Amphoterin and Advanced Glycation End Products on Colon Cancer Cells. Int. J. Cancer 2003, 104, 722–727. [Google Scholar] [CrossRef]
  107. Sajithlal, G.; Huttunen, H.; Rauvala, H.; Münch, G. Receptor for Advanced Glycation End Products Plays a More Important Role in Cellular Survival than in Neurite Outgrowth during Retinoic Acid-Induced Differentiation of Neuroblastoma Cells. J. Biol. Chem. 2002, 277, 6888–6897. [Google Scholar] [CrossRef] [Green Version]
  108. Tang, C.-H.; Keng, Y.-T.; Liu, J.-F. HMGB-1 Induces Cell Motility and A5β1 Integrin Expression in Human Chondrosarcoma Cells. Cancer Lett. 2012, 322, 98–106. [Google Scholar] [CrossRef]
  109. Peng, T.; Hu, M.; Wu, T.; Chen, Z.; Zhang, C.; Huang, S.; Zhou, X. Effects of High-mobility Group Box 1 Knockdown on Proliferation, Migration and Invasion of the HONE-1 Human Nasopharyngeal Carcinoma Cell Line. Mol. Med. Rep. 2015, 12, 7531–7537. [Google Scholar] [CrossRef] [Green Version]
  110. Arumugam, T.; Ramachandran, V.; Gomez, S.B.; Schmidt, A.M.; Logsdon, C.D. S100P-Derived RAGE Antagonistic Peptide Reduces Tumor Growth and Metastasis. Clin. Cancer Res. 2012, 18, 4356–4364. [Google Scholar] [CrossRef] [Green Version]
  111. Yaser, A.-M.; Huang, Y.; Zhou, R.-R.; Hu, G.-S.; Xiao, M.-F.; Huang, Z.-B.; Duan, C.-J.; Tian, W.; Tang, D.-L.; Fan, X.-G. The Role of Receptor for Advanced Glycation End Products (RAGE) in the Proliferation of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2012, 13, 5982–5997. [Google Scholar] [CrossRef] [Green Version]
  112. Gong, W.; Wang, Z.-Y.; Chen, G.-X.; Liu, Y.-Q.; Gu, X.-Y.; Liu, W.-W. Invasion Potential of H22 Hepatocarcinoma Cells Is Increased by HMGB1-Induced Tumor NF-ΚB Signaling via Initiation of HSP70. Oncol. Rep. 2013, 30, 1249–1256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Takeuchi, A.; Yamamoto, Y.; Munesue, S.; Harashima, A.; Watanabe, T.; Yonekura, H.; Yamamoto, H.; Tsuchiya, H. Low Molecular Weight Heparin Suppresses Receptor for Advanced Glycation End Products-mediated Expression of Malignant Phenotype in Human Fibrosarcoma Cells. Cancer Sci. 2013, 104, 740–749. [Google Scholar] [CrossRef] [Green Version]
  114. Berridge, M.J.; Lipp, P.; Bootman, M.D. The Versatility and Universality of Calcium Signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef] [PubMed]
  115. Kretsinger, R.H. Why Cells Must Export Calcium; Wiley-Liss: New York, NY, USA, 1990; pp. 439–457. [Google Scholar]
  116. Kretsinger, R.H.; Wasserman, R.H. Structure and Evolution of Calcium-Modulated Protein. Crit. Rev. Biochem. 1980, 8, 119–174. [Google Scholar] [CrossRef] [PubMed]
  117. Kretsinger, R.H.; Tolbert, D.; Nakayama, S.; Pearson, W. The EF-Hand, Homologs and Analogs. In Novel Calcium-Binding Proteins; Springer: Berlin/Heidelberg, Germany, 1991; pp. 17–37. [Google Scholar]
  118. Heizmann, C.W.; Fritz, G.; Schafer, B.W. S100 Proteins: Structure, Functions and Pathology. Front. Biosci. 2002, 7, 1356–1368. [Google Scholar]
  119. Donato, R. Intracellular and Extracellular Roles of S100 Proteins. Microsc. Res. Tech. 2003, 60, 540–551. [Google Scholar] [CrossRef]
  120. Ostendorp, T.; Leclerc, E.; Galichet, A.; Koch, M.; Demling, N.; Weigle, B.; Heizmann, C.W.; Kroneck, P.M.H.; Fritz, G. Structural and Functional Insights into RAGE Activation by Multimeric S100B. EMBO J. 2007, 26, 3868–3878. [Google Scholar] [CrossRef]
  121. Chaudhuri, D.; Horrocks, W.D.; Amburgey, J.C.; Weber, D.J. Characterization of Lanthanide Ion Binding to the EF-Hand Protein S100β by Luminescence Spectroscopy. Biochemistry 1997, 36, 9674–9680. [Google Scholar] [CrossRef]
  122. Mishra, S.K.; Siddique, H.R.; Saleem, M. S100A4 Calcium-Binding Protein Is Key Player in Tumor Progression and Metastasis: Preclinical and Clinical Evidence. Cancer Metastasis Rev. 2012, 31, 163–172. [Google Scholar] [CrossRef]
  123. Boye, K.; Mælandsmo, G.M. S100A4 and Metastasis: A Small Actor Playing Many Roles. Am. J. Pathol. 2010, 176, 528–535. [Google Scholar] [CrossRef]
  124. Yammani, R.R.; Carlson, C.S.; Bresnick, A.R.; Loeser, R.F. 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, 2901–2911. [Google Scholar] [CrossRef] [PubMed]
  125. Garrett, S.C.; Varney, K.M.; Weber, D.J.; Bresnick, A.R. S100A4, a Mediator of Metastasis. J. Biol. Chem. 2006, 281, 677–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Dahlmann, M.; Okhrimenko, A.; Marcinkowski, P.; Osterland, M.; Herrmann, P.; Smith, J.; Heizmann, C.W.; Schlag, P.M.; Stein, U. RAGE Mediates S100A4-Induced Cell Motility via MAPK/ERK and Hypoxia Signaling and Is a Prognostic Biomarker for Human Colorectal Cancer Metastasis. Oncotarget 2014, 5, 3220. [Google Scholar] [CrossRef] [Green Version]
  127. Grotterød, I.; Mælandsmo, G.M.; Boye, K. Signal Transduction Mechanisms Involved in S100A4-Induced Activation of the Transcription Factor NF-ΚB. BMC Cancer 2010, 10, 241. [Google Scholar] [CrossRef] [PubMed]
  128. Haase-Kohn, C.; Wolf, S.; Herwig, N.; Mosch, B.; Pietzsch, J. Metastatic Potential of B16-F10 Melanoma Cells Is Enhanced by Extracellular S100A4 Derived from RAW264. 7 Macrophages. Biochem. Biophys. Res. Commun. 2014, 446, 143–148. [Google Scholar] [CrossRef]
  129. Haase-Kohn, C.; Wolf, S.; Lenk, J.; Pietzsch, J. Copper-Mediated Cross-Linking of S100A4, but Not of S100A2, Results in Proinflammatory Effects in Melanoma Cells. Biochem. Biophys. Res. Commun. 2011, 413, 494–498. [Google Scholar] [CrossRef]
  130. Herwig, N.; Belter, B.; Pietzsch, J. Extracellular S100A4 Affects Endothelial Cell Integrity and Stimulates Transmigration of A375 Melanoma Cells. Biochem. Biophys. Res. Commun. 2016, 477, 963–969. [Google Scholar] [CrossRef]
  131. Nikitenko, L.L.; Lloyd, B.H.; Rudland, P.S.; Fear, S.; Barraclough, R. Localisation by in Situ Hybridisation of S100A4 (P9Ka) MRNA in Primary Human Breast Tumour Specimens. Int. J. Cancer 2000, 86, 219–228. [Google Scholar] [CrossRef]
  132. 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, 1086–1093. [Google Scholar] [CrossRef] [Green Version]
  133. Boye, K.; Grotterød, I.; Aasheim, H.; Hovig, E.; Mælandsmo, G.M. Activation of NF-κB by Extracellular S100A4: Analysis of Signal Transduction Mechanisms and Identification of Target Genes. Int. J. Cancer 2008, 123, 1301–1310. [Google Scholar] [CrossRef]
  134. Schmidt-Hansen, B.; Örnås, D.; Grigorian, M.; Klingelhöfer, 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, 5487–5495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Novitskaya, V.; Grigorian, M.; Kriajevska, M.; Tarabykina, S.; Bronstein, I.; Berezin, V.; Bock, E.; Lukanidin, E. Oligomeric Forms of the Metastasis-Related Mts1 (S100A4) Protein Stimulate Neuronal Differentiation in Cultures of Rat Hippocampal Neurons. J. Biol. Chem. 2000, 275, 41278–41286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Semov, A.; Moreno, M.J.; 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, 20833–20841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Kiryushko, D.; Novitskaya, V.; Soroka, V.; Klingelhofer, J.; Lukanidin, E.; Berezin, V.; Bock, E. Molecular Mechanisms of Ca2+ Signaling in Neurons Induced by the S100A4 Protein. Mol. Cell. Biol. 2006, 26, 3625–3638. [Google Scholar] [CrossRef]
  138. Baudier, J.; Glasser, N.; Gerard, D. Ions Binding to S100 Proteins. I. Calcium-and Zinc-Binding Properties of Bovine Brain S100 Alpha Alpha, S100a (Alpha Beta), and S100b (Beta Beta) Protein: Zn2+ Regulates Ca2+ Binding on S100b Protein. J. Biol. Chem. 1986, 261, 8192–8203. [Google Scholar] [CrossRef]
  139. Kligman, D.; Hilt, D.C. The S100 Protein Family. Trends Biochem. Sci. 1988, 13, 437–443. [Google Scholar] [CrossRef]
  140. Vallely, K.M.; Rustandi, R.R.; Ellis, K.C.; Varlamova, O.; Bresnick, A.R.; Weber, D.J. Solution Structure of Human Mts1 (S100A4) as Determined by NMR Spectroscopy. Biochemistry 2002, 41, 12670–12680. [Google Scholar] [CrossRef]
  141. Malashkevich, V.N.; Varney, K.M.; Garrett, S.C.; Wilder, P.T.; Knight, D.; Charpentier, T.H.; Ramagopal, U.A.; Almo, S.C.; Weber, D.J.; Bresnick, A.R. Structure of Ca2+-Bound S100A4 and Its Interaction with Peptides Derived from Nonmuscle Myosin-IIA. Biochemistry 2008, 47, 5111–5126. [Google Scholar] [CrossRef] [Green Version]
  142. Gingras, A.R.; Basran, J.; Prescott, A.; Kriajevska, M.; Bagshaw, C.R.; Barsukov, I.L. Crystal Structure of the Ca2+-Form and Ca2+-Binding Kinetics of Metastasis-Associated Protein, S100A4. FEBS Lett. 2008, 582, 1651–1656. [Google Scholar] [CrossRef] [Green Version]
  143. Leclerc, E.; Fritz, G.; Weibel, M.; Heizmann, C.W.; Galichet, A. S100B and S100A6 Differentially Modulate Cell Survival by Interacting with Distinct RAGE (Receptor for Advanced Glycation End Products) Immunoglobulin Domains. J. Biol. Chem. 2007, 282, 31317–31331. [Google Scholar] [CrossRef] [Green Version]
  144. Leśniak, W.; Słomnicki, Ł.P.; Filipek, A. S100A6–New Facts and Features. Biochem. Biophys. Res. Commun. 2009, 390, 1087–1092. [Google Scholar] [CrossRef]
  145. Yan, S.F.; Ramasamy, R.; Schmidt, A.M. Receptor for AGE (RAGE) and Its Ligands—Cast into Leading Roles in Diabetes and the Inflammatory Response. J. Mol. Med. 2009, 87, 235–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Moroz, O.V.; Wilson, K.S.; Bronstein, I.B. The Role of Zinc in the S100 Proteins: Insights from the X-Ray Structures. Amino Acids 2011, 41, 761–772. [Google Scholar] [CrossRef] [PubMed]
  147. Filipek, A.; Leśniak, W. Current View on Cellular Function of S100A6 and Its Ligands, CacyBP/SIP and Sgt1. Postep. Biochem. 2018, 64, 242–252. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, N.; Ma, Q.; Wang, Y.; Ma, G.; Zhai, H. CacyBP/SIP Expression Is Involved in the Clinical Progression of Breast Cancer. World J. Surg. 2010, 34, 2545–2552. [Google Scholar] [CrossRef]
  149. Topolska-Woś, A.M.; Chazin, W.J.; Filipek, A. CacyBP/SIP—Structure and Variety of Functions. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2016, 1860, 79–85. [Google Scholar] [CrossRef]
  150. Leong, S.; Christopherson, R.; Baxter, R. Profiling of Apoptotic Changes in Human Breast Cancer Cells Using SELDI-TOF Mass Spectrometry. Cell. Physiol. Biochem. 2007, 20, 579–590. [Google Scholar] [CrossRef]
  151. Mohan, S.K.; Gupta, A.A.; Yu, C. Interaction of the S100A6 Mutant (C3S) with the V Domain of the Receptor for Advanced Glycation End Products (RAGE). Biochem. Biophys. Res. Commun. 2013, 434, 328–333. [Google Scholar] [CrossRef]
  152. Li, F.; Men, X.; Zhang, W. S100 Protein in Breast Tumor. Indian J. Cancer 2014, 51, 67. [Google Scholar]
  153. Cross, S.S.; Hamdy, F.C.; Deloulme, J.-C.; Rehman, I. Expression of S100 Proteins in Normal Human Tissues and Common Cancers Using Tissue Microarrays: S100A6, S100A8, S100A9 and S100A11 Are All Overexpressed in Common Cancers. Histopathology 2005, 46, 256–269. [Google Scholar] [CrossRef]
  154. Ito, Y.; Yoshida, H.; Tomoda, C.; Uruno, T.; Miya, A.; Kobayashi, K.; Matsuzuka, F.; Kakudo, K.; Kuma, K.; Miyauchi, A. Expression of S100A2 and S100A6 in Thyroid Carcinomas. Histopathology 2005, 46, 569–575. [Google Scholar] [CrossRef] [PubMed]
  155. Zhao, M.; Wang, K.; Tan, Z.; Zheng, C.; Liang, Z.; Zhao, J. Identification of Potential Therapeutic Targets for Papillary Thyroid Carcinoma by Bioinformatics Analysis. Oncol. Lett. 2016, 11, 51–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Chen, X.; Liu, X.; Lang, H.; Zhang, S.; Luo, Y.; Zhang, J. S100 Calcium-Binding Protein A6 Promotes Epithelial-Mesenchymal Transition through β-Catenin in Pancreatic Cancer Cell Line. PLoS ONE 2015, 10, e0121319. [Google Scholar] [CrossRef] [PubMed]
  157. Lyu, X.-J.; Li, H.-Z.; Ma, X.; Li, X.-T.; Gao, Y.; Ni, D.; Shen, D.-L.; Gu, L.-Y.; Wang, B.-J.; Zhang, Y. Elevated S100A6 (Calcyclin) Enhances Tumorigenesis and Suppresses CXCL14-Induced Apoptosis in Clear Cell Renal Cell Carcinoma. Oncotarget 2015, 6, 6656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Tamai, H.; Miyake, K.; Yamaguchi, H.; Shimada, T.; Dan, K.; Inokuchi, K. Inhibition of S100A6 Induces GVL Effects in MLL/AF4-Positive ALL in Human PBMC-SCID Mice. Bone Marrow Transplant. 2014, 49, 699–703. [Google Scholar] [CrossRef] [PubMed]
  159. Wolf, R.; Howard, O.M.Z.; Dong, H.-F.; Voscopoulos, C.; Boeshans, K.; Winston, J.; Divi, R.; Gunsior, M.; Goldsmith, P.; Ahvazi, B. Chemotactic Activity of S100A7 (Psoriasin) Is Mediated by the Receptor for Advanced Glycation End Products and Potentiates Inflammation with Highly Homologous but Functionally Distinct S100A15. J. Immunol. 2008, 181, 1499–1506. [Google Scholar] [CrossRef] [Green Version]
  160. Zhang, H.; Zhao, Q.; Chen, Y.; Wang, Y.; Gao, S.; Mao, Y.; Li, M.; Peng, A.; He, D.; Xiao, X. Selective Expression of S100A7 in Lung Squamous Cell Carcinomas and Large Cell Carcinomas but Not in Adenocarcinomas and Small Cell Carcinomas. Thorax 2008, 63, 352–359. [Google Scholar] [CrossRef] [Green Version]
  161. Guilloteau, K.; Paris, I.; Pedretti, N.; Boniface, K.; Juchaux, F.; Huguier, V.; Guillet, G.; Bernard, F.-X.; Lecron, J.-C.; Morel, F. Skin Inflammation Induced by the Synergistic Action of IL-17A, IL-22, Oncostatin M, IL-1α, and TNF-α Recapitulates Some Features of Psoriasis. J. Immunol. 2010, 184, 5263–5270. [Google Scholar] [CrossRef] [Green Version]
  162. Brodersen, D.E.; Etzerodt, M.; Madsen, P.; Celis, J.E.; Thøgersen, H.C.; Nyborg, J.; Kjeldgaard, M. EF-Hands at Atomic Resolution: The Structure of Human Psoriasin (S100A7) Solved by MAD Phasing. Structure 1998, 6, 477–489. [Google Scholar] [CrossRef] [Green Version]
  163. Celis, J.E.; Rasmussen, H.H.; Vorum, H.; Madsen, P.; Honore, B.; Wolf, H.; Orntoft, T.F. Bladder Squamous Cell Carcinomas Express Psoriasin and Externalize It to the Urine. J. Urol. 1996, 155, 2105–2112. [Google Scholar] [CrossRef]
  164. Ralhan, R.; DeSouza, L.V.; Matta, A.; Tripathi, S.C.; Ghanny, S.; Gupta, S.D.; Bahadur, S.; Siu, K.W.M. Discovery and Verification of Head-and-Neck Cancer Biomarkers by Differential Protein Expression Analysis Using ITRAQ Labeling, Multidimensional Liquid Chromatography, and Tandem Mass Spectrometry. Mol. Cell. Proteom. 2008, 7, 1162–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Webb, M.; Emberley, E.D.; Lizardo, M.; Alowami, S.; Qing, G.; Alfia’ar, A.; Snell-Curtis, L.J.; Niu, Y.; Civetta, A.; Myal, Y. Expression Analysis of the Mouse S100A7/Psoriasin Gene in Skin Inflammation and Mammary Tumorigenesis. BMC Cancer 2005, 5, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Brouard, M.C.; Saurat, J.-H.; Ghanem, G.; Siegenthaler, G. Urinary Excretion of Epidermal-Type Fatty Acid-Binding Protein and S100A7 Protein in Patients with Cutaneous Melanoma. Melanoma Res. 2002, 12, 627–631. [Google Scholar] [CrossRef] [PubMed]
  167. Moog-Lutz, C.; Bouillet, P.; Régnier, C.H.; Tomasetto, C.; Mattei, M.; Chenard, M.; Anglard, P.; Rio, M.; Basset, P. Comparative Expression of the Psoriasin (S100A7) and S100C Genes in Breast Carcinoma and Co-localization to Human Chromosome 1q21-q22. Int. J. Cancer 1995, 63, 297–303. [Google Scholar] [CrossRef] [PubMed]
  168. Enerbäck, C.; Porter, D.A.; Seth, P.; Sgroi, D.; Gaudet, J.; Weremowicz, S.; Morton, C.C.; Schnitt, S.; Pitts, R.L.; Stampl, J. Psoriasin Expression in Mammary Epithelial Cells in Vitro and in Vivo. Cancer Res. 2002, 62, 43–47. [Google Scholar]
  169. León, R.; Murray, J.I.; Cragg, G.; Farnell, B.; West, N.R.; Pace, T.C.S.; Watson, P.H.; Bohne, C.; Boulanger, M.J.; Hof, F. Identification and Characterization of Binding Sites on S100A7, a Participant in Cancer and Inflammation Pathways. Biochemistry 2009, 48, 10591–10600. [Google Scholar] [CrossRef]
  170. Tian, T.; Li, X.; Hua, Z.; Ma, J.; Wu, X.; Liu, Z.; Chen, H.; Cui, Z. S100A7 Promotes the Migration, Invasion and Metastasis of Human Cervical Cancer Cells through Epithelial–Mesenchymal Transition. Oncotarget 2017, 8, 24964. [Google Scholar] [CrossRef] [Green Version]
  171. Yin, C.; Li, H.; Zhang, B.; Liu, Y.; Lu, G.; Lu, S.; Sun, L.; Qi, Y.; Li, X.; Chen, W. RAGE-Binding S100A8/A9 Promotes the Migration and Invasion of Human Breast Cancer Cells through Actin Polymerization and Epithelial–Mesenchymal Transition. Breast Cancer Res. Treat. 2013, 142, 297–309. [Google Scholar] [CrossRef]
  172. Vetter, S.W.; Leclerc, E. Novel Aspects of Calmodulin Target Recognition and Activation. Eur. J. Biochem. 2003, 270, 404–414. [Google Scholar] [CrossRef]
  173. Iotzova-Weiss, G.; Dziunycz, P.J.; Freiberger, S.N.; Läuchli, S.; Hafner, J.; Vogl, T.; French, L.E.; Hofbauer, G.F.L. S100A8/A9 Stimulates Keratinocyte Proliferation in the Development of Squamous Cell Carcinoma of the Skin via the Receptor for Advanced Glycation-End Products. PLoS ONE 2015, 10, e0120971. [Google Scholar] [CrossRef] [Green Version]
  174. Hunter, M.J.; Chazin, W.J. High Level Expression and Dimer Characterization of the S100 EF-Hand Proteins, Migration Inhibitory Factor-Related Proteins 8 and 14. J. Biol. Chem. 1998, 273, 12427–12435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Leukert, N.; Sorg, C.; Roth, J. Molecular Basis of the Complex Formation between the Two Calcium-Binding Proteins S100A8 (MRP8) and S100A9 (MRP14). Biol. Chem. 2005, 386, 429–434. [Google Scholar] [CrossRef]
  176. Pröpper, C.; Huang, X.; Roth, J.; Sorg, C.; Nacken, W. Analysis of the MRP8-MRP14 Protein-Protein Interaction by the Two-Hybrid System Suggests a Prominent Role of the C-Terminal Domain of S100 Proteins in Dimer Formation. J. Biol. Chem. 1999, 274, 183–188. [Google Scholar] [CrossRef] [Green Version]
  177. Nacken, W.; Roth, J.; Sorg, C.; Kerkhoff, C. S100A9/S100A8: Myeloid Representatives of the S100 Protein Family as Prominent Players in Innate Immunity. Microsc. Res. Tech. 2003, 60, 569–580. [Google Scholar] [CrossRef] [PubMed]
  178. Stříž, I.; Trebichavský, I. Calprotectin—A Pleiotropic Molecule in Acute and Chronic Inflammation. Physiol. Res. 2004, 53, 245–253. [Google Scholar] [CrossRef] [PubMed]
  179. Ghavami, S.; Rashedi, I.; Dattilo, B.M.; Eshraghi, M.; Chazin, W.J.; Hashemi, M.; Wesselborg, S.; Kerkhoff, C.; Los, M. S100A8/A9 at Low Concentration Promotes Tumor Cell Growth via RAGE Ligation and MAP Kinase-dependent Pathway. J. Leukoc. Biol. 2008, 83, 1484–1492. [Google Scholar] [CrossRef] [PubMed]
  180. Vogl, T.; Tenbrock, K.; Ludwig, S.; Leukert, N.; Ehrhardt, C.; Van Zoelen, M.A.D.; Nacken, W.; Foell, D.; Van Der Poll, T.; Sorg, C.; et al. Mrp8 and Mrp14 Are Endogenous Activators of Toll-like Receptor 4, Promoting Lethal, Endotoxin-Induced Shock. Nat. Med. 2007, 13, 1042–1049. [Google Scholar] [CrossRef]
  181. Loser, K.; Vogl, T.; Voskort, M.; Lueken, A.; Kupas, V.; Nacken, W.; Klenner, L.; Kuhn, A.; Foell, D.; Sorokin, L. The Toll-like Receptor 4 Ligands Mrp8 and Mrp14 Are Crucial in the Development of Autoreactive CD8+ T Cells. Nat. Med. 2010, 16, 713–717. [Google Scholar] [CrossRef]
  182. Hermani, A.; De Servi, B.; Medunjanin, S.; Tessier, P.A.; Mayer, D. S100A8 and S100A9 Activate MAP Kinase and NF-ΚB Signaling Pathways and Trigger Translocation of RAGE in Human Prostate Cancer Cells. Exp. Cell Res. 2006, 312, 184–197. [Google Scholar] [CrossRef]
  183. Greten, F.R.; Eckmann, L.; Greten, T.F.; Park, J.M.; Li, Z.-W.; Egan, L.J.; Kagnoff, M.F.; Karin, M. IKKβ Links Inflammation and Tumorigenesis in a Mouse Model of Colitis-Associated Cancer. Cell 2004, 118, 285–296. [Google Scholar] [CrossRef] [Green Version]
  184. Karin, M.; Greten, F.R. NF-ΚB: Linking Inflammation and Immunity to Cancer Development and Progression. Nat. Rev. Immunol. 2005, 5, 749–759. [Google Scholar] [CrossRef] [PubMed]
  185. Németh, J.; Stein, I.; Haag, D.; Riehl, A.; Longerich, T.; Horwitz, E.; Breuhahn, K.; Gebhardt, C.; Schirmacher, P.; Hahn, M. S100A8 and S100A9 Are Novel Nuclear Factor Kappa B Target Genes during Malignant Progression of Murine and Human Liver Carcinogenesis. Hepatology 2009, 50, 1251–1262. [Google Scholar] [CrossRef] [PubMed]
  186. Itou, H.; Yao, M.; Fujita, I.; Watanabe, N.; Suzuki, M.; Nishihira, J.; Tanaka, I. The Crystal Structure of Human MRP14 (S100A9), a Ca2+-Dependent Regulator Protein in Inflammatory Process. J. Mol. Biol. 2002, 316, 265–276. [Google Scholar] [CrossRef] [PubMed]
  187. Korndörfer, I.P.; Brueckner, F.; Skerra, A. The Crystal Structure of the Human (S100A8/S100A9) 2 Heterotetramer, Calprotectin, Illustrates How Conformational Changes of Interacting α-Helices Can Determine Specific Association of Two EF-Hand Proteins. J. Mol. Biol. 2007, 370, 887–898. [Google Scholar] [CrossRef]
  188. Santamaria-Kisiel, L.; Rintala-Dempsey, A.C.; Shaw, G.S. Calcium-Dependent and-Independent Interactions of the S100 Protein Family. Biochem. J. 2006, 396, 201–214. [Google Scholar] [CrossRef] [Green Version]
  189. Ishikawa, K.; Nakagawa, A.; Tanaka, I.; Suzuki, M.; Nishihira, J. The Structure of Human MRP8, a Member of the S100 Calcium-Binding Protein Family, by MAD Phasing at 1.9 Å Resolution. Acta Crystallogr. Sect. D Biol. Crystallogr. 2000, 56, 559–566. [Google Scholar] [CrossRef]
  190. Pietas, A.; Schlüns, K.; Marenholz, I.; Schäfer, B.W.; Heizmann, C.W.; Petersen, I. Molecular Cloning and Characterization of the Human S100A14 Gene Encoding a Novel Member of the S100 Family. Genomics 2002, 79, 513–522. [Google Scholar] [CrossRef]
  191. Jin, Q.; Chen, H.; Luo, A.; Ding, F.; Liu, Z. S100A14 Stimulates Cell Proliferation and Induces Cell Apoptosis at Different Concentrations via Receptor for Advanced Glycation End Products (RAGE). PLoS ONE 2011, 6, e19375. [Google Scholar] [CrossRef]
  192. Bresnick, A.R.; Weber, D.J.; Zimmer, D.B. S100 Proteins in Cancer. Nat. Rev. Cancer 2015, 15, 96–109. [Google Scholar] [CrossRef] [Green Version]
  193. Sapkota, D.; Bruland, O.; Costea, D.E.; Haugen, H.; Vasstrand, E.N.; Ibrahim, S.O. S100A14 Regulates the Invasive Potential of Oral Squamous Cell Carcinoma Derived Cell-Lines in Vitro by Modulating Expression of Matrix Metalloproteinases, MMP1 and MMP9. Eur. J. Cancer 2011, 47, 600–610. [Google Scholar] [CrossRef]
  194. Sapkota, D.; Costea, D.E.; Blø, M.; Bruland, O.; Lorens, J.B.; Vasstrand, E.N.; Ibrahim, S.O. S100A14 Inhibits Proliferation of Oral Carcinoma Derived Cells through G1-Arrest. Oral Oncol. 2012, 48, 219–225. [Google Scholar] [CrossRef] [PubMed]
  195. Xu, C.; Chen, H.; Wang, X.; Gao, J.; Che, Y.; Li, Y.; Ding, F.; Luo, A.; Zhang, S.; Liu, Z. S100A14, a Member of the EF-Hand Calcium-Binding Proteins, Is Overexpressed in Breast Cancer and Acts as a Modulator of HER2 Signaling. J. Biol. Chem. 2014, 289, 827–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Bigelow, R.L.H.; Williams, B.J.; Carroll, J.L.; Daves, L.K.; Cardelli, J.A. TIMP-1 Overexpression Promotes Tumorigenesis of MDA-MB-231 Breast Cancer Cells and Alters Expression of a Subset of Cancer Promoting Genes in Vivo Distinct from Those Observed in Vitro. Breast Cancer Res. Treat. 2009, 117, 31–44. [Google Scholar] [CrossRef]
  197. Zhu, M.; Wang, H.; Cui, J.; Li, W.; An, G.; Pan, Y.; Zhang, Q.; Xing, R.; Lu, Y. Calcium-Binding Protein S100A14 Induces Differentiation and Suppresses Metastasis in Gastric Cancer. Cell Death Dis. 2017, 8, e2938. [Google Scholar] [CrossRef] [PubMed]
  198. Lavoie, C.; Meerloo, T.; Lin, P.; Farquhar, M.G. Calnuc, an EF-Hand Ca2+-Binding Protein, Is Stored and Processed in the Golgi and Secreted by the Constitutive-like Pathway in AtT20 Cells. Mol. Endocrinol. 2002, 16, 2462–2474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Meng, D.-F.; Sun, R.; Liu, G.-Y.; Peng, L.-X.; Zheng, L.-S.; Xie, P.; Lin, S.-T.; Mei, Y.; Qiang, Y.-Y.; Li, C.-Z. S100A14 Suppresses Metastasis of Nasopharyngeal Carcinoma by Inhibition of NF-KB Signaling through Degradation of IRAK1. Oncogene 2020, 39, 5307–5322. [Google Scholar] [CrossRef]
  200. Medapati, M.R.; Dahlmann, M.; Ghavami, S.; Pathak, K.A.; Lucman, L.; Klonisch, T.; Hoang-Vu, C.; Stein, U.; Hombach-Klonisch, S. RAGE Mediates the Pro-Migratory Response of Extracellular S100A4 in Human Thyroid Cancer Cells. Thyroid 2015, 25, 514–527. [Google Scholar] [CrossRef]
  201. Adam, P.J.; Boyd, R.; Tyson, K.L.; Fletcher, G.C.; Stamps, A.; Hudson, L.; Poyser, H.R.; Redpath, N.; Griffiths, M.; Steers, G. Comprehensive Proteomic Analysis of Breast Cancer Cell Membranes Reveals Unique Proteins with Potential Roles in Clinical Cancer. J. Biol. Chem. 2003, 278, 6482–6489. [Google Scholar] [CrossRef] [Green Version]
  202. Yao, R.; Lopez-Beltran, A.; Maclennan, G.T.; Montironi, R.; Eble, J.N.; Cheng, L. Expression of S100 Protein Family Members in the Pathogenesis of Bladder Tumors. Anticancer. Res. 2007, 27, 3051–3058. [Google Scholar]
  203. Smirnov, D.A.; Zweitzig, D.R.; Foulk, B.W.; Miller, M.C.; Doyle, G.V.; Pienta, K.J.; Meropol, N.J.; Weiner, L.M.; Cohen, S.J.; Moreno, J.G. Global Gene Expression Profiling of Circulating Tumor Cells. Cancer Res. 2005, 65, 4993–4997. [Google Scholar] [CrossRef] [Green Version]
  204. Chen, H.; Yu, D.; Luo, A.; Tan, W.; Zhang, C.; Zhao, D.; Yang, M.; Liu, J.; Lin, D.; Liu, Z. Functional Role of S100A14 Genetic Variants and Their Association with Esophageal Squamous Cell Carcinoma. Cancer Res. 2009, 69, 3451–3457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Bertini, I.; Borsi, V.; Cerofolini, L.; Das Gupta, S.; Fragai, M.; Luchinat, C. Solution Structure and Dynamics of Human S100A14. JBIC J. Biol. Inorg. Chem. 2013, 18, 183–194. [Google Scholar] [CrossRef] [PubMed]
  206. Nogueira-Machado, J.A.; M de Oliveira Volpe, C. HMGB-1 as a Target for Inflammation Controlling. Recent Pat. Endocr. Metab. Immune Drug Discov. 2012, 6, 201–209. [Google Scholar] [CrossRef] [PubMed]
  207. Arnesano, F.; Banci, L.; Bertini, I.; Fantoni, A.; Tenori, L.; Viezzoli, M.S. Structural Interplay between Calcium (II) and Copper (II) Binding to S100A13 Protein. Angew. Chem. 2005, 117, 6499–6502. [Google Scholar] [CrossRef]
  208. Otterbein, L.R.; Kordowska, J.; Witte-Hoffmann, C.; Wang, C.-L.A.; Dominguez, R. Crystal Structures of S100A6 in the Ca2+-Free and Ca2+-Bound States: The Calcium Sensor Mechanism of S100 Proteins Revealed at Atomic Resolution. Structure 2002, 10, 557–567. [Google Scholar] [CrossRef] [Green Version]
  209. Drohat, A.C.; Amburgey, J.C.; Abildgaard, F.; Starich, M.R.; Baldisseri, D.; Weber, D.J. Solution Structure of Rat Apo-S100B (Ββ) as Determined by NMR Spectroscopy. Biochemistry 1996, 35, 11577–11588. [Google Scholar] [CrossRef]
  210. Smith, S.P.; Shaw, G.S. A Novel Calcium-Sensitive Switch Revealed by the Structure of Human S100B in the Calcium-Bound Form. Structure 1998, 6, 211–222. [Google Scholar] [CrossRef]
  211. Kiess, A.P.; Cho, S.Y.; Pomper, M.G. Translational Molecular Imaging of Prostate Cancer. Curr. Radiol. Rep. 2013, 1, 216–226. [Google Scholar] [CrossRef] [Green Version]
  212. Marenholz, I.; Heizmann, C.W. S100A16, a Ubiquitously Expressed EF-Hand Protein Which Is up-Regulated in Tumors. Biochem. Biophys. Res. Commun. 2004, 313, 237–244. [Google Scholar] [CrossRef]
  213. Heizmann, C.W.; Fritz, G. The Family of S100 Cell Signaling Proteins. In Handbook of Cell Signaling; Elsevier: Amsterdam, The Netherlands, 2010; pp. 983–993. [Google Scholar]
  214. He, M.; Kubo, H.; Morimoto, K.; Fujino, N.; Suzuki, T.; Takahasi, T.; Yamada, M.; Yamaya, M.; Maekawa, T.; Yamamoto, Y. Receptor for Advanced Glycation End Products Binds to Phosphatidylserine and Assists in the Clearance of Apoptotic Cells. EMBO Rep. 2011, 12, 358–364. [Google Scholar] [CrossRef] [Green Version]
  215. Andersen, J.S.; Lam, Y.W.; Leung, A.K.L.; Ong, S.-E.; Lyon, C.E.; Lamond, A.I.; Mann, M. Nucleolar Proteome Dynamics. Nature 2005, 433, 77–83. [Google Scholar] [CrossRef] [PubMed]
  216. Sturchler, E.; Cox, J.A.; Durussel, I.; Weibel, M.; Heizmann, C.W. S100A16, a Novel Calcium-Binding Protein of the EF-Hand Superfamily. J. Biol. Chem. 2006, 281, 38905–38917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Sapkota, D.; Costea, D.E.; Ibrahim, S.O.; Johannessen, A.C.; Bruland, O. S100A14 Interacts with S100A16 and Regulates Its Expression in Human Cancer Cells. PLoS ONE 2013, 8, e76058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Zhou, W.; Pan, H.; Xia, T.; Xue, J.; Cheng, L.; Fan, P.; Zhang, Y.; Zhu, W.; Xue, Y.; Liu, X. Up-Regulation of S100A16 Expression Promotes Epithelial-Mesenchymal Transition via Notch1 Pathway in Breast Cancer. J. Biomed. Sci. 2014, 21, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Sapkota, D.; Bruland, O.; Parajuli, H.; Osman, T.A.; Teh, M.-T.; Johannessen, A.C.; Costea, D.E. S100A16 Promotes Differentiation and Contributes to a Less Aggressive Tumor Phenotype in Oral Squamous Cell Carcinoma. BMC Cancer 2015, 15, 631. [Google Scholar] [CrossRef] [Green Version]
  220. Kobayashi, M.; Nagashio, R.; Saito, K.; Aguilar-Bonavides, C.; Ryuge, S.; Katono, K.; Igawa, S.; Tsuchiya, B.; Jiang, S.-X.; Ichinoe, M. Prognostic Significance of S100A16 Subcellular Localization in Lung Adenocarcinoma. Hum. Pathol. 2018, 74, 148–155. [Google Scholar] [CrossRef] [PubMed]
  221. Schmitt, T.; Ogris, C.; Sonnhammer, E.L.L. FunCoup 3.0: Database of Genome-Wide Functional Coupling Networks. Nucleic Acids Res. 2014, 42, D380–D388. [Google Scholar] [CrossRef]
  222. Price, D.J.; Avraham, S.; Feuerstein, J.; Fu, Y.; Avraham, H.K. The Invasive Phenotype in HMT-3522 Cells Requires Increased EGF Receptor Signaling through Both PI 3-Kinase and ERK 1, 2 Pathways. Cell Commun. Adhes. 2002, 9, 87–102. [Google Scholar] [CrossRef]
  223. Peng, S.; Zhang, Y.; Zhang, J.; Wang, H.; Ren, B. ERK in Learning and Memory: A Review of Recent Research. Int. J. Mol. Sci. 2010, 11, 222–232. [Google Scholar] [CrossRef] [Green Version]
  224. Zhu, W.; Xue, Y.; Liang, C.; Zhang, R.; Zhang, Z.; Li, H.; Su, D.; Liang, X.; Zhang, Y.; Huang, Q. S100A16 Promotes Cell Proliferation and Metastasis via AKT and ERK Cell Signaling Pathways in Human Prostate Cancer. Tumor Biol. 2016, 37, 12241–12250. [Google Scholar] [CrossRef]
  225. Babini, E.; Bertini, I.; Borsi, V.; Calderone, V.; Hu, X.; Luchinat, C.; Parigi, G. Structural Characterization of Human S100A16, a Low-Affinity Calcium Binder. JBIC J. Biol. Inorg. Chem. 2011, 16, 243–256. [Google Scholar] [CrossRef] [PubMed]
  226. Steiner, J.; Bogerts, B.; Schroeter, M.L.; Bernstein, H.-G. S100B Protein in Neurodegenerative Disorders. Clin. Chem. Lab. Med. 2011, 49, 409–424. [Google Scholar] [CrossRef] [PubMed]
  227. Schmidt, A.P.; Tort, A.B.L.; Amaral, O.B.; Schmidt, A.P.; Walz, R.; Vettorazzi-Stuckzynski, J.; Martins-Costa, S.H.; Ramos, J.G.L.; Souza, D.O.; Portela, L.V.C. Serum S100B in Pregnancy-Related Hypertensive Disorders: A Case–Control Study. Clin. Chem. 2004, 50, 435–438. [Google Scholar] [CrossRef] [PubMed]
  228. Harpio, R.; Einarsson, R. S100 Proteins as Cancer Biomarkers with Focus on S100B in Malignant Melanoma. Clin. Biochem. 2004, 37, 512–518. [Google Scholar] [CrossRef] [PubMed]
  229. Hamberg, A.P.; Korse, C.M.; Bonfrer, J.M.G.; De Gast, G.C. Serum S100B Is Suitable for Prediction and Monitoring of Response to Chemoimmunotherapy in Metastatic Malignant Melanoma. Melanoma Res. 2003, 13, 45–49. [Google Scholar] [CrossRef]
  230. Huang, M.-Y.; Wang, H.-M.; Chang, H.-J.; Hsiao, C.-P.; Wang, J.-Y.; Lin, S.-R. Overexpression of S100B, TM4SF4, and OLFM4 Genes Is Correlated with Liver Metastasis in Taiwanese Colorectal Cancer Patients. DNA Cell Biol. 2012, 31, 43–49. [Google Scholar] [CrossRef] [Green Version]
  231. Hwang, C.-C.; Chai, H.-T.; Chen, H.-W.; Tsai, H.-L.; Lu, C.-Y.; Yu, F.-J.; Huang, M.-Y.; Wang, J.-Y. S100B Protein Expressions as an Independent Predictor of Early Relapse in UICC Stages II and III Colon Cancer Patients after Curative Resection. Ann. Surg. Oncol. 2011, 18, 139–145. [Google Scholar] [CrossRef]
  232. Villarreal, A.; Aviles Reyes, R.X.; Angelo, M.F.; Reines, A.G.; Ramos, A.J. S100B Alters Neuronal Survival and Dendrite Extension via RAGE-mediated NF-κB Signaling. J. Neurochem. 2011, 117, 321–332. [Google Scholar] [CrossRef]
  233. Wang, H.; Zhang, L.; Zhang, I.Y.; Chen, X.; Da Fonseca, A.; Wu, S.; Ren, H.; Badie, S.; Sadeghi, S.; Ouyang, M. S100B Promotes Glioma Growth through Chemoattraction of Myeloid-Derived Macrophages. Clin. Cancer Res. 2013, 19, 3764–3775. [Google Scholar] [CrossRef] [Green Version]
  234. Yang, T.; Cheng, J.; Yang, Y.; Qi, W.; Zhao, Y.; Long, H.; Xie, R.; Zhu, B. S100B Mediates Stemness of Ovarian Cancer Stem-like Cells through Inhibiting P53. Stem Cells 2017, 35, 325–336. [Google Scholar] [CrossRef] [Green Version]
  235. Zhang, F.; Banker, G.; Liu, X.; Suwanabol, P.A.; Lengfeld, J.; Yamanouchi, D.; Kent, K.C.; Liu, B. The Novel Function of Advanced Glycation End Products in Regulation of MMP-9 Production. J. Surg. Res. 2011, 171, 871–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Donato, R. RAGE: A Single Receptor for Several Ligands and Different Cellular Responses: The Case of Certain S100 Proteins. Curr. Mol. Med. 2007, 7, 711–724. [Google Scholar] [CrossRef] [PubMed]
  237. Baudier, J.; Delphin, C.; Grunwald, D.; Khochbin, S.; Lawrence, J.J. Characterization of the Tumor Suppressor Protein P53 as a Protein Kinase C Substrate and a S100b-Binding Protein. Proc. Natl. Acad. Sci. USA 1992, 89, 11627–11631. [Google Scholar] [CrossRef] [Green Version]
  238. Rustandi, R.R.; Drohat, A.C.; Baldisseri, D.M.; Wilder, P.T.; Weber, D.J. The Ca2+-Dependent Interaction of S100B (Ββ) with a Peptide Derived from P53. Biochemistry 1998, 37, 1951–1960. [Google Scholar] [CrossRef] [PubMed]
  239. Delphin, C.; Ronjat, M.; Deloulme, J.C.; Garin, G.; Debussche, L.; Higashimoto, Y.; Sakaguchi, K.; Baudier, J. Calcium-Dependent Interaction of S100B with the C-Terminal Domain of the Tumor Suppressor P53. J. Biol. Chem. 1999, 274, 10539–10544. [Google Scholar] [CrossRef] [Green Version]
  240. Lin, J.; Yang, Q.; Yan, Z.; Markowitz, J.; Wilder, P.T.; Carrier, F.; Weber, D.J. Inhibiting S100B Restores P53 Levels in Primary Malignant Melanoma Cancer Cells. J. Biol. Chem. 2004, 279, 34071–34077. [Google Scholar] [CrossRef] [Green Version]
  241. Lin, J.; Blake, M.; Tang, C.; Zimmer, D.; Rustandi, R.R.; Weber, D.J.; Carrier, F. Inhibition of P53 Transcriptional Activity by the S100B Calcium-Binding Protein. J. Biol. Chem. 2001, 276, 35037–35041. [Google Scholar] [CrossRef]
  242. Wilder, P.T.; Lin, J.; Bair, C.L.; Charpentier, T.H.; Yang, D.; Liriano, M.; Varney, K.M.; Lee, A.; Oppenheim, A.B.; Adhya, S. Recognition of the Tumor Suppressor Protein P53 and Other Protein Targets by the Calcium-Binding Protein S100B. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2006, 1763, 1284–1297. [Google Scholar] [CrossRef] [Green Version]
  243. Drohat, A.C.; Baldisseri, D.M.; Rustandi, R.R.; Weber, D.J. Solution Structure of Calcium-Bound Rat S100B (Ββ) as Determined by Nuclear Magnetic Resonance Spectroscopy. Biochemistry 1998, 37, 2729–2740. [Google Scholar] [CrossRef]
  244. Rust, R.R.; Baldisseri, D.M.; Weber, D.J. Structure of the Negative Regulatory Domain of P53 Bound to S100B (Ββ). Nat. Struct. Biol. 2000, 7, 570–574. [Google Scholar]
  245. Amburgey, J.C.; Abildgaard, F.; Starich, M.R.; Shah, S.; Hilt, D.C.; Weber, D.J. 1 H, 13 C and 15 N NMR Assignments and Solution Secondary Structure of Rat Apo-S100β. J. Biomol. NMR 1995, 6, 171–179. [Google Scholar] [CrossRef] [PubMed]
  246. Fritz, G.; Heizmann, C.W. 3D Structures of the Calcium and Zinc Binding S100 Proteins. In Handbook of Metalloproteins; Wiley: Hoboken, NJ, USA, 2004; Volume 3, pp. 529–540. [Google Scholar]
  247. Xie, J.; Méndez, J.D.; Méndez-Valenzuela, V.; Aguilar-Hernández, M.M. Cellular Signalling of the Receptor for Advanced Glycation End Products (RAGE). Cell. Signal. 2013, 25, 2185–2197. [Google Scholar] [CrossRef] [PubMed]
  248. Ostendorp, T.; Diez, J.; Heizmann, C.W.; Fritz, G. The Crystal Structures of Human S100B in the Zinc-and Calcium-Loaded State at Three PH Values Reveal Zinc Ligand Swapping. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2011, 1813, 1083–1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Sorci, G.; Riuzzi, F.; Agneletti, A.L.; Marchetti, C.; Donato, R. S100B Causes Apoptosis in a Myoblast Cell Line in a RAGE-independent Manner. J. Cell. Physiol. 2004, 199, 274–283. [Google Scholar] [CrossRef]
  250. Huttunen, H.J.; Kuja-Panula, J.; Sorci, G.; Agneletti, A.L.; Donato, R.; Rauvala, H. Coregulation of Neurite Outgrowth and Cell Survival by Amphoterin and S100 Proteins through Receptor for Advanced Glycation End Products (RAGE) Activation. J. Biol. Chem. 2000, 275, 40096–40105. [Google Scholar] [CrossRef] [Green Version]
  251. Marsden, B.J.; Shaw, G.S.; Sykes, B.D. Calcium Binding Proteins. Elucidating the Contributions to Calcium Affinity from an Analysis of Species Variants and Peptide Fragments. Biochem. Cell Biol. 1990, 68, 587–601. [Google Scholar] [CrossRef]
  252. Averboukh, L.; Liang, P.; Kantoff, P.W.; Pardee, A.B. Regulation of S100P Expression by Androgen. Prostate 1996, 29, 350–355. [Google Scholar] [CrossRef]
  253. Logsdon, C.D.; Simeone, D.M.; Binkley, C.; Arumugam, T.; Greenson, J.K.; Giordano, T.J.; Misek, D.E.; Hanash, S. Molecular Profiling of Pancreatic Adenocarcinoma and Chronic Pancreatitis Identifies Multiple Genes Differentially Regulated in Pancreatic Cancer. Cancer Res. 2003, 63, 2649–2657. [Google Scholar]
  254. Beer, D.G.; Kardia, S.L.R.; Huang, C.-C.; Giordano, T.J.; Levin, A.M.; Misek, D.E.; Lin, L.; Chen, G.; Gharib, T.G.; Thomas, D.G. Gene-Expression Profiles Predict Survival of Patients with Lung Adenocarcinoma. Nat. Med. 2002, 8, 816–824. [Google Scholar] [CrossRef]
  255. Fuentes, M.K.; Nigavekar, S.S.; Arumugam, T.; Logsdon, C.D.; Schmidt, A.M.; Park, J.C.; Huang, E.H. RAGE Activation by S100P in Colon Cancer Stimulates Growth, Migration, and Cell Signaling Pathways. Dis. Colon Rectum 2007, 50, 1230–1240. [Google Scholar] [CrossRef]
  256. Wang, G.; Platt-Higgins, A.; Carroll, J.; de Silva Rudland, S.; Winstanley, J.; Barraclough, R.; Rudland, P.S. Induction of Metastasis by S100P in a Rat Mammary Model and Its Association with Poor Survival of Breast Cancer Patients. Cancer Res. 2006, 66, 1199–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Rehbein, G.; Simm, A.; Hofmann, H.-S.; Silber, R.-E.; Bartling, B. Molecular Regulation of S100P in Human Lung Adenocarcinomas. Int. J. Mol. Med. 2008, 22, 69–77. [Google Scholar] [CrossRef] [PubMed]
  258. Wang, Q.; Zhang, Y.-N.; Lin, G.-L.; Qiu, H.-Z.; Wu, B.; Wu, H.-Y.; Zhao, Y.; Chen, Y.-J.; Lu, C.-M. S100P, a Potential Novel Prognostic Marker in Colorectal Cancer. Oncol. Rep. 2012, 28, 303–310. [Google Scholar] [PubMed]
  259. Tothova, V.; Gibadulinova, A. S100P, a Peculiar Member of S100 Family of Calcium-Binding Proteins Implicated in Cancer. Acta Virol. 2013, 57, 238–246. [Google Scholar] [CrossRef] [Green Version]
  260. Lee, Y.-C.; Volk, D.E.; Thiviyanathan, V.; Kleerekoper, Q.; Gribenko, A.V.; Zhang, S.; Gorenstein, D.G.; Makhatadze, G.I.; Luxon, B.A. Letter to the Editor: NMR Structure of the Apo-S100P Protein. J. Biomol. NMR 2004, 29, 399–402. [Google Scholar] [CrossRef]
  261. Austermann, J.; Nazmi, A.R.; Müller-Tidow, C.; Gerke, V. Characterization of the Ca2+-Regulated Ezrin-S100P Interaction and Its Role in Tumor Cell Migration∗. J. Biol. Chem. 2008, 283, 29331–29340. [Google Scholar] [CrossRef] [Green Version]
  262. Cavallaro, U.G.O.; Christofori, G. Multitasking in Tumor Progression: Signaling Functions of Cell Adhesion Molecules. Ann. New York Acad. Sci. 2004, 1014, 58–66. [Google Scholar] [CrossRef]
  263. Beauchemin, N.; Arabzadeh, A. Carcinoembryonic Antigen-Related Cell Adhesion Molecules (CEACAMs) in Cancer Progression and Metastasis. Cancer Metastasis Rev. 2013, 32, 643–671. [Google Scholar] [CrossRef]
  264. Johnson, J.P.; Stade, B.G.; Holzmann, B.; Schwäble, W.; Riethmüller, G. De Novo Expression of Intercellular-Adhesion Molecule 1 in Melanoma Correlates with Increased Risk of Metastasis. Proc. Natl. Acad. Sci. USA 1989, 86, 641–644. [Google Scholar] [CrossRef]
  265. Lin, Y.-C.; Shun, C.-T.; Wu, M.-S.; Chen, C.-C. A Novel Anticancer Effect of Thalidomide: Inhibition of Intercellular Adhesion Molecule-1–Mediated Cell Invasion and Metastasis through Suppression of Nuclear Factor-ΚB. Clin. Cancer Res. 2006, 12, 7165–7173. [Google Scholar] [CrossRef] [Green Version]
  266. Schröder, C.; Witzel, I.; Müller, V.; Krenkel, S.; Wirtz, R.M.; Jänicke, F.; Schumacher, U.; Milde-Langosch, K. Prognostic Value of Intercellular Adhesion Molecule (ICAM)-1 Expression in Breast Cancer. J. Cancer Res. Clin. Oncol. 2011, 137, 1193–1201. [Google Scholar] [CrossRef] [PubMed]
  267. Usami, Y.; Ishida, K.; Sato, S.; Kishino, M.; Kiryu, M.; Ogawa, Y.; Okura, M.; Fukuda, Y.; Toyosawa, S. Intercellular Adhesion Molecule-1 (ICAM-1) Expression Correlates with Oral Cancer Progression and Induces Macrophage/Cancer Cell Adhesion. Int. J. Cancer 2013, 133, 568–578. [Google Scholar] [CrossRef] [PubMed]
  268. Farahani, E.; Patra, H.K.; Jangamreddy, J.R.; Rashedi, I.; Kawalec, M.; Rao Pariti, R.K.; Batakis, P.; Wiechec, E. Cell Adhesion Molecules and Their Relation to (Cancer) Cell Stemness. Carcinogenesis 2014, 35, 747–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Buckley, S.T.; Medina, C.; Kasper, M.; Ehrhardt, C. Interplay between RAGE, CD44, and Focal Adhesion Molecules in Epithelial-Mesenchymal Transition of Alveolar Epithelial Cells. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2011, 300, L548–L559. [Google Scholar] [CrossRef] [Green Version]
  270. Sessa, L.; Gatti, E.; Zeni, F.; Antonelli, A.; Catucci, A.; Koch, M.; Pompilio, G.; Fritz, G.; Raucci, A.; Bianchi, M.E. The Receptor for Advanced Glycation End-Products (RAGE) Is Only Present in Mammals, and Belongs to a Family of Cell Adhesion Molecules (CAMs). PLoS ONE 2014, 9, e86903. [Google Scholar] [CrossRef]
  271. Paschos, K.A.; Canovas, D.; Bird, N.C. The Role of Cell Adhesion Molecules in the Progression of Colorectal Cancer and the Development of Liver Metastasis. Cell. Signal. 2009, 21, 665–674. [Google Scholar] [CrossRef]
  272. Zecchini, S.; Cavallaro, U. Neural Cell Adhesion Molecule in Cancer: Expression and Mechanisms. In Structure and Function of the Neural Cell Adhesion Molecule NCAM; Springer: New York, NY, USA, 2010; pp. 319–333. [Google Scholar]
  273. Bergom, C.; Gao, C.; Newman, P.J. Mechanisms of PECAM-1-Mediated Cytoprotection and Implications for Cancer Cell Survival. Leuk. Lymphoma 2005, 46, 1409–1421. [Google Scholar] [CrossRef]
  274. Weichert, W.; Knösel, T.; Bellach, J.; Dietel, M.; Kristiansen, G. ALCAM/CD166 Is Overexpressed in Colorectal Carcinoma and Correlates with Shortened Patient Survival. J. Clin. Pathol. 2004, 57, 1160–1164. [Google Scholar] [CrossRef]
  275. Roland, C.L.; Harken, A.H.; Sarr, M.G.; Barnett Jr, C.C. ICAM-1 Expression Determines Malignant Potential of Cancer. Surgery 2007, 141, 705–707. [Google Scholar] [CrossRef]
  276. Donin, N.; Jurianz, K.; Ziporen, L.; Schultz, S.; Kirschfink, M.; Fishelson, Z. Complement Resistance of Human Carcinoma Cells Depends on Membrane Regulatory Proteins, Protein Kinases and Sialic Acid. Clin. Exp. Immunol. 2003, 131, 254–263. [Google Scholar] [CrossRef]
  277. Okroj, M.; Hsu, Y.-F.; Ajona, D.; Pio, R.; Blom, A.M. Non-Small Cell Lung Cancer Cells Produce a Functional Set of Complement Factor I and Its Soluble Cofactors. Mol. Immunol. 2008, 45, 169–179. [Google Scholar] [CrossRef] [PubMed]
  278. Ajona, D.; Castano, Z.; Garayoa, M.; Zudaire, E.; Pajares, M.J.; Martinez, A.; Cuttitta, F.; Montuenga, L.M.; Pio, R. Expression of Complement Factor H by Lung Cancer Cells: Effects on the Activation of the Alternative Pathway of Complement. Cancer Res. 2004, 64, 6310–6318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  279. Ajona, D.; Ortiz-Espinosa, S.; Pio, R. Complement Anaphylatoxins C3a and C5a: Emerging Roles in Cancer Progression and Treatment. Semin. Cell Dev. Biol. 2019, 85, 153–163. [Google Scholar] [CrossRef] [PubMed]
  280. Ajona, D.; Hsu, Y.-F.; Corrales, L.; Montuenga, L.M.; Pio, R. Down-Regulation of Human Complement Factor H Sensitizes Non-Small Cell Lung Cancer Cells to Complement Attack and Reduces in Vivo Tumor Growth. J. Immunol. 2007, 178, 5991–5998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  281. Reid, K. Complement Component C1q: Historical Perspective of a Functionally Versatile, and Structurally Unusual, Serum Protein. Front. Immunol. 2018, 9, 764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Teh, B.K.; Yeo, J.G.; Chern, L.M.; Lu, J. C1q Regulation of Dendritic Cell Development from Monocytes with Distinct Cytokine Production and T Cell Stimulation. Mol. Immunol. 2011, 48, 1128–1138. [Google Scholar] [CrossRef] [PubMed]
  283. Lauvrak, V.; Brekke, O.H.; Ihle, Ø.; Lindqvist, B.H. Identification and Characterisation of C1q-Binding Phage Displayed Peptides. Biol. Chem. 1997, 378, 1509–1520. [Google Scholar] [CrossRef]
  284. Goodman, E.B.; Anderson, D.C.; Tenner, A.J. C1q Triggers Neutrophil Superoxide Production by a Unique CD18-dependent Mechanism. J. Leukoc. Biol. 1995, 58, 168–176. [Google Scholar] [CrossRef]
  285. Ma, W.; Rai, V.; Hudson, B.I.; Song, F.; Schmidt, A.M.; Barile, G.R. RAGE Binds C1q and Enhances C1q-Mediated Phagocytosis. Cell. Immunol. 2012, 274, 72–82. [Google Scholar] [CrossRef]
  286. Pamplona, R. Advanced Lipoxidation End-Products. Chem. -Biol. Interact. 2011, 192, 14–20. [Google Scholar] [CrossRef]
  287. Viedma-Poyatos, Á.; González-Jiménez, P.; Langlois, O.; Spickett, C.M.; Pérez-Sala, D. Protein Lipoxidation: Basic Concepts and Emerging Roles. Antioxidants 2021, 10, 295. [Google Scholar] [CrossRef] [PubMed]
  288. Dianzani, M.U. Lipid Peroxidation and Cancer: A Critical Reconsideration. Tumori J. 1989, 75, 351–357. [Google Scholar] [CrossRef] [PubMed]
  289. Biasi, F.; Vizio, B.; Mascia, C.; Gaia, E.; Zarkovic, N.; Chiarpotto, E.; Leonarduzzi, G.; Poli, G. C-Jun N-Terminal Kinase Upregulation as a Key Event in the Proapoptotic Interaction between Transforming Growth Factor-Β1 and 4-Hydroxynonenal in Colon Mucosa. Free. Radic. Biol. Med. 2006, 41, 443–454. [Google Scholar] [CrossRef]
  290. Mol, M.; Degani, G.; Coppa, C.; Baron, G.; Popolo, L.; Carini, M.; Aldini, G.; Vistoli, G.; Altomare, A. Advanced Lipoxidation End Products (ALEs) as RAGE Binders: Mass Spectrometric and Computational Studies to Explain the Reasons Why. Redox Biol. 2019, 23, 101083. [Google Scholar] [CrossRef]
  291. Rakhesh, M.; Cate, M.; Vijay, R.; Shrikant, A.; Shanjana, A. A TLR4-Interacting Peptide Inhibits Lipopolysaccharide-Stimulated Inflammatory Responses, Migration and Invasion of Colon Cancer SW480 Cells. Oncoimmunology 2012, 1, 1495–1506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Liu, X.; Liang, J.; Li, G. Lipopolysaccharide Promotes Adhesion and Invasion of Hepatoma Cell Lines HepG2 and HepG2. 2.15. Mol. Biol. Rep. 2010, 37, 2235–2239. [Google Scholar] [CrossRef]
  293. Huang, T.A.O.; Chen, Z.; Fang, L. Curcumin Inhibits LPS-Induced EMT through Downregulation of NF-ΚB-Snail Signaling in Breast Cancer Cells. Oncol. Rep. 2013, 29, 117–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Yamamoto, Y.; Harashima, A.; Saito, H.; Tsuneyama, K.; Munesue, S.; Motoyoshi, S.; Han, D.; Watanabe, T.; Asano, M.; Takasawa, S. Septic Shock Is Associated with Receptor for Advanced Glycation End Products Ligation of LPS. J. Immunol. 2011, 186, 3248–3257. [Google Scholar] [CrossRef] [Green Version]
  295. Nakashima, H.; Nakamura, M.; Yamaguchi, H.; Yamanaka, N.; Akiyoshi, T.; Koga, K.; Yamaguchi, K.; Tsuneyoshi, M.; Tanaka, M.; Katano, M. Nuclear Factor-ΚB Contributes to Hedgehog Signaling Pathway Activation through Sonic Hedgehog Induction in Pancreatic Cancer. Cancer Res. 2006, 66, 7041–7049. [Google Scholar] [CrossRef]
  296. Kojima, M.; Morisaki, T.; Izuhara, K.; Uchiyama, A.; Matsunari, Y.; Katano, M.; Tanaka, M. Lipopolysaccharide Increases Cyclo-Oxygenase-2 Expression in a Colon Carcinoma Cell Line through Nuclear Factor-ΚB Activation. Oncogene 2000, 19, 1225–1231. [Google Scholar] [CrossRef] [Green Version]
  297. Wilkie, T.; Verma, A.K.; Zhao, H.; Charan, M.; Ahirwar, D.K.; Kant, S.; Pancholi, V.; Mishra, S.; Ganju, R.K. Lipopolysaccharide from the Commensal Microbiota of the Breast Enhances Cancer Growth: Role of S100A7 and TLR4. Mol. Oncol. 2021, 16, 1508–1522. [Google Scholar] [CrossRef] [PubMed]
  298. Nasser, M.W.; Wani, N.A.; Ahirwar, D.K.; Powell, C.A.; Ravi, J.; Elbaz, M.; Zhao, H.; Padilla, L.; Zhang, X.; Shilo, K. RAGE Mediates S100A7-Induced Breast Cancer Growth and Metastasis by Modulating the Tumor Microenvironment. Cancer Res. 2015, 75, 974–985. [Google Scholar] [CrossRef] [Green Version]
  299. You, L.; Cui, H.; Zhao, F.; Sun, H.; Zhong, H.; Zhou, G.; Chen, X. Inhibition of HMGB1/RAGE Axis Suppressed the Lipopolysaccharide (LPS)-Induced Vicious Transformation of Cervical Epithelial Cells. Bioengineered 2021, 12, 4995–5003. [Google Scholar] [CrossRef] [PubMed]
  300. Sharaf, H.; Matou-Nasri, S.; Wang, Q.; Rabhan, Z.; Al-Eidi, H.; Al Abdulrahman, A.; Ahmed, N. Advanced Glycation Endproducts Increase Proliferation, Migration and Invasion of the Breast Cancer Cell Line MDA-MB-231. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 429–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Allmen, E.U.; Koch, M.; Fritz, G.; Legler, D.F. V Domain of RAGE Interacts with AGEs on Prostate Carcinoma Cells. Prostate 2008, 68, 748–758. [Google Scholar] [CrossRef] [Green Version]
  302. Poser, I.; Bosserhoff, A.K. Transcription Factors Involved in Development and Progression of Malignant Melanoma. Histol. Histopathol. 2004, 19, 173–188. [Google Scholar]
  303. Hernandez, J.L.; Padilla, L.; Dakhel, S.; Coll, T.; Hervas, R.; Adan, J.; Masa, M.; Mitjans, F.; Martinez, J.M.; Coma, S. Therapeutic Targeting of Tumor Growth and Angiogenesis with a Novel Anti-S100A4 Monoclonal Antibody. PLoS ONE 2013, 8, e72480. [Google Scholar] [CrossRef] [Green Version]
  304. Dakhel, S.; Padilla, L.; Adan, J.; Masa, M.; Martinez, J.M.; Roque, L.; Coll, T.; Hervas, R.; Calvis, C.; Messeguer, R. S100P Antibody-Mediated Therapy as a New Promising Strategy for the Treatment of Pancreatic Cancer. Oncogenesis 2014, 3, e92. [Google Scholar] [CrossRef] [Green Version]
  305. Li, A.; Shi, D.; Xu, B.; Wang, J.; Tang, Y.; Xiao, W.; Shen, G.; Deng, W.; Zhao, C. S100A6 Promotes Cell Proliferation in Human Nasopharyngeal Carcinoma via the P38/MAPK Signaling Pathway. Mol. Carcinog. 2017, 56, 972–984. [Google Scholar] [CrossRef]
  306. Sakurai, M.; Miki, Y.; Takagi, K.; Suzuki, T.; Ishida, T.; Ohuchi, N.; Sasano, H. Interaction with Adipocyte Stromal Cells Induces Breast Cancer Malignancy via S100A7 Upregulation in Breast Cancer Microenvironment. Breast Cancer Res. 2017, 19, 70. [Google Scholar] [CrossRef]
  307. Basnet, S.; Sharma, S.; Costea, D.E.; Sapkota, D. Expression Profile and Functional Role of S100A14 in Human Cancer. Oncotarget 2019, 10, 2996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  308. Ichikawa, M.; Williams, R.; Wang, L.; Vogl, T.; Srikrishna, G. S100A8/A9 Activate Key Genes and Pathways in Colon Tumor Progression. Mol. Cancer Res. 2011, 9, 133–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  309. Wan, S.; Taccioli, C.; Jiang, Y.; Chen, H.; Smalley, K.J.; Huang, K.; Liu, X.; Farber, J.L.; Croce, C.M.; Fong, L.Y.Y. Zinc Deficiency Activates S100A8 Inflammation in the Absence of COX-2 and Promotes Murine Oral-esophageal Tumor Progression. Int. J. Cancer 2011, 129, 331–345. [Google Scholar] [CrossRef]
  310. De Ponti, A.; Wiechert, L.; Schneller, D.; Pusterla, T.; Longerich, T.; Hogg, N.; Vogel, A.; Schirmacher, P.; Hess, J.; Angel, P. A Pro-Tumorigenic Function of S100A8/A9 in Carcinogen-Induced Hepatocellular Carcinoma. Cancer Lett. 2015, 369, 396–404. [Google Scholar] [CrossRef] [PubMed]
  311. Yang, T.; Cheng, J.; You, J.; Yan, B.; Liu, H.; Li, F. S100B Promotes Chemoresistance in Ovarian Cancer Stem Cells by Regulating P53. Oncol. Rep. 2018, 40, 1574–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Tan, B.; Shen, L.; Yang, K.; Huang, D.; Li, X.; Li, Y.; Zhao, L.; Chen, J.; Yi, Q.; Xu, H. C6 Glioma-Conditioned Medium Induces Malignant Transformation of Mesenchymal Stem Cells: Possible Role of S100B/RAGE Pathway. Biochem. Biophys. Res. Commun. 2018, 495, 78–85. [Google Scholar] [CrossRef] [PubMed]
  313. Seguella, L.; Capuano, R.; Pesce, M.; Annunziata, G.; Pesce, M.; de Conno, B.; Sarnelli, G.; Aurino, L.; Esposito, G. S100B Protein Stimulates Proliferation and Angiogenic Mediators Release through RAGE/PAkt/MTOR Pathway in Human Colon Adenocarcinoma Caco-2 Cells. Int. J. Mol. Sci. 2019, 20, 3240. [Google Scholar] [CrossRef] [Green Version]
  314. Liu, Y.; Wang, C.; Shan, X.; Wu, J.; Liu, H.; Liu, H.; Zhang, J.; Xu, W.; Sha, Z.; He, J. S100P Is Associated with Proliferation and Migration in Nasopharyngeal Carcinoma. Oncol. Lett. 2017, 14, 525–532. [Google Scholar] [CrossRef]
  315. Arumugam, T.; Ramachandran, V.; Logsdon, C.D. Effect of Cromolyn on S100P Interactions with RAGE and Pancreatic Cancer Growth and Invasion in Mouse Models. J. Natl. Cancer Inst. 2006, 98, 1806–1818. [Google Scholar] [CrossRef]
  316. Köhler, G.; Milstein, C. Derivation of Specific Antibody-producing Tissue Culture and Tumor Lines by Cell Fusion. Eur. J. Immunol. 1976, 6, 511–519. [Google Scholar] [CrossRef]
  317. Healey, G.D.; Pan-Castillo, B.; Garcia-Parra, J.; Davies, J.; Roberts, S.; Jones, E.; Dhar, K.; Nandanan, S.; Tofazzal, N.; Piggott, L. Antibody Drug Conjugates against the Receptor for Advanced Glycation End Products (RAGE), a Novel Therapeutic Target in Endometrial Cancer. J. Immunother. Cancer 2019, 7, 280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Esumi, S.; Kitamura, Y.; Yokota-Kumasaki, H.; Ushio, S.; Yamada-Takemoto, A.; Nagai, R.; Ogawa, A.; Kawasaki, Y.; Sendo, T. Effects of Magnesium Oxide on the Serum Duloxetine Concentration and Antidepressant-like Effects of Duloxetine in Rats. Biol. Pharm. Bull. 2018, 41, 1727–1731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  319. Mizumoto, S.; Takahashi, J.; Sugahara, K. Receptor for Advanced Glycation End Products (RAGE) Functions as Receptor for Specific Sulfated Glycosaminoglycans, and Anti-RAGE Antibody or Sulfated Glycosaminoglycans Delivered in Vivo Inhibit Pulmonary Metastasis of Tumor Cells. J. Biol. Chem. 2012, 287, 18985–18994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  320. Muthyalaiah, Y.S.; Jonnalagadda, B.; John, C.M.; Arockiasamy, S. Impact of Advanced Glycation End products (AGEs) and its receptor (RAGE) on cancer metabolic signaling pathways and its progression. Glycoconj J. 2021, 38, 717–734. [Google Scholar] [CrossRef]
  321. Paleiron, N.; Bylicki, O.; André, M.; Rivière, E.; Grassin, F.; Robinet, G.; Chouaïd, C. Targeted Therapy for Localized Non-Small-Cell Lung Cancer: A Review. OncoTargets Ther. 2016, 9, 4099. [Google Scholar]
  322. Liang, H.; Zhong, Y.; Zhou, S.; Peng, L. Knockdown of RAGE Expression Inhibits Colorectal Cancer Cell Invasion and Suppresses Angiogenesis in Vitro and in Vivo. Cancer Lett. 2011, 313, 91–98. [Google Scholar] [CrossRef]
  323. Yu, Y.X.; Pan, W.C.; Cheng, Y.F. Silencing of Advanced Glycosylation and Glycosylation and Product-Specific Receptor (RAGE) Inhibits the Metastasis and Growth of Non-Small Cell Lung Cancer. Am. J. Transl. Res. 2017, 9, 2760. [Google Scholar]
  324. Finkenzeller, G.; Sparacio, A.; Technau, A.; Marmé, D.; Siemeister, G. Sp1 Recognition Sites in the Proximal Promoter of the Human Vascular Endothelial Growth Factor Gene Are Essential for Platelet-Derived Growth Factor-Induced Gene Expression. Oncogene 1997, 15, 669–676. [Google Scholar] [CrossRef] [Green Version]
  325. Li, J.; Wu, P.-W.; Zhou, Y.; Dai, B.; Zhang, P.-F.; Zhang, Y.-H.; Liu, Y.; Shi, X.-L. Rage Induces Hepatocellular Carcinoma Proliferation and Sorafenib Resistance by Modulating Autophagy. Cell Death Dis. 2018, 9, 225. [Google Scholar] [CrossRef] [Green Version]
  326. Abdelrahim, M.; Smith, R.; Burghardt, R.; Safe, S. Role of Sp Proteins in Regulation of Vascular Endothelial Growth Factor Expression and Proliferation of Pancreatic Cancer Cells. Cancer Res. 2004, 64, 6740–6749. [Google Scholar] [CrossRef] [Green Version]
  327. Matsuoka, H.; Makimura, C.; Koyama, A.; Otsuka, M.; Okamoto, W.; Fujisaka, Y.; Kaneda, H.; Tsurutani, J.; Nakagawa, K. Pilot Study of Duloxetine for Cancer Patients with Neuropathic Pain Non-Responsive to Pregabalin. Anticancer. Res. 2012, 32, 1805–1809. [Google Scholar] [PubMed]
  328. Voigtlaender, M.; Langer, F. Low-Molecular-Weight Heparin in Cancer Patients: Overview and Indications. Hämostaseologie 2019, 39, 67–75. [Google Scholar] [CrossRef]
  329. Dowling, R.J.O.; Goodwin, P.J.; Stambolic, V. Understanding the Benefit of Metformin Use in Cancer Treatment. BMC Med. 2011, 9, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  330. Bodmer, M.; Meier, C.; Krähenbühl, S.; Jick, S.S.; Meier, C.R. Long-Term Metformin Use Is Associated with Decreased Risk of Breast Cancer. Diabetes Care 2010, 33, 1304–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Gao, H.; Zhang, I.Y.; Zhang, L.; Song, Y.; Liu, S.; Ren, H.; Liu, H.; Zhou, H.; Su, Y.; Yang, Y. S100B Suppression Alters Polarization of Infiltrating Myeloid-Derived Cells in Gliomas and Inhibits Tumor Growth. Cancer Lett. 2018, 439, 91–100. [Google Scholar] [CrossRef] [PubMed]
  332. Col, N.F.; Ochs, L.; Springmann, V.; Aragaki, A.K.; Chlebowski, R.T. Metformin and Breast Cancer Risk: A Meta-Analysis and Critical Literature Review. Breast Cancer Res. Treat. 2012, 135, 639–646. [Google Scholar] [CrossRef]
  333. Currie, C.J.; Poole, C.D.; Jenkins-Jones, S.; Gale, E.A.M.; Johnson, J.A.; Morgan, C.L. Mortality after Incident Cancer in People with and without Type 2 Diabetes: Impact of Metformin on Survival. Diabetes Care 2012, 35, 299–304. [Google Scholar] [CrossRef] [Green Version]
  334. Ishibashi, Y.; Matsui, T.; Takeuchi, M.; Yamagishi, S. Metformin Inhibits Advanced Glycation End Products (AGEs)-Induced Growth and VEGF Expression in MCF-7 Breast Cancer Cells by Suppressing AGEs Receptor Expression via AMP-Activated Protein Kinase. Horm. Metab. Res. 2013, 45, 387–390. [Google Scholar] [CrossRef]
  335. Pellegrini, L.; Xue, J.; Larson, D.; Pastorino, S.; Jube, S.; Forest, K.H.; Saad-Jube, Z.S.; Napolitano, A.; Pagano, I.; Negi, V.S. HMGB1 Targeting by Ethyl Pyruvate Suppresses Malignant Phenotype of Human Mesothelioma. Oncotarget 2017, 8, 22649. [Google Scholar] [CrossRef] [Green Version]
  336. Chen, B.; Na, F.; Yang, H.; Li, R.; Li, M.; Sun, X.; Hu, B.; Huang, G.; Lan, J.; Xu, H. Ethyl Pyruvate Alleviates Radiation-Induced Lung Injury in Mice. Biomed. Pharmacother. 2017, 92, 468–478. [Google Scholar] [CrossRef]
  337. Liu, Q.; Huo, Y.; Zheng, H.; Zhao, J.; Jia, L.; Wang, P. Ethyl Pyruvate Suppresses the Growth, Invasion and Migration and Induces the Apoptosis of Non-small Cell Lung Cancer Cells via the HMGB1/RAGE Axis and the NF-κB/STAT3 Pathway. Oncol. Rep. 2019, 42, 817–825. [Google Scholar] [CrossRef] [PubMed]
  338. Inada, M.; Shindo, M.; Kobayashi, K.; Sato, A.; Yamamoto, Y.; Akasaki, Y.; Ichimura, K.; Tanuma, S. Anticancer Effects of a Non-Narcotic Opium Alkaloid Medicine, Papaverine, in Human Glioblastoma Cells. PLoS ONE 2019, 14, e0216358. [Google Scholar] [CrossRef] [PubMed]
  339. Lee, S.; Piao, C.; Kim, G.; Kim, J.Y.; Choi, E.; Lee, M. Production and Application of HMGB1 Derived Recombinant RAGE-Antagonist Peptide for Anti-Inflammatory Therapy in Acute Lung Injury. Eur. J. Pharm. Sci. 2018, 114, 275–284. [Google Scholar] [CrossRef] [PubMed]
  340. Park, I.-H.; Chung, S.-K.; Lee, K.-B.; Yoo, Y.-C.; Kim, S.-K.; Kim, G.-S.; Song, K.-S. An Antioxidant Hispidin from the Mycelial Cultures of Phellinus Linteus. Arch. Pharmacal Res. 2004, 27, 615–618. [Google Scholar] [CrossRef] [PubMed]
  341. El-Far, A.H.A.M.; Munesue, S.; Harashima, A.; Sato, A.; Shindo, M.; Nakajima, S.; Inada, M.; Tanaka, M.; Takeuchi, A.; Tsuchiya, H. In Vitro Anticancer Effects of a RAGE Inhibitor Discovered Using a Structure-Based Drug Design System. Oncol. Lett. 2018, 15, 4627–4634. [Google Scholar] [CrossRef] [Green Version]
  342. Chandimali, N.; JIN, W.O.O.Y.; KWON, T. Combination Effects of Hispidin and Gemcitabine via Inhibition of Stemness in Pancreatic Cancer Stem Cells. Anticancer. Res. 2018, 38, 3967–3975. [Google Scholar] [CrossRef]
  343. Song, T.-Y.; Yang, N.-C.; Chen, C.-L.; Thi, T.L.V. Protective Effects and Possible Mechanisms of Ergothioneine and Hispidin against Methylglyoxal-Induced Injuries in Rat Pheochromocytoma Cells. Oxidative Med. Cell. Longev. 2017, 2017, 4824371. [Google Scholar] [CrossRef]
Figure 1. (A) Head-to-toe organization of receptor for advanced glycation end products (RAGE), consisting of a signal protein (residues 1–22), extracellular domains (V (residues 23–116), C1 (residues 124–221), and C2 (residues 227–317), a transmembrane domain (residues 343–363), and a cytosolic domain (residues 364–404); and (B) a hypothetical model of the extracellular region of RAGE with V, C1, and C2 domains.
Figure 1. (A) Head-to-toe organization of receptor for advanced glycation end products (RAGE), consisting of a signal protein (residues 1–22), extracellular domains (V (residues 23–116), C1 (residues 124–221), and C2 (residues 227–317), a transmembrane domain (residues 343–363), and a cytosolic domain (residues 364–404); and (B) a hypothetical model of the extracellular region of RAGE with V, C1, and C2 domains.
Ijms 24 00266 g001
Figure 2. Schematic illustration of the functions of a receptor for advanced glycation end products (RAGE) in the cancer microenvironment: (A) binding of RAGE and its ligands promotes angiogenesis and cancer development by upregulating PKI3/mTOR/Akt, MAPKs, MMPs, VEGF, NF-κB, and downregulating p53 signaling; (B) secretion of HMGB1 from inflamed or injured cells followed by its binding with RAGE results in the increased release of proinflammatory cytokines via NF-κB activation and ensuing harmful inflammatory responses; (C) HMGB1 operates via NF-κB and epigenetic pathways in the cancer microenvironment to exert its long-lasting effects on surviving tumor cells, immune cells, and stromal cells. Together with STAT3, NF-κB controls genes that promote metastasis, angiogenesis, and cancer development; (D) upregulation of VEGF leads to angiogenesis resulting in cancer progression; and (E) increased expression of MMPs and MMPKs promotes metastasis in various cancers.
Figure 2. Schematic illustration of the functions of a receptor for advanced glycation end products (RAGE) in the cancer microenvironment: (A) binding of RAGE and its ligands promotes angiogenesis and cancer development by upregulating PKI3/mTOR/Akt, MAPKs, MMPs, VEGF, NF-κB, and downregulating p53 signaling; (B) secretion of HMGB1 from inflamed or injured cells followed by its binding with RAGE results in the increased release of proinflammatory cytokines via NF-κB activation and ensuing harmful inflammatory responses; (C) HMGB1 operates via NF-κB and epigenetic pathways in the cancer microenvironment to exert its long-lasting effects on surviving tumor cells, immune cells, and stromal cells. Together with STAT3, NF-κB controls genes that promote metastasis, angiogenesis, and cancer development; (D) upregulation of VEGF leads to angiogenesis resulting in cancer progression; and (E) increased expression of MMPs and MMPKs promotes metastasis in various cancers.
Ijms 24 00266 g002
Figure 3. Three-dimensional representation of S100A6 and receptor for advanced glycation end products (RAGE) interacting amino acid residues: (A) S100A6 residues that interact with RAGE; and (B) residues in the V-domain of RAGE that interact with S100A6.
Figure 3. Three-dimensional representation of S100A6 and receptor for advanced glycation end products (RAGE) interacting amino acid residues: (A) S100A6 residues that interact with RAGE; and (B) residues in the V-domain of RAGE that interact with S100A6.
Ijms 24 00266 g003
Figure 4. 3D depictions of S100P and RAGE interacting residues: (A) S100P residues that interact with RAGE; and (B) residues found on the RAGE V-segment that interact with S100P.
Figure 4. 3D depictions of S100P and RAGE interacting residues: (A) S100P residues that interact with RAGE; and (B) residues found on the RAGE V-segment that interact with S100P.
Ijms 24 00266 g004
Figure 5. RAGE inhibitors such as duloxetine, hispidin, ergothioneine, ethyl pyruvate low-molecular-weight heparins (LMWHs), and papaverine diminish the formation of AGEs, S100B, and HMGB1 and have an impact on downstream signaling (red arrow in downward direction). However, these inhibitors upregulate NF-κB (green arrow upward direction) and downregulate STAT3 (red arrow downward direction) which are primarily involved in the regulation of cancer progression. Moreover, another green arrow indicates the suppression of cancer progression (green arrow in downward direction).
Figure 5. RAGE inhibitors such as duloxetine, hispidin, ergothioneine, ethyl pyruvate low-molecular-weight heparins (LMWHs), and papaverine diminish the formation of AGEs, S100B, and HMGB1 and have an impact on downstream signaling (red arrow in downward direction). However, these inhibitors upregulate NF-κB (green arrow upward direction) and downregulate STAT3 (red arrow downward direction) which are primarily involved in the regulation of cancer progression. Moreover, another green arrow indicates the suppression of cancer progression (green arrow in downward direction).
Ijms 24 00266 g005
Table 1. RAGE–ligand interaction in different malignancies.
Table 1. RAGE–ligand interaction in different malignancies.
S. No.RAGE–Ligand InteractionCancer TypeIn Vitro/In Vivo StudyReferences
1.RAGE–AGEsHuman oral cancer, breast cancer, prostate carcinoma cellsIn vitro[2,3,4,5]
2.RAGE–HMGB1Gastric cancer, colon cancer, breast cancer, melanoma, prostate cancer, hepatocellular carcinomaIn vitro[2,6,7,8,9,10,11]
3.RAGE–S100A4Human colorectal cancer, melanoma, human osteosarcoma, human pancreatic cancer, thyroid cancer (human specimens)In vitro/In vivo[12,13,14,15,16]
4.RAGE–S100A6Nasopharyngeal carcinomaIn vitro[10]
5.RAGE–S100A7Breast cancer,
human cervical cancer
In vitro/In vivo[17,18]
6.RAGE–S100A8Human breast cancer, human prostate cancer, colon tumor, oral-esophageal tumor, squamous cell carcinomaIn vitro/In vivo[19,20,21,22,23]
7.RAGE–S100A9Human hepatocellular carcinomaIn vitro[24]
8.RAGE–S100A14Esophageal squamous
cell carcinoma
In vitro[25]
9.RAGE–S100A16Human prostate cancerIn vitro[26]
10.RAGE–S100BOvarian cancer, glioma (C6), human colon adenocarcinomaIn vitro[27,28,29,30]
11.RAGE–S100PNasopharyngeal carcinoma, colon
cancer, pancreatic cancer,
In vitro/In vivo[16,31,32]
12.RAGE–LPSLiver cancer cells, colorectal cancer, and leukemia cellsIn vitro/In vivo[33,34]
Table 2. RAGE inhibitors are designed for diverse human malignancies.
Table 2. RAGE inhibitors are designed for diverse human malignancies.
S. No.RAGE InhibitorsIn Vitro/In Vivo FindingsRAGE PathwaysStudiesCancer TypesReferences
1.RGBO1In vitro/In vivoAKT, BCL2, and cyclin D1HEC1A, HEC1B, HEC50/Murine xenograft modelEndometrial cancer (EC)[35,36]
2.SorafenibIn vitro/In vivoAMPK/mTOR signalingHepG2, HCCLM3, Huh7, Bel7402/xenografts and orthotopic nude mice modelsHepatocellular carcinoma (HCC)[37]
3.S100B inhibitor /K-Luc (Duloxetine)In vitro/In vivoSTAT3GL261/Murine modelGliomas[38]
4.Sulfated
Glycosaminoglycans
(Chondroitin sulfate (CS), Heparan sulfate (HS))
In vivoRAGE-mediated pathwayMale C57BL/6
mice
Lung metastasis[39,319]
5.S100P-derived inhibitorIn vitro/In vivovia NF-κBC6-glioma, MPanc-96 for pancreatic cancer/Murine modelPancreatic tumors and Glioma tumor growth[33]
6.RAGE inhibitor FPS-ZM1/ PI3K inhibitor LY294002In vitro/In vivoPI3K/AKTSiHa, CaSki, C33A, MS751/xenograft mouse modelCervical squamous cell carcinoma (CSCC)[40]
7.GefitinibIn vitroERK1/2 MAPKGES-1Gastric carcinoma cells[41]
8.Metformin/RAGE-AbIn vitroAMP-activated
protein kinase
MCF-7Breast cancer cells[42]
9.(RAP) Potent LRP1 antagonistIn vivoNF-κB signalingNude micePancreatic tumors, glioma tumor growth[43]
10.CromolynIn vivoNF-κB signalingMale CB17 SCID micePancreatic cancer[31]
11.Ethyl pyruvateIn vitro/In vivoNF-κB/STAT3 signalingA549, H520, and PC-9 cell lines/orthotopic
MM xenograft model
Human malignant mesothelioma/non-small cell lung cancer cells[44]
12.PapaverineIn vitroNF-κB signalingHT1080, U87MG,
and T98G cell lines
Human fibrosarcoma, human glioblastoma[45]
13.Ergothioneine and HispidinIn vitroNF-κB signalingPC12 cell linesRat pheochromocytoma[46]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Faruqui, T.; Khan, M.S.; Akhter, Y.; Khan, S.; Rafi, Z.; Saeed, M.; Han, I.; Choi, E.-H.; Yadav, D.K. RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int. J. Mol. Sci. 2023, 24, 266. https://doi.org/10.3390/ijms24010266

AMA Style

Faruqui T, Khan MS, Akhter Y, Khan S, Rafi Z, Saeed M, Han I, Choi E-H, Yadav DK. RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. International Journal of Molecular Sciences. 2023; 24(1):266. https://doi.org/10.3390/ijms24010266

Chicago/Turabian Style

Faruqui, Tabrez, Mohd Sajid Khan, Yusuf Akhter, Salman Khan, Zeeshan Rafi, Mohd Saeed, Ihn Han, Eun-Ha Choi, and Dharmendra Kumar Yadav. 2023. "RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review" International Journal of Molecular Sciences 24, no. 1: 266. https://doi.org/10.3390/ijms24010266

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop