International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cancer is a paramount societal, public health and economic challenge in the twenty-first century, accounting for nearly 16.8% of global deaths (one in six deaths) and 22.8% of deaths attributable to non-communicable diseases (1). The disease is responsible for 3 in 10 premature deaths from non-communicable diseases worldwide (30.3% among individuals aged 30-69 years), ranking amongst the three principal causes of mortality within this demographic across 177 of 183 nations (2). In the United States in 2025, there will be ~2,041,910 new cancer diagnoses, equating to ~5,600 cases daily (3). Oncological research has predominantly focused on intrinsic characteristics of neoplastic cells, with comparatively limited attention to extracellular stromal components. The extracellular matrix (ECM) constitutes a crucial element of tumor stroma. Alterations in ECM protein expression represent significant mechanisms influencing tumor cell proliferation, migration and invasion, with Fibulin2 serving as a typical protein. Even intracellular modifications in Fibulin2 expression may have a substantial impact on the ECM due to the secretory properties of Fibulin2. Furthermore, Fibulin2 is considered to be a promising biomarker in multi-tumor research contexts (4-7).
Fibulin2 was initially characterized by Pan et al (8), who identified it in a murine fibroblast cDNA library using the Fibulin1 cDNA probe. Fibulin2 is expressed in humans and mice (Fig. 1A and B). In human physiology, Fibulin2 is highly expressed in cardiac tissue, placenta, testes and the urinary bladder, but expressed at low levels in brain tissue, splenic parenchyma and bone marrow (Fig. 1A). Fibulin2 exhibits aberrant expression across malignancies of diverse organs (9), such as colorectal cancer (4) and lung cancer (10). The protein structures of human and murine Fibulin2 exhibit remarkable homology (8,11) (Fig. 2A and B), providing a robust theoretical basis for using murine cellular systems and animal models as surrogates for human studies in oncological research. In the Na, I, II and III domains, the amino acid sequence identity is ~90%, whereas the Nb domain shows only 62% sequence identity (8,11). Human Fibulin2 contains 18 exons (11) (Fig. 3), while mouse Fibulin2 contains 17 exons (8). Surface plasmon resonance and/or solid-phase microplate analyses have shown that Fibulin2 has high binding affinity for Perlecan (12,13), laminin5 (14,15), Fibronectin (16), collagen XVIII (17), Aggrecan, Versican, Brevican lectin domains (18), the C-terminal region V of Perlecan (19), and the short arms of Laminin-5 and Laminin-1 (20), which helps maintain the ECM (21). Consequently, ECM stabilization mediated by Fibulin2 represents a pivotal mechanism underlying tumor suppression.
Nevertheless, Fibulin2 does not universally exert inhibitory effects across all malignancies; rather, it acts as a promoter in certain neoplastic contexts. For instance, increased Fibulin2 expression inhibits cell proliferation in nasopharyngeal carcinoma (22), Kaposi's sarcoma (23) and breast cancer (24). Conversely, suppression of Fibulin2 expression effectively attenuates lung adenocarcinoma progression (10). Therefore, the present review summarizes the diverse roles of Fibulin2 across various malignancies, while examining underlying mechanisms, alongside comprehensive analysis of the potential of Fibulin2 as a biomarker. Regarding the mechanistic influence of Fibulin2 on tumor development and its utility as a tumor marker, several critical issues persist. The mechanisms governing Fibulin2 function across numerous malignancies require further elucidation. The role and mechanistic basis of the effects of Fibulin2 on tumor stroma remain poorly understood. Furthermore, its clinical application as a biomarker is not yet mature. Therefore, the present overview provides guidance for the clinical translation of Fibulin2 as both a therapeutic target and predictive marker in oncological diseases.
Fibulin2 acts as an upstream regulator of the Ras-MEK-ERK1/2 pathway in hepatocellular carcinoma and the TGF-β/ Smad2/TGFB induced factor homeobox 2 (TGIF2) pathway or β-catenin in gastric cancer (25-27). Differences in Fibulin2 expression between tumor tissues and adjacent normal tissues lead to the activation of these pathways (25,26). However, to the best of our knowledge, the factors causing altered Fibulin2 expression in liver and gastric cancer, as well as its upstream regulators, remain unclear. In p53-null immortalized mouse keratinocytes and RasV12-transformed mouse keratinocytes, which are skin cancer models, Fibulin2 is regulated by the upstream molecule Integrin α3β1 (28). Changes in Fbln2 expression in lung adenocarcinoma cells alter the expression of downstream Integrin genes, including Itga1, Itga2, Itga10 and Itgb1 (10). A reciprocal regulatory relationship may exist between Fibulin2 and certain Integrin proteins, which warrants further investigation. In nasopharyngeal carcinoma, Fbln2 acts as an upstream inhibitor of VEGF-165, VEGF-189 and MMP-2. The upstream factors regulating Fbln2 expression in nasopharyngeal carcinoma remain to be elucidated (22). Given that Fibulin2 is a secreted protein, it likely exerts autocrine or paracrine effects on its producing cells or neighboring cells, potentially binding to receptors such as Integrin proteins to establish feedback loops. However, such feedback mechanisms have not received sufficient attention in current tumor-related studies, and further research is needed to demonstrate their existence across various tumor types. In the extracellular space, Fibulin2 interacts with multiple proteins: Integrin β1 in non-small cell lung cancer (29), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5 and ADAMTS-12 in breast cancer (30), Nidogen1 in colorectal cancer (CRC) (31), and mucin 4 (MUC4) in pancreatic cancer (32). These are protein-protein interactions rather than upstream-downstream regulatory relationships (Fig. 4). In breast cancer, ADAMTS-5 cleaves Fibulin2, and this role can be inhibited by ADAMTS-12 (30). Differences in mechanisms of Fibulin2 regulating tumor development across tumor types are shown in Table I.
In the enteric nervous system of patients with colorectal carcinoma, Ndrg4 expression is absent or markedly reduced (33), while enteric nerve cells concurrently secrete higher levels of Fibulin2. Fibulin2 and Nidogen1 promote human colorectal carcinoma cell proliferation and migration. A study substantiated its conclusions by stimulating cells with Fibulin2 and Nidogen1 proteins, then assessing cell proliferation and migration (31). However, the study failed to demonstrate the isolated effects of Fibulin2 on CRC cells and mechanisms (31). In an experiment investigating subcutaneous tumor formation in lung adenocarcinoma, tumors were generated in mice by injecting tumor cells in PBS subcutaneously into the right flank. Tumors formed by Fbln2-knockdown cells exhibited reduced size, weight and tissue consistency compared with those formed by normal cells (10). In vitro experiments revealed that Fbln2-knockdown cells, compared with normal cells, exhibited markedly attenuated type I collagen adhesion capacity alongside markedly reduced tumor cellular pseudopodia (10). Itga1, Itga2, Itga10 and Itgb1 were notably reduced following Fbln2 depletion in lung adenocarcinoma cells (10). These genes encode Integrin proteins α1β1, α2β1, α10β1 and α11β1, respectively, which can bind to collagen (34). In summary, Fibulin2 is closely associated with other ECM proteins in lung adenocarcinoma. Aberrant Fibulin2 expression leads to abnormal changes of other ECM proteins such as integrin proteins and collagen (10). Immunohistochemistry revealed Fibulin2 upregulation in lung cancer tissues compared with adjacent normal tissues. In vitro validation relied exclusively on Fbln2 knockdown. Incorporating overexpression experiments would enhance methodological rigor (10). In p53-null immortalized mouse keratinocytes and RasV12-transformed mouse keratinocytes, knockdown of Fbln2 diminished cell invasion; however, subcutaneous injection of Fbln2-knockdown cells failed to produce a notable reduction in tumor growth volume compared with controls (28). Fibulin2 expression was regulated by Integrin α3β1 and promoted the invasion of immortalized/transformed keratinocytes (28). The research was limited by overly simplistic mechanistic exploration, necessitating further investigation of more specific mechanisms. In mouse non-small cell lung cancer tissue sections, ECM-expressed Fibulin2 interacts with cell membrane-expressed Integrin β1. The interaction between Integrin β1 and Fibulin2 mediates tumor cellular adhesion to the ECM while conferring chemotherapy drug resistance (29). The study employed immunofluorescence double-labeling experiments to demonstrate partial colocalization of Integrin β1 and Fibulin2, subsequently inferring disease mechanisms (29). Additional experiments are necessary, including experiments verifying whether Integrin β1 is a receptor of Fibulin2 and assessing its impact on tumors. In human hepatocellular carcinoma cells, Fibulin2 promotes carcinoma cell proliferation while inhibiting apoptosis through Ras-MEK-ERK1/2 signaling pathway activation (25). In in vitro experiments on liver cancer cells, Fbln2 knockdown and overexpression were performed, and the expression changes of key proteins in the Ras-MEK-ERK1/2 signaling pathway were detected by western blotting (WB) (25). Fibulin2 exerts tumor-promoting effects in both in vitro and in vivo experiments (25). However, this merely establishes an association between Fibulin2 and the Ras-MEK-ERK1/2 signaling pathway rather than a direct regulatory relationship, as indirect mechanisms may exist. Future research could use glutathione S-transferase (GST) pull-down to further verify the direct interactions between Fibulin2 and these pathway proteins. Fibulin2 expression alterations within neoplastic tissues and corresponding modifications in tumor cell proliferation, migration and invasion (demonstrating the pro-carcinogenic properties of Fibulin2) are shown in Table II.
Table IIChanges in Fibulin2 expression in tumor tissue and changes in tumor cell proliferation, migration and invasion (Fibulin2 exerts a pro-carcinogenic effect). |
Fibulin2 is associated with Integrin proteins in mechanistic investigations of lung adenocarcinoma (10), non-small cell lung carcinoma (29), p53-null immortalized murine keratinocytes and RasV12-transformed murine keratinocytes (28). In a hepatocellular carcinoma study, the authors did not study Integrin proteins (25), the Ras-MEK-ERK1/2 signaling pathway constitutes a crucial downstream pathway of Integrin according to previous investigations (35-37). Integrins, as principal ECM cell-surface receptors, represent essential mediators of cellular communication with the tumor microenvironment (TME) (35,38,39). Integrins comprise a family of 24 heterodimeric receptors (40). Integrins consist of two subunits: α and β subunits, which have extensive extracellular domains and short cytoplasmic domains (41,42). These interact with ECM proteins outside the cell membrane while connecting to downstream pathways, including the Ras-MEK-ERK1/2 and FAK/SFK pathways in the cytoplasm (35). When ECM protein, including Fibulin2, Nidogen, Collagen and Laminin, or Integrin expression is altered, fibrosis or remodeling can change tissue stiffness, and downstream Integrin pathways such as the Ras-MEK-ERK1/2 and FAK/SFK pathways may be activated, affecting tumor migration, invasion and proliferation (35,43,44) (Fig. 5).
Most studies examining the tumor-promoting functions of Fibulin2 are simplistic. Future research may include additional experiments to enhance persuasiveness. Furthermore, other factors impact future drug development. For example, studies have used immortalized cell lines such as the HCT116 and Caco-2 human CRC cell lines (31), and mouse models such as nude mice subcutaneously injected with SNU398 cells (25). These models differ substantially from primary cells and the human body, potentially leading to the failure of related drugs in human applications. Inferring disease mechanisms solely from protein spatial associations, exemplified by partial Integrin β1 and Fibulin2 colocalization in lung carcinoma tissue sections (29), may fail to establish causative relationships, resulting in ineffective therapeutic targets. Focusing on single signaling pathways, such as the Ras-MEK-ERK1/2 signaling cascade in hepatocellular carcinoma (25), while disregarding other potential pathways, may lead to compensatory pathway activation and therapeutic resistance after single-target inhibition, thereby limiting the applicability of conclusions. To the best of our knowledge, the impact of changes in Fibulin2 expression on tumor stroma activity [which depends on cancer-associated fibroblasts (CAFs)] has not been investigated. Tumor stromal activity and changes in CAF properties substantially influence tumor development. In advanced-stage malignancies, activated stroma promotes further genetic and epigenetic alterations in carcinoma cells (45-47) while supporting carcinoma progression (48). CAFs are one of the cell types in the TME. CAFs exhibit proliferative, migratory and high secretory activities (49,50). Due to their altered morphology [multispindled rather than single-spindled (51)] and Acta2 and prolyl endopeptidase expression, CAFs are frequently referred to as myofibroblasts (50). CAFs secrete ECM factors, including tenascin, periostin, secreted protein acidic and rich in cysteine, and collagens (51-58). The expression levels of MMPs and other enzymes that degrade and metabolize the ECM are increased in CAFs, enabling cell penetration through the ECM (59-61). CAFs also secrete high levels of growth factors, cytokines and chemokines, promoting intrinsic malignant hallmarks of carcinoma cells through autocrine and paracrine mechanisms (62). In these studies where Fibulin2 had a tumor-promoting effect (10,25,28,31), while Fibulin2 was secreted by tumor cells and acted on the stroma, it remained unclear how the stroma reciprocally affects tumor cells. Whether tumor cell secretion of Fibulin2 impacts CAFs requires further exploration.
Alterations in Fibulin2 expression in tumor tissues and corresponding changes in tumor cell proliferation, migration and invasion (demonstrating the anti-carcinogenic properties of Fibulin2) are summarized in Table III. In human gastric cancer, Fibulin2 expression in malignant tissues is reduced compared with that in adjacent normal tissues, and is negatively associated with β-catenin (26). β-catenin is a bifunctional protein with both cell adhesion and signal transduction activities, and was initially identified through a study of the cell adhesion molecule E-cadherin (63). β-catenin is an essential component of the Wnt/β-catenin signaling pathway, serving a pivotal role in gastric carcinoma pathogenesis and progression (64). A study has established direct or indirect regulatory relationships between the Fibulin family and β-catenin, with both substantially influencing gastric carcinoma development (65). β-catenin is typically localized in the cytoplasm, with its expression suppressed through ubiquitin/proteasome-mediated protein degradation (66). Following construction of Fbln2 overexpression plasmids and transfection of them into AGS and SGC-790 human gastric cancer cell lines, WB analysis has confirmed that Fibulin2 overexpression downregulated β-catenin and its downstream c-myc and cyclin D1, thereby inhibiting the proliferation of gastric cancer cells (26). Immunohistochemistry demonstrated downregulation of Fibulin2 in gastric cancer tissues compared with adjacent normal tissues (26). In vitro validation relied exclusively on Fbln2 overexpression, and incorporation of knockdown experiments would enhance methodological rigor (26). Another study has confirmed that reduced Fibulin2 expression in gastric carcinoma tissues promotes tumor cell proliferation and metastasis through activation of the TGF-β/Smad2/TGIF2 pathway (27). The study used normal and Fbln2-knockdown human gastric cancer cell lines for RNA sequencing, demonstrating upregulation of the TGF-β signaling pathway in Fbln2-knockdown gastric cancer cells (27). Following Fbln2 knockdown and overexpression in gastric carcinoma cells, WB analysis revealed corresponding increases and decreases in Smad2 and TGIF2 phosphorylation (27). Addition of TGF-β signaling pathway inhibitor reversed these expression changes (27). The specific mechanism by which decreased Fbln2 expression in cells alters the activity of the TGF-β pathway requires further exploration. This establishes an association rather than a direct regulatory relationship between Fibulin2 and the TGF-β/Smad2/TGIF2 pathway, as indirect mechanisms cannot be excluded. Future research could use GST pull-down to demonstrate direct interactions between Fibulin2 and pathway proteins.
Table IIIChanges in Fibulin2 expression in tumor tissue and changes in tumor cell proliferation, migration and invasion (Fibulin2 exerts an anti-carcinogenic effect). |
Kaposi's sarcoma, which originates from human vascular endothelial cells, is characterized by vascular proliferation caused by HIV infection of endothelial cells (23). In cutaneous microvascular endothelial cells after 10 days of infection, Fibulin2 protein and mRNA expression are decreased by 50- and 26-fold, respectively. Simultaneously, mRNA levels of the ECM-binding partners fibronectin and tropoelastin are decreased 5- and 25-fold, respectively. This weakens the binding of Fibulin2 to ECM proteins, including fibronectin and tropoelastin, compromising basement membrane (BM) stability and promoting tumor progression (23). Fibronectin is crucial for numerous cell functions, including migration, proliferation and differentiation (67). Transcriptional downregulation of fibronectin has been associated with highly metastatic breast carcinoma cells in murine models (68). Tropoelastin is the soluble precursor of elastin, inducible by ultraviolet irradiation and degradable by MMP-12 (69). Loss of tropoelastin causes developmental tissue disorders, including aneurysms, atherosclerosis and reduced skin elasticity (70). Tropoelastin and fibronectin are essential ECM proteins critical for wound healing (71). The study investigating Kaposi's sarcoma failed to establish causative relationships between Fibulin2 alterations and changes in fibronectin or tropoelastin (23). Future research should include cellular Fbln2 knockdown and overexpression for validation.
In astrocytoma research, U251 cells (a human glioma cell line) exhibited reduced migration and invasion following Fibulin2 overexpression. The conclusion that Fibulin2 inhibits astrocytoma was confirmed by comparing Fbln2-overexpressing U251 cells with negative controls transfected with empty vectors (7). A previous study revealed that across different astrocytoma grades, Fibulin2 inhibits tumor development (7). The authors suggested an ECM-stabilizing function of Fibulin2; however, experimental verification is required (7). During pancreatic carcinoma development, MUC4 protein expression is markedly upregulated on pancreatic carcinoma cell surfaces and MUC4 binds to Fibulin2 (32). This binding interferes with the normal interaction between Fibulin2 and the G1 domain of Nidogen (NIDO), disrupting BM integrity (32) (Fig. 6). Tumor cells consequently breach the BM more readily, facilitating invasion and metastasis. In pancreatic cancer tissue sections, immunofluorescence double-labeling experiments have shown colocalization of MUC4 and Fibulin2 (32). However, immunofluorescence double-labeling experiments merely suggest potential connections between these proteins and do not convincingly demonstrate the roles of MUC4 and Fibulin2 in pancreatic carcinoma (32). Future studies may use conditional knockout of MUC4 and Fibulin2 in murine models for further validation. Additionally, numerous previous articles and reviews have erroneously characterized Fibulin2 as an oncogenic protein in pancreatic carcinoma when citing the conclusions of this study about pancreatic carcinoma. The original study provides no evidence to suggest that Fibulin2 promotes pancreatic carcinoma metastasis and invasion (32).
Breast carcinoma is a prevalent malignancy in women with a high incidence (72). In a breast carcinoma study, transfection of Fbln2 plasmids into human breast carcinoma cell lines reduced tumor cell migration and invasion compared with those of negative control lentiviral vector groups (24). Fibulin2 expression is reduced in breast carcinoma tissues compared with adjacent normal tissues (73). Knockdown of Fbln2 is associated with disruption of the type IV collagen sheath surrounding breast cells in vitro (74); Ibrahim et al (74) hypothesized that its downregulation may be associated with BM disruption and early invasion. However, the BM composition is complex, precluding exclusion of compensation by other Fibulin family members (such as Fibulin1 and Fibulin5) or ECM proteins, which requires demonstration (75). A study involving 272 patients with breast carcinoma from a Norwegian hospital found a positive association between elevated perivascular Fibulin2 expression in breast carcinoma and survival rates (76). Elevated Fibulin2 expression was associated with luminal breast carcinoma, pronounced elastic tissue hyperplasia in the tumor stroma and favorable prognosis, while reduced expression was associated with the basal-like phenotype, triple-negative breast carcinoma, interval breast carcinoma, vascular invasion and poor prognosis (76). The study lacked mechanistic exploration and only reported observed associations.
The TME is currently being investigated as a novel therapeutic target for cancer. ADAMTS is secreted by carcinoma and stromal cells, potentially modifying the TME through various mechanisms, thereby promoting or inhibiting tumors (77). ADAMTS-12 is a secreted metalloproteinase that has functions in tissue remodeling and cell migration or adhesion (78,79). In 293-EBNA cells, yeast two-hybrid screening and co-immunoprecipitation experiments demonstrated that the carboxyl-terminal region of Fibulin2 interacts with the spacer region of ADAMTS-12 (80). When Fibulin2 and ADAMTS-12 were concurrently overexpressed in breast cancer cells, they reduced cell invasion and migration in vitro, inhibited cell migration on relevant matrices, decreased mammosphere unit formation, and suppressed tumor growth in vivo (80). Fibulin2 and ADAMTS-12 expression in breast cancer tissue was negatively associated with histopathological tumor stage, with patients exhibiting the best prognosis when both were highly expressed (80). ADAMTS-4 and ADAMTS-5 are members of the ADAMTS secreted metalloproteinase family (81). A previous study has indicated that both ADAMTS-5 and Fibulin2 are expressed in the stromal and epithelial components of breast cancer tissue, with immunofluorescence double staining showing partial colocalization (30). ADAMTS-4 and ADAMTS-5, predominantly ADAMTS-5, can specifically cleave Fibulin2, enhancing breast carcinoma cell migration and invasion, while increasing the tumorigenic potential (30) (Fig. 7). The study compared the invasion of breast cancer cell lines with simultaneous overexpression of ADAMTS-5 and Fibulin2, breast cancer cell lines with overexpression of ADAMTS-5 or Fibulin2 alone and the control group (30). The invasion capacity of MCF-7 cells was increased by the simultaneous overexpression of Fibulin-2 and ADAMTS-5 compared with the control group and overexpression of Fibulin2 alone (30). However, the invasion capacity of MCF-7 cells was decreased by the simultaneous overexpression of Fibulin-2 and ADAMTS-5 compared with overexpression of ADAMTS-5 alone (30). ADAMTS-5-mediated Fibulin2 degradation is inhibited by ADAMTS-12 (Fig. 7) (30). However, in vivo experiments only showed partial colocalization of ADAMTS-5 or ADAMTS-12 with Fibulin2 via immunofluorescence or immunohistochemistry, which is insufficient to establish their relationship with breast cancer (30). Future research using conditional gene-knockout mice to substantiate the relationship between these proteins and breast cancer would be more compelling. When mammary fibroblasts are cultured in medium containing excess Fibulin2 and ADAMTS-5, the expression levels of α-smooth muscle actin (α-SMA) increase. Stimulation with excess Fibulin2 alone can also increase α-SMA expression levels, as observed by WB (the authors did not conduct statistical analysis), indicating that Fibulin2 promotes breast fibroblast activation (30). Breast fibroblasts are a key component of the tumor stroma (82). This indicates that changes in Fibulin2 expression in breast cancer cells can activate stromal fibroblasts and potentially drive their differentiation into CAFs, a phenomenon closely related to the secretory properties of Fibulin2 (30). Similar mechanisms may occur in other tumor types; however, they have received limited attention to date and warrant further investigation.
Due to the opposing roles of different ADAMTS subtypes in breast cancer, developing highly specific inhibitors that selectively block the tumor-promoting activities of ADAMTS subtypes without affecting their potential tumor-suppressive effects remains challenging. Small-molecule inhibitors targeting aggrecanases, including ADAMTS-4 and ADAMTS-5, have advanced to a phase III clinical trial for orthopedic applications (83). This provides valuable insights for the development of ADAMTS-targeted therapeutics for breast cancer treatment.
Cancer progression is accompanied by uncontrolled tumor growth, local invasion and metastasis; processes that depend heavily on the proteolytic activities of multiple MMPs. These enzymes affect tissue integrity by degrading ECM components such as Fibulin2 (84). In a nasopharyngeal carcinoma study involving samples from 30 patients, Fbln2 levels in carcinoma tissues substantially exceeded those in normal tissues, as detected by gene chip technology (22). In nasopharyngeal cancer cell lines, overexpression of Fbln2s, which encodes the short isoform of Fibulin2, downregulated the expression levels of VEGF-165, VEGF-189 and MMP-2 compared with those in the control group (22). Sustained angiogenesis is indispensable for both tumor growth and metastasis (85). MMP-2 is an effective gelatinase capable of cleaving protein components of the ECM and is involved in the invasion and metastasis process of tumor cells (86). Evidence has indicated that Fibulin2 is cleaved by MMP-2 (87), and inhibits nasopharyngeal carcinoma cell migration and invasion by strictly regulating MMP-2 expression (22). However, the study could not establish a direct regulatory relationship between Fbln2 and VEGF-165, VEGF-189 or MMP-2, because there may be unknown molecules or signaling pathways mediating the interaction between Fbln2 and VEGF-165, VEGF-189 or MMP-2. WB analysis of osteosarcoma cell line lysates has indicated multiple Fibulin2 fragments (87). Gelatinase MMP-2 facilitates Fibulin2 degradation through MMP-dependent mechanisms in osteosarcoma cell lines (87). Following addition of an MMP-2 inhibitor to the cell culture medium, WB indicated no change in the cleaved Fibulin2 fragments, indicating that unidentified mechanisms require exploration (87). This study using an osteosarcoma cell line relied entirely on in vitro experiments, and in vivo experiments are required for verification, as the in vivo environment is more complex than the in vitro cell culture environment (87).
Most documented MMP inhibitors exhibit non-specific binding and have diminished efficacy, attributable to their pronounced sequence homology with other MMPs (88). To date, no effective MMP inhibitor has successfully completed clinical trials and secured regulatory approval from the Food and Drug Administration for tumor treatment (84,88).
Studies on the tumor-inhibitory effect of Fibulin2 predominantly focus on preserving BM structural integrity (7,32,74). Few studies have investigated the mechanisms of interaction between Fibulin2 and classical signaling pathways. Only the association among Fibulin2, β-catenin and the TGF-β/Smad2/TGIF2 pathways has been demonstrated in gastric cancer studies (26,27). An intricate association exists between the β-catenin and TGF-β signaling pathways and the aforementioned Integrins (89,90). Whether Fibulin2 indirectly regulates the β-catenin and TGF-β/Smad2/TGIF2 pathways through Integrins warrants comprehensive investigation. Some methodological considerations profoundly impact future pharmaceutical development efforts. For example, the use of established cell lines, such as the HGC27 and MKN28 cell lines, to study gastric cancer fails to replicate the complexity of primary cells isolated from living tissues (27). Inferring causal relationships in disease mechanisms based solely on gene or protein expression associations or simultaneous alterations, exemplified by MUC4 and Fibulin2 in pancreatic cancer (32), and simultaneous changes in Fibulin2, fibronectin and tropoelastin in Kaposi's sarcoma (23), may result in ineffective drug targets. Focusing on signaling pathways, such as the TGF-β/TGIF2 pathway in gastric cancer (27), while disregarding other potential pathways, may lead to therapeutic resistance due to the activation of compensatory pathways following single-target inhibition. The role of tumor stroma in inhibiting or promoting tumors is associated not only with signaling pathway activation but is also closely associated with stromal cells, such as the activity or senescence state of fibroblasts (91). Secretion of growth factors with inhibitory functions by non-activated fibroblasts (92) or upregulation of the tissue inhibitor of metalloproteinases (TIMP) family are potential mechanisms by which tumor stroma suppresses neoplastic progression (93,94). Notably, both MMPs and the previously discussed ADAMTS are strictly regulated by TIMPs in normal tissues (95,96). Expression of TIMP family proteins in fibroblasts governs ECM structural organization and stromal cell architecture (97). These proteins function as endogenous negative regulators of MMP activity, and numerous malignancies exhibit aberrant TIMP and/or MMP expression patterns (93,98-99). Loss or reduction of TIMP expression leads to enhanced MMP functionality, facilitating stromal activation and subsequent tumor progression (100,101). TIMP overexpression attenuates tumorigenesis, growth, angiogenesis and metastasis in some cancer types, such as pancreatic cancer (94). Whether altered Fibulin2 expression in tumor cells influences fibroblast activity and stromal TIMP family expression requires further elucidation.
Studies on Fibulin2 as a biomarker are summarized in Table IV. For case-control studies involving human populations in Table IV, according to the 2011 Evidence Hierarchy of the Oxford Centre for Evidence-based Medicine, the evidence level of most studies was 4 (102).
In breast cancer mouse models, Fibulin2 expression in plasma was substantially higher than that in normal mice, indicating its potential as a plasma biomarker (103). The study used mice rather than humans as research subjects. Due to species differences, the effectiveness of the marker in humans requires verification, limiting the applicability of the conclusions. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and label-free quantitative methods, differences in Fibulin2 expression have been detected in the sera of patients with advanced colon cancer and healthy individuals. Fibulin2 is a diagnostic marker for advanced colon cancer (4). The sample sizes for the patient and normal control groups were only 8 and 10, respectively, in the study investigating advanced colon cancer, representing inadequate sample sizes (4). Consequently, the cohort lacks representativeness, predisposing it to false positives or false negatives. Future research should expand the sample size to enhance the robustness of the conclusions. In a study on patients with lung cancer, Fbln2 gene expression levels of the enriched epithelial cells of peripheral blood lymphocytes were found to be decreased ~2-fold in patients with metastatic and non-metastatic lung cancer compared with healthy controls (104). Fbln2 is a potential biomarker to distinguish patients with lung cancer from healthy controls (104).
Surgical specimens of astrocytomas have been analyzed using LC-MS/MS, WB and reverse transcription-quantitative PCR, revealing that Fibulin2 was downregulated in grade II/III/IV astrocytomas compared with grade I astrocytomas, with negative expression being associated with advanced clinical stages (7). The sample sizes of patients with different grades of glioma were only 5 each, representing small sample sizes that may reduce diagnostic accuracy. Fibulin2 expression in the plasma of patients with grade II meningioma is higher than that in grade I patients, indicating that Fibulin2 is a potential marker to distinguish between patients with grade II and I meningioma (5,6).
In studies of Fibulin2 as a marker to predict the prognosis of patients (74,105), comprehensive analysis of human sample data from multiple databases yielded information on patients with breast cancer, including their stages, molecular subtypes and treatment conditions. Based on the Kaplan-Meier plotter dataset, after chemotherapy, high Fbln2 expression was associated with improved overall survival in Her2− patients. In grade 2 patients (including those after chemotherapy/hormonal therapy), unstratified patients with breast cancer, Her2− patients, Luminal− patients and estrogen receptor+ patients after chemotherapy, high Fbln2 expression was associated with improved recurrence-free survival. Conversely, in grade 3 patients, low Fbln2 expression was associated with improved overall survival. In Her2+ patients, estrogen receptor− patients and grade 3 patients, low Fbln2 expression was associated with improved recurrence-free survival. Analysis of sample data from patients with breast cancer with different molecular subtypes in The Cancer Genome Atlas dataset revealed that in Luminal B patients, low Fbln2 expression was associated with improved overall survival. In Her2+ patients, high Fbln2 expression was associated with improved overall survival (105). Another study used the Kaplan-Meier plotter dataset to collect the mRNA expression and survival data of patients with breast cancer, analyzing the association between Fbln2 expression and prognosis in different subgroups (74). In patients with lymph node-negative and intermediate-grade breast cancer, high Fbln2 mRNA expression was associated with improved distant metastasis-free survival compared with that of patients with low expression. By contrast, in patients with high-grade breast cancer, the opposite was true: Elevated Fbln2 mRNA expression was associated with poorer prognosis than low expression (74). The conclusions of both studies were derived from database analyses (74,105). However, samples from public databases are prone to heterogeneity, so clinical validation of their findings is necessary. In a study analyzing human meningioma tissue samples, high-grade histopathology was associated with elevated Ki-67 and Fibulin2 expression. The higher the histological grade of meningioma was based on histopathology, the poorer the prognosis of patients. Higher grades were associated with increased risks of recurrence, progression and mortality. The younger age group (<50 years) exhibited higher Fibulin2 expression than the older age group (>50 years). Fibulin2 expression exhibited an association with age and histopathological grading (106). The sample sizes for low-grade and high-grade gliomas were both 25, representing relatively small cohorts. Furthermore, as the study was a cross-sectional study, it could not determine whether elevated Fibulin2 expression was the cause or consequence of increased meningioma grading.
In a study examining patients with hypertrophic cardiomyopathy (HCM), serum Fibulin2 expression in patients with HCM markedly exceeded that of normal individuals, suggesting that Fibulin2 may be beneficial for HCM diagnosis (107). This retrospective study included 95 consecutive patients with obstructive HCM eligible for septal myectomy surgery. However, HCM has numerous subtypes, with sampling limited to specific subtypes, introducing bias that affects the generalizability of the conclusions to HCM overall. In a study examining patients with infections, plasma Fibulin2 expression in patients with infections was higher than that in normal individuals (108). All patients in the study were from the emergency department, and the disease types included were limited, leading to selection bias in the study population. The applicability of the conclusions supporting Fibulin2 as a clinical diagnostic marker for infection remains limited. Immunohistochemical expression of Fibulin2 distinguishes liver stellate cells from liver myofibroblasts after liver injury, suggesting that liver myofibroblasts are a unique cell population involved in matrix production during liver fibrosis caused by chronic liver injury (109). Differences in Fibulin2 expression have been observed between liver stellate cells and liver myofibroblasts in normal livers, livers with acute damage and livers with chronic damage (109). This study provides a novel marker for researchers to label liver myofibroblasts in related research, although its application remains limited to basic research. Future research should explore whether Fibulin2 can be translated into a clinical diagnostic marker for liver-related diseases based on this finding.
Although Fibulin2 shows considerable potential as a therapeutic target and biomarker, several challenges and limitations remain. Addressing these impediments is crucial for successful clinical translation.
Currently, studies on Fibulin2 receptors remain limited. In infected bone marrow mesenchymal stem cells, Fibulin2 binds to the transmembrane receptor Notch2 (110). In a rat model of neuropathic pain, Fibulin2 expression in the spinal dorsal horn was upregulated (111). Fibulin2 specifically binds to the B1a subunit of γ-aminobutyric acid B receptor, inhibiting its activity and thereby exacerbating pain (111). Following multiple sclerosis, Fibulin2 suppresses oligodendrocyte generation through Notch pathway activation (112). In tumor research, Integrin proteins have been demonstrated to be receptors of Fibulin2 (29). The mechanistic evidence in numerous studies exhibits oversimplification (10,22,28,29). These studies predominantly involve Fbln2 knockdown or overexpression, followed by detection of expression changes in specific key proteins in vitro. Other studies primarily encompass influencing the alteration of the BM (7,32,74). The impact of Fibulin2 on neoplastic processes is complex, requiring further investigation into more comprehensive mechanistic pathways. This would facilitate the design of precision-targeted pharmaceuticals against disease-associated molecular targets.
Due to the complexity of the function of Fibulin2, targeting Fibulin2 may induce various adverse effects and toxicities. Beyond its role in tumors, Fibulin2 serves a pivotal role in maintaining tissue integrity (74,75,113), regulating fibrosis and immunity (114-120), and supporting tissue development and repair (121-126). For example, Fibulin2-deficient mouse pups developed blisters, demonstrating that Fibulin2 deficiency during development could cause BM rupture. However, adult Fibulin2-deficient mice exhibited no blisters, likely due to compensatory mechanisms mediated by other ECM components (75,113). Reduced Fbln2 expression disrupts sheath formation in the mammary epithelium and downregulates Integrin β1 expression, compromising BM integrity. Fibulin2 serves a crucial role in stabilizing the BM structure of the mammary epithelium (74). The Fibulin2 protein exhibits close associations with fibrosis. By establishing cardiac hypertrophy models in Fibulin2 homozygous and wild-type mice via angiotensin II infusion, Fibulin2 knockout has been shown to inhibit myocardial fibrosis via H&E staining, Masson staining and immunohistochemistry analysis (114). This finding was corroborated by observations that Fbln2 reduced fibrosis markers in primary cardiomyocyte fibroblasts from Fibulin2 homozygous and wild-type mice stimulated with TGF-β1 (114). Consistent results were obtained when establishing myocardial infarction models in Fibulin2 homozygous and wild-type mice: The survival rate of Fibulin2 homozygous mice was higher than that of wild-type mice (115). A 5-day mouse model of skin injury revealed markedly elevated Fibulin2 expression in mice with skin injuries compared with normal mice, as detected by nucleic acid analysis and immunofluorescence (116). Unilateral ureteral obstruction leads to renal fibrosis. Immunofluorescence staining in mice after 7 days of unilateral ureteral obstruction showed higher Fibulin2 expression compared with that in normal mice (117). After establishing a liver fibrosis model in rats, immunofluorescence revealed higher Fibulin2 expression in rats with liver fibrosis compared with normal rats (118). Fibulin2 is upregulated in patients with idiopathic pulmonary fibrosis (119). Fibulin2 inhibition suppresses α-SMA, collagen type Iα1 and fibronectin expression in human lung fibroblast-derived MRC-5 cells (119). Fibulin2 influences immune function, with reduced levels being associated with immune impairment after bone trauma (120). In terms of tissue development and repair, Fibulin2 is a key mediator of the neurogenic effect of TGF-β1 on adult neural stem cells (121). Fibulin2-mediated TGF-β signaling in astrocyte extracellular vesicles promotes synapse formation (122). During myoblast differentiation, Fbln2 expression is upregulated, and Fbln2 is indispensable for myoblast differentiation (123-125). Fibulin2 regulates smooth muscle cell migration during blood vessel wall repair (126).
Consequently, the functions of Fibulin2 exhibit extraordinary complexity. Although targeting of Fibulin2 has therapeutic potential in tumor treatment, it may cause immunosuppression (120), disrupt the balance between ECM production and degradation, and induce severe complications such as excessive collagen deposition and fibrosis (114-119).
To the best of our knowledge, currently, there are no clinical studies on Fibulin2 as a therapeutic target. Existing clinical studies related to Fibulin2 only focus on its use as a marker (5,6,7,107,108). The clinical translation of Fibulin2-targeted drugs for cancer treatment faces numerous obstacles, although current research has not yet reached this stage. Beyond the aforementioned incomplete understanding of mechanisms, there are substantial challenges in clinical drug trials themselves. The following issues are similarly prevalent in the research and development of oncology drugs. For example, determining the optimal dosing and regimens for targeted drugs is challenging due to their narrow therapeutic window and potential toxicity (127). Careful dose escalation studies and monitoring of adverse reactions are necessary. Additionally, for combination therapies involving targeted drugs and other anticancer drugs, synergistic dose regimens need to be determined. Establishing appropriate clinical trial endpoints and efficacy measures is quite complex, as traditional endpoints such as overall survival and progression-free survival may inadequately reflect the therapeutic effects and potential long-term implications of targeted drugs (128). Determining the optimal combination, sequence and potential interactions between treatments is challenging. Extensive preclinical and clinical studies are necessary to evaluate synergistic effects, additional benefits or potential antagonistic interactions of treatments to maximize therapeutic efficacy.
Biomarkers should meet strict criteria to ensure concordance between measured and actual physiological values, including accuracy, precision, sensitivity, reproducibility, stability (129,130), specificity, dynamics, detectability and minimal invasiveness (131). Fibulin2 exhibits significant expression differences across distinct stages of tumor development and between normal and diseased conditions (4,5,6,7,103). Fibulin2 is also a potentially effective biomarker in studies of infection and fibrosis (108,109). However, numerous studies are limited by inadequate sample sizes. Furthermore, Fibulin2 can serve as a marker in studies on different diseases, and thus, does not meet the specificity criteria for a marker. Its low specificity fundamentally hinders the clinical utility of Fibulin2 as a single biomarker. In breast cancer, meningioma, infection and HCM, Fibulin2 in human serum samples is detected using ELISA (5,103,107,108). ELISA has considerable advantages, including high sensitivity, high specificity (132) and the ability to analyze multiple samples simultaneously within short timeframes (133). This ensures practical feasibility and convenience for large-scale screening programs and facilitates high-throughput sample processing (133). However, its inherent limitations include lengthy sample pretreatment and purification procedures, unsuitability for rapid detection, high cost, lack of real-time detection capabilities (134), and the need for a relatively large sample volume (100-200 µl) (135). As a biomarker, Fibulin2 should not merely aid in staging disease progression or pathological exclusion but also provide therapeutic guidance. This requires that Fibulin2 demonstrates systematic changes corresponding to dynamic disease progression. Most studies so far have not conducted comprehensive dynamic monitoring.
At present, there is a lack of pharmaceutical investigations targeting Fibulin2 for tumor treatment. A key contributing factor to this scarcity is the multifaceted complexity of its biological functions as aforementioned. Fibulin2-directed therapeutic interventions may induce considerable toxicity and adverse sequelae (74,75,113-126). Next-generation small-molecule inhibitors, novel monoclonal antibodies, ligand traps, gene editing, RNA interference technologies, nanomaterials and peptide-based therapies are emerging targeted treatments in cancer. Beyond the need to further elucidate the mechanistic basis of the role of Fibulin2 in tumors, the strategic application of these emerging technologies in prospective Fibulin2-targed drugs will prove instrumental in enhancing therapeutic specificity and potency while reducing adverse effects.
Integrating Fibulin2-targeted treatment with other modalities is a strategic approach to enhance antitumor efficacy and circumvent resistance mechanisms. For example, synergistic combinations with immunotherapeutic agents, particularly immune checkpoint inhibitors (such as anti-programmed cell death protein 1 and anti-cytotoxic T-lymphocyte-associated protein 4), show promise by reactivating antitumor immune responses by reducing immunosuppression (136,137). Combining Fibulin2-targeted treatment with adoptive cell therapy, such as chimeric antigen receptor-T cells, may enhance cellular persistence and effectiveness (138). Co-administering signaling pathway inhibitors with Fibulin2-targeted treatment helps attenuate parallel signaling pathways, thereby blocking the compensatory mechanisms exploited by malignant cells. This strategy aims to enhance comprehensive antitumor activity and delay drug resistance. Integrating Fibulin2-targeted treatment with traditional chemotherapy and radiotherapy increases tumor susceptibility to these treatments. Concomitant use of epigenetic modulators can enhance therapeutic responsiveness by systematically reprogramming the TME, which is effective in addressing epigenetic changes that promote cancer progression (139).
Given the limited specificity of Fibulin2 as a single biomarker, integrating multiple biomarkers is advantageous to address this limitation. While there are currently no reports on the use of Fibulin2 in combination with other markers for tumor detection, the use of microRNAs (miRNAs) and other proteins as combined markers for early-stage cancer diagnosis has demonstrated the considerable potential of combined marker detection, offering valuable insights for combining Fibulin2 with other markers (140,141). For example, Yu et al (140) described a multi-marker diagnostic method for early-stage hepatocellular carcinoma, using α fetoprotein and miRNA-125b as combined markers to simultaneously improve diagnostic sensitivity and specificity. Yuan et al (141) proposed a new combination of circulating miRNAs and plasma protein biomarkers for pancreatic cancer diagnosis. These combined indicators exhibited higher specificity in distinguishing pancreatic cancer from other gastrointestinal cancers than CA19-9 and individual indicators (141). Combining Fibulin2 with other markers such as miRNAs for early cancer screening, diagnosis and prognosis may enhance specificity compared with using Fibulin2 alone. The introduction of artificial intelligence (AI) and machine learning can facilitate the identification of novel potential combined biomarkers. AI algorithms excel in analyzing and identifying unique combinations of gene mutations and using large-scale genomic databases to identify cancer-specific markers (142).
The quantification of Fibulin2 in serum and plasma is performed using traditional ELISA. ELISA detection requires air-conditioned laboratories, refrigeration facilities for chemicals and reagents, a reliable power supply, precision-calibrated equipment, and highly trained personnel (135). Some laboratories or hospitals still lack access to affordable infrastructure for these complex diagnostic tests. To address these deficiencies, laboratories worldwide have evaluated and implemented various modified forms of ELISA. These innovative ELISA formats have greater potential for clinical translation due to their low cost, short detection time, portability and reduced reagent needs (143-146). For example, microfluidic-based ELISA, paper-ELISA and aptamer-ELISA can mitigate the limitations of traditional ELISA to some extent (143-146).
The oncogenic or suppressive effects of altered Fibulin2 expression in malignant cells vary markedly across diverse tumors. The contributing factors include distinct mechanisms, including the activation of different signaling pathways and the complex relationships with BM or other ECM proteins, tumor development stage, species and tumor origin. Although current mechanistic studies help clarify aspects of pathogenic processes, several outstanding issues remain to be resolved. These include validating the upstream and downstream molecules of Fibulin2, confirming the existence of feedback loops, exploring alternative pathways, and clarifying the relationship between Fibulin2 and stromal fibroblasts. These considerations are crucial to improve the safety and effectiveness of future relevant drug research. The use of Fibulin2 alone as a marker for tumor diagnosis or staging remains not well-established. Future research should further explore methods to enhance specificity, such as identifying novel and specific markers to be used in combination with Fibulin2 as a dual-marker system.
Not applicable.
YY completed most of the work, wrote the original draft and created illustrations. ZW wrote and revised the article, and collected the literature. LW revised the article and collected the literature. JF reviewed and edited the manuscript, provided resources, and acquired funding. ZL supervised the study, edited and reviewed the manuscript, was involved in project administration, and acquired funding. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
TME |
tumor microenvironment |
|
CAF |
cancer-associated fibroblast |
|
ECM |
extracellular matrix |
|
BM |
basement membrane |
|
TIMP |
tissue inhibitor of metalloproteinases |
|
WB |
western blotting |
|
ADAMTS |
a disintegrin and metalloproteinase with thrombospondin motifs |
|
AI |
artificial intelligence |
|
HCM |
hypertrophic cardiomyopathy |
|
CRC |
colorectal cancer |
Not applicable.
The present study was supported by grants from the Key Research and Development Program of Sichuan Province (grant no. 2023YFQ0009), the Chongqing Municipality Science-Health Joint Traditional Chinese Medicine Research Project (grant no. 2023DBXM010), and the Sichuan Provincial Central Government-Guided Local Science and Technology Development Special Project (grant no. 2023ZYD0288).
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Bray F, Laversanne M, Weiderpass E and Soerjomataram I: The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 127:3029–3030. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Kratzer TB, Giaquinto AN, Sung H and Jemal A: Cancer statistics, 2025. CA Cancer J Clin. 75:10–45. 2025.PubMed/NCBI | |
|
Takakura D, Ohashi S, Kobayashi N, Tokuhisa M, Ichikawa Y and Kawasaki N: Targeted O-glycoproteomics for the development of diagnostic markers for advanced colorectal cancer. Front Oncol. 13:11049362023. View Article : Google Scholar : PubMed/NCBI | |
|
Sofela AA, Hilton DA, Ammoun S, Baiz D, Adams CL, Ercolano E, Jenkinson MD, Kurian KM, Teo M, Whitfield PC, et al: Fibulin-2: A novel biomarker for differentiating grade II from grade I meningiomas. Int J Mol Sci. 22:5602021. View Article : Google Scholar : PubMed/NCBI | |
|
Sofela AA, McGavin L, Whitfield PC and Hanemann CO: Biomarkers for differentiating grade II meningiomas from grade I: A systematic review. Br J Neurosurg. 35:696–702. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ren T, Lin S, Wang Z and Shang A: Differential proteomics analysis of low- and high-grade of astrocytoma using iTRAQ quantification. Onco Targets Ther. 9:5883–5895. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Pan TC, Sasaki T, Zhang RZ, Fässler R, Timpl R and Chu ML: Structure and expression of fibulin-2, a novel extracellular matrix protein with multiple EGF-like repeats and consensus motifs for calcium binding. J Cell Biol. 123:1269–1277. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Hui D and Fu X: Roles of Fibulin-2 in carcinogenesis. Med Sci Monit. 26:e9180992020.PubMed/NCBI | |
|
Baird BN, Schliekelman MJ, Ahn YH, Chen Y, Roybal JD, Gill BJ, Mishra DK, Erez B, O'Reilly M, Yang Y, et al: Fibulin-2 is a driver of malignant progression in lung adenocarcinoma. PLoS One. 8:e670542013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang RZ, Pan TC, Zhang ZY, Mattei MG, Timpl R and Chu ML: Fibulin-2 (FBLN2): Human cDNA sequence, mRNA expression, and mapping of the gene on human and mouse chromosomes. Genomics. 22:425–430. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Brown JC, Sasaki T, Göhring W, Yamada Y and Timpl R: The C-terminal domain V of perlecan promotes beta1 integrin-mediated cell adhesion, binds heparin, nidogen and fibulin-2 and can be modified by glycosaminoglycans. Eur J Biochem. 250:39–46. 1997. View Article : Google Scholar | |
|
Hopf M, Göhring W, Kohfeldt E, Yamada Y and Timpl R: Recombinant domain IV of perlecan binds to nidogens, laminin-nidogen complex, fibronectin, fibulin-2 and heparin. Eur J Biochem. 259:917–925. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Sasaki T, Göhring W, Mann K, Brakebusch C, Yamada Y, Fässler R and Timpl R: Short arm region of laminin-5 gamma2 chain: Structure, mechanism of processing and binding to heparin and proteins. J Mol Biol. 314:751–763. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Talts JF, Andac Z, Göhring W, Brancaccio A and Timpl R: Binding of the G domains of laminin alpha1 and alpha2 chains and perlecan to heparin, sulfatides, alpha-dystroglycan and several extracellular matrix proteins. EMBO J. 18:863–870. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Sasaki T, Wiedemann H, Matzner M, Chu ML and Timpl R: Expression of fibulin-2 by fibroblasts and deposition with Fibronectin into a fibrillar matrix. J Cell Sci. 109(Pt 12): 2895–2904. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Sasaki T, Fukai N, Mann K, Göhring W, Olsen BR and Timpl R: Structure, function and tissue forms of the C-terminal globular domain of collagen XVIII containing the angiogenesis inhibitor endostatin. EMBO J. 17:4249–4256. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Olin AI, Mörgelin M, Sasaki T, Timpl R, Heinegård D and Aspberg A: The proteoglycans aggrecan and Versican form networks with fibulin-2 through their lectin domain binding. J Biol Chem. 276:1253–1261. 2001. View Article : Google Scholar | |
|
Friedrich MV, Göhring W, Mörgelin M, Brancaccio A, David G and Timpl R: Structural basis of glycosaminoglycan modification and of heterotypic interactions of perlecan domain V. J Mol Biol. 294:259–270. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Utani A, Nomizu M and Yamada Y: Fibulin-2 binds to the short arms of laminin-5 and laminin-1 via conserved amino acid sequences. J Biol Chem. 272:2814–2820. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
de Vega S, Iwamoto T and Yamada Y: Fibulins: Multiple roles in matrix structures and tissue functions. Cell Mol Life Sci. 66:1890–1902. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Law EW, Cheung AK, Kashuba VI, Pavlova TV, Zabarovsky ER, Lung HL, Cheng Y, Chua D, Kwong DLK, Tsao SW, et al: Anti-angiogenic and tumor-suppressive roles of candidate tumor-suppressor gene, Fibulin-2, in nasopharyngeal carcinoma. Oncogene. 31:728–738. 2012. View Article : Google Scholar | |
|
Alcendor DJ, Knobel S, Desai P, Zhu WQ and Hayward GS: KSHV regulation of fibulin-2 in Kaposi's sarcoma: Implications for tumorigenesis. Am J Pathol. 179:1443–1454. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Duan L, Zhang Y, Zhao H, Yang X and Zhang C: Correlation of Fibulin-2 expression with proliferation, migration and invasion of breast cancer cells. Oncol Lett. 20:1945–1951. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hu X, Liu T, Li L, Gan H, Wang T, Pang P and Mao J: Fibulin-2 facilitates malignant progression of hepatocellular carcinoma. Turk J Gastroenterol. 34:635–644. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma H, Lian C and Song Y: Fibulin-2 inhibits development of gastric cancer by downregulating β-catenin. Oncol Lett. 18:2799–2804. 2019.PubMed/NCBI | |
|
Zhou M, Mao X, Shen K, Zhan Q, Ni H, Liu C, Huang Z and Li R: FBLN2 inhibits gastric cancer proliferation and metastasis via the TGFβ/TGIF2 pathway. Pathol Res Pract. 269:1558992025. View Article : Google Scholar | |
|
Missan DS, Chittur SV and DiPersio CM: Regulation of fibulin-2 gene expression by integrin α3β1 contributes to the invasive phenotype of transformed keratinocytes. J Invest Dermatol. 134:2418–2427. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang MR, Chen RJ, Zhao F, Zhang HH, Bi QY, Zhang YN, Zhang YQ, Wu ZC and Ji XM: Effect of Wenxia Changfu formula combined with cisplatin reversing non-small cell lung cancer cell adhesion-mediated drug resistance. Front Pharmacol. 11:5001372020. View Article : Google Scholar : PubMed/NCBI | |
|
Fontanil T, Álvarez-Teijeiro S, Villaronga MÁ, Mohamedi Y, Solares L, Moncada-Pazos A, Vega JA, García-Suárez O, Pérez-Basterrechea M, García-Pedrero JM, et al: Cleavage of Fibulin-2 by the aggrecanases ADAMTS-4 and ADAMTS-5 contributes to the tumorigenic potential of breast cancer cells. Oncotarget. 8:13716–13729. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Vaes N, Schonkeren SL, Rademakers G, Holland AM, Koch A, Gijbels MJ, Keulers TG, de Wit M, Moonen L, Van der Meer JRM, et al: Loss of enteric neuronal Ndrg4 promotes colorectal cancer via increased release of Nid1 and Fbln2. EMBO Rep. 22:e519132021. View Article : Google Scholar : PubMed/NCBI | |
|
Senapati S, Gnanapragassam VS, Moniaux N, Momi N and Batra SK: Role of MUC4-NIDO domain in the MUC4-mediated metastasis of pancreatic cancer cells. Oncogene. 31:3346–3356. 2012. View Article : Google Scholar : | |
|
Melotte V, Lentjes MH, van den Bosch SM, Hellebrekers DM, de Hoon JP, Wouters KA, Daenen KL, Partouns-Hendriks IE, Stessels F, Louwagie J, et al: N-Myc downstream-regulated gene 4 (NDRG4): A candidate tumor suppressor gene and potential biomarker for colorectal cancer. J Natl Cancer Inst. 101:916–927. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Barczyk M, Carracedo S and Gullberg D: Integrins. Cell Tissue Res. 339:269–280. 2010. View Article : Google Scholar | |
|
Cooper J and Giancotti FG: Integrin signaling in cancer: Mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell. 35:347–367. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Nobre LT, Vidal AA, Almeida-Lima J, Oliveira RM, Paredes-Gamero EJ, Medeiros VP, Trindade ES, Franco CR, Nader HB and Rocha HA: Fucan effect on CHO cell proliferation and migration. Carbohydr Polym. 98:224–232. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fischer C, Sanchez-Ruderisch H, Welzel M, Wiedenmann B, Sakai T, André S, Gabius HJ, Khachigian L, Detjen KM and Rosewicz S: Galectin-1 interacts with the {alpha}5{beta}1 fibronectin receptor to restrict carcinoma cell growth via induction of p21 and p27. J Biol Chem. 280:37266–37277. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Hynes RO: Integrins: Bidirectional, allosteric signaling machines. Cell. 110:673–687. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Hamidi H and Ivaska J: Every step of the way: Integrins in cancer progression and metastasis. Nat Rev Cancer. 18:533–548. 2019. View Article : Google Scholar : | |
|
Humphries JD, Byron A and Humphries MJ: Integrin ligands at a glance. J Cell Sci. 119:3901–3903. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Springer TA and Wang JH: The three-dimensional structure of integrins and their ligands, and sconformational regulation of cell adhesion. Adv Protein Chem. 68:29–63. 2004. View Article : Google Scholar | |
|
Arnaout MA, Mahalingam B and Xiong JP: Integrin structure, allostery, and bidirectional signaling. Annu Rev Cell Dev Biol. 21:381–410. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Erler JT, Bennewith KL, Nicolau M, Dornhofer N, Kong C, Le QT, Chi JT, Jeffrey SS and Giaccia AJ: Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 440:1222–1226. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, et al: Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 139:891–906. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Couillard J, Demers M, Lavoie G and St-Pierre Y: The role of DNA hypomethylation in the control of stromelysin gene expression. Biochem Biophys Res Commun. 342:1233–1239. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Hanson JA, Gillespie JW, Grover A, Tangrea MA, Chuaqui RF, Emmert-Buck MR, Tangrea JA, Libutti SK, Linehan WM and Woodson KG: Gene promoter methylation in prostate tumor-associated stromal cells. J Natl Cancer Inst. 98:255–261. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Lin HJ, Zuo T, Lin CH, Kuo CT, Liyanarachchi S, Sun S, Shen R, Deatherage DE, Potter D, Asamoto L, et al: Breast cancer-associated fibroblasts confer AKT1-mediated epigenetic silencing of Cystatin M in epithelial cells. Cancer Res. 68:10257–10266. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Hanahan D and Coussens LM: Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 21:309–322. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Ostman A and Augsten M: Cancer-associated fibroblasts and tumor growth-bystanders turning into key players. Curr Opin Genet Dev. 19:67–73. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Micallef L, Vedrenne N, Billet F, Coulomb B, Darby IA and Desmoulière A: The myofibroblast, multiple origins for major roles in normal and pathological tissue repair. Fibrogenesis Tissue Repair. 5(Suppl 1): S52012. View Article : Google Scholar : PubMed/NCBI | |
|
Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Neuzillet C, Tijeras-Raballand A, Cros J, Faivre S, Hammel P and Raymond E: Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev. 32:585–602. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rønnov-Jessen L, Petersen OW and Bissell MJ: Cellular changes involved in conversion of normal to malignant breast: Importance of the stromal reaction. Physiol Rev. 76:69–125. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Chiquet-Ehrismann R, Mackie EJ, Pearson CA and Sakakura T: Tenascin: An extracellular matrix protein involved in tissue interactions during fetal development and oncogenesis. Cell. 47:131–139. 1986. View Article : Google Scholar : PubMed/NCBI | |
|
Kyutoku M, Taniyama Y, Katsuragi N, Shimizu H, Kunugiza Y, Iekushi K, Koibuchi N, Sanada F, Oshita Y and Morishita R: Role of periostin in cancer progression and metastasis: Inhibition of breast cancer progression and metastasis by anti-periostin antibody in a murine model. Int J Mol Med. 28:181–186. 2011.PubMed/NCBI | |
|
Mackie EJ, Chiquet-Ehrismann R, Pearson CA, Inaguma Y, Taya K, Kawarada Y and Sakakura T: Tenascin is a stromal marker for epithelial malignancy in the mammary gland. Proc Natl Acad Sci USA. 84:4621–4625. 1987. View Article : Google Scholar : PubMed/NCBI | |
|
Ouyang G, Liu M, Ruan K, Song G, Mao Y and Bao S: Upregulated expression of periostin by hypoxia in non-small-cell lung cancer cells promotes cell survival via the Akt/PKB pathway. Cancer Lett. 281:213–219. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Ruan K, Bao S and Ouyang G: The multifaceted role of periostin in tumorigenesis. Cell Mol Life Sci. 66:2219–2230. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Boire A, Covic L, Agarwal A, Jacques S, Sherifi S and Kuliopulos A: PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell. 120:303–313. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Sternlicht MD, Lochter A, Sympson CJ, Huey B, Rougier JP, Gray JW, Pinkel D, Bissell MJ and Werb Z: The stromal proteinase MMP3/stromelysin-1 promotes mammary carcinogenesis. Cell. 98:137–146. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Hotary KB, Allen ED, Brooks PC, Datta NS, Long MW and Weiss SJ: Membrane type I matrix metalloproteinase usurps tumor growth control imposed by the three-dimensional extracellular matrix. Cell. 114:33–45. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Valkenburg KC, de Groot AE and Pienta KJ: Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 15:366–381. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Nusse R and Clevers H: Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Melnik S, Dvornikov D, Müller-Decker K, Depner S, Stannek P, Meister M, Warth A, Thomas M, Muley T, Risch A, et al: Cancer cell specific inhibition of Wnt/β-catenin signaling by forced intracellular acidification. Cell Discov. 4:372018. View Article : Google Scholar | |
|
Deng YZ, Yao F, Li JJ, Mao ZF, Hu PT, Long LY, Li G, Ji XD, Shi S, Guan DX, et al: RACK1 suppresses gastric tumorigenesis by stabilizing the β-catenin destruction complex. Gastroenterology. 142:812–823.e15. 2012. View Article : Google Scholar | |
|
Austinat M, Dunsch R, Wittekind C, Tannapfel A, Gebhardt R and Gaunitz F: Correlation between beta-catenin mutations and expression of Wnt-signaling target genes in hepatocellular carcinoma. Mol Cancer. 7:212008. View Article : Google Scholar : PubMed/NCBI | |
|
Brenner KA, Corbett SA and Schwarzbauer JE: Regulation of fibronectin matrix assembly by activated Ras in transformed cells. Oncogene. 19:3156–3163. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Werbajh SE, Urtreger AJ, Puricelli LI, de Lustig ES, de Kier Joffé E and Kornblihtt AR: Downregulation of fibronectin transcription in highly metastatic adenocarcinoma cells. FEBS Lett. 440:277–281. 1998. View Article : Google Scholar | |
|
Seo JY, Lee SH, Youn CS, Choi HR, Rhie GE, Cho KH, Kim KH, Park KC, Eun HC and Chung JH: Ultraviolet radiation increases Tropoelastin mRNA expression in the epidermis of human skin in vivo. J Invest Dermatol. 116:915–919. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Taddese S, Weiss AS, Jahreis G, Neubert RH and Schmelzer CE: In vitro degradation of human tropoelastin by MMP-12 and the generation of matrikines from domain 24. Matrix Biol. 28:84–91. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Wang Y, Feng J, Ni Y, Zhang T, Cao Y, Zhou M and Zhao C: Mechanism and application of fibrous proteins in diabetic wound healing: A literature review. Front Endocrinol (Lausanne). 15:14305432024. View Article : Google Scholar : PubMed/NCBI | |
|
Giaquinto AN, Sung H, Newman LA, Freedman RA, Smith RA, Star J, Jemal A and Siegel RL: Breast cancer statistics 2024. CA Cancer J Clin. 74:477–495. 2024.PubMed/NCBI | |
|
Yi CH, Smith DJ, West WW and Hollingsworth MA: Loss of fibulin-2 expression is associated with breast cancer progression. Am J Pathol. 170:1535–1545. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Ibrahim AM, Sabet S, El-Ghor AA, Kamel N, Anis SE, Morris JS and Stein T: Fibulin-2 is required for basement membrane integrity of mammary epithelium. Sci Rep. 8:141392018. View Article : Google Scholar : PubMed/NCBI | |
|
Longmate WM, Monichan R, Chu ML, Tsuda T, Mahoney MG and DiPersio CM: Reduced fibulin-2 contributes to loss of basement membrane integrity and skin blistering in mice lacking Integrin α3β1 in the epidermis. J Invest Dermatol. 134:1609–1617. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Klingen TA, Chen Y, Aas H, Wik E and Akslen LA: Fibulin-2 expression associates with vascular invasion and patient survival in breast cancer. PLoS One. 16:e02497672021. View Article : Google Scholar : PubMed/NCBI | |
|
Cal S and López-Otín C: ADAMTS proteases and cancer. Matrix Biol. 44-46:77–85. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Noël A, Gutiérrez-Fernández A, Sounni NE, Behrendt N, Maquoi E, Lund IK, Cal S, Hoyer-Hansen G and López-Otín C: New and paradoxical roles of matrix metalloproteinases in the tumor microenvironment. Front Pharmacol. 3:1402012. View Article : Google Scholar : PubMed/NCBI | |
|
Luan Y, Kong L, Howell DR, Ilalov K, Fajardo M, Bai XH, Di Cesare PE, Goldring MB, Abramson SB and Liu CJ: Inhibition of ADAMTS-7 and ADAMTS-12 degradation of cartilage oligomeric matrix protein by alpha-2-macroglobulin. Osteoarthritis Cartilage. 16:1413–1420. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Fontanil T, Rúa S, Llamazares M, Moncada-Pazos A, Quirós PM, García-Suárez O, Vega JA, Sasaki T, Mohamedi Y, Esteban MM, et al: Interaction between the ADAMTS-12 metalloprotease and fibulin-2 induces tumor-suppressive effects in breast cancer cells. Oncotarget. 5:1253–1264. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Kelwick R, Desanlis I, Wheeler GN and Edwards DR: The ADAMTS (A DisIntegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol. 16:1132015. View Article : Google Scholar : PubMed/NCBI | |
|
Yu Y, Xiao CH, Tan LD, Wang QS, Li XQ and Feng YM: Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signaling. Br J Cancer. 110:724–732. 2014. View Article : Google Scholar | |
|
Cuffaro D, Ciccone L, Rossello A, Nuti E and Santamaria S: Targeting Aggrecanases for osteoarthritis therapy: From zinc chelation to exosite inhibition. J Med Chem. 65:13505–13532. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Niland S, Riscanevo AX and Eble JA: Matrix metalloproteinases shape the tumor microenvironment in cancer progression. Int J Mol Sci. 23:1462021. View Article : Google Scholar | |
|
Ghalehbandi S, Yuzugulen J, Pranjol MZI and Pourgholami MH: The role of VEGF in cancer-induced angiogenesis and research progress of drugs targeting VEGF. Eur J Pharmacol. 949:1755862023. View Article : Google Scholar : PubMed/NCBI | |
|
Reddy RA, Varshini MS and Kumar RS: Matrix metalloproteinase-2 (MMP-2): As an essential factor in cancer progression. Recent Pat Anticancer Drug Discov. 20:26–44. 2025. View Article : Google Scholar | |
|
Hergeth SP, Aicher WK, Essl M, Schreiber TD, Sasaki T and Klein G: Characterization and functional analysis of osteoblast-derived fibulins in the human hematopoietic stem cell niche. Exp Hematol. 36:1022–1034. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Rashid ZA and Bardaweel SK: Novel matrix metalloproteinase-9 (MMP-9) inhibitors in cancer treatment. Int J Mol Sci. 24:121332023. View Article : Google Scholar : PubMed/NCBI | |
|
Astudillo P: Extracellular matrix stiffness and Wnt/β-catenin signaling in physiology and disease. Biochem Soc Trans. 48:1187–1198. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Henderson NC, Rieder F and Wynn TA: Fibrosis: From mechanisms to medicines. Nature. 587:555–566. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Caligiuri G and Tuveson DA: Activated fibroblasts in cancer: Perspectives and challenges. Cancer Cell. 41:434–449. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, Washington MK, Neilson EG and Moses HL: TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 303:848–851. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Cruz-Munoz W and Khokha R: The role of tissue inhibitors of metalloproteinases in tumorigenesis and metastasis. Crit Rev Clin Lab Sci. 45:291–338. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Bloomston M, Shafii A, Zervos EE and Rosemurgy AS: TIMP-1 overexpression in pancreatic cancer attenuates tumor growth, decreases implantation and metastasis, and inhibits angiogenesis. J Surg Res. 102:39–44. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J and Khalil RA: Matrix metalloproteinases in normal pregnancy and preeclampsia. Prog Mol Biol Transl Sci. 148:87–165. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy G: Tissue inhibitors of metalloproteinases. Genome Biol. 12:2332011. View Article : Google Scholar : PubMed/NCBI | |
|
Shimoda M, Jackson HW and Khokha R: Tumor suppression by stromal TIMPs. Mol Cell Oncol. 3:e9750822016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhai LL, Cai CY, Wu Y and Tang ZG: Correlation and prognostic significance of MMP-2 and TFPI-2 differential expression in pancreatic carcinoma. Int J Clin Exp Pathol. 8:682–691. 2015.PubMed/NCBI | |
|
Song G, Xu S, Zhang H, Wang Y, Xiao C, Jiang T, Wu L, Zhang T, Sun X, Zhong L, et al: TIMP1 is a prognostic marker for the progression and metastasis of colon cancer through FAK-PI3K/AKT and MAPK pathway. J Exp Clin Cancer Res. 35:1482016. View Article : Google Scholar : PubMed/NCBI | |
|
Shimoda M, Principe S, Jackson HW, Luga V, Fang H, Molyneux SD, Shao YW, Aiken A, Waterhouse PD, Karamboulas C, et al: Loss of the Timp gene family is sufficient for the acquisition of the CAF-like cell state. Nat Cell Biol. 16:889–901. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Tjomsland V, Pomianowska E, Aasrum M, Sandnes D, Verbeke CS and Gladhaug IP: Profile of MMP and TIMP expression in human pancreatic stellate cells: Regulation by IL-1α and TGFβ and implications for migration of pancreatic cancer cells. Neoplasia. 18:447–456. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
https://gdt.gradepro.org/app/handbook/handbook.html#h.9rdbelsnu4iy. Accessed 22 Jul 2023 | |
|
Whiteaker JR, Zhang H, Zhao L, Wang P, Kelly-Spratt KS, Ivey RG, Piening BD, Feng LC, Kasarda E, Gurley KE, et al: Integrated pipeline for mass spectrometry-based discovery and confirmation of biomarkers demonstrated in a mouse model of breast cancer. J Proteome Res. 6:3962–3975. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Avsar M, Tambas M, Yalniz Z, Akdeniz D, Tuncer SB, Kilic S, Erdogan OS, Ciftci R, Dagoglu N, Vatansever S and Yazici H: The expression level of fibulin-2 in the circulating RNA (ctRNA) of epithelial tumor cells of peripheral blood and tumor tissue of patients with metastatic lung cancer. Mol Biol Rep. 46:4001–4008. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
WalyEldeen AA, Sabet S, Anis SE, Stein T and Ibrahim AM: FBLN2 is associated with basal cell markers Krt14 and ITGB1 in mouse mammary epithelial cells and has a preferential expression in molecular subtypes of human breast cancer. Breast Cancer Res Treat. 208:6872024. View Article : Google Scholar | |
|
Setiawati Y, Alimuddin T, Mulyani H and Kamelia M: The prognostic role of fibulin-2 and Ki-67 index in patients with meningioma: A study among minangkabau ethnicity. Asian Pac J Cancer Prev. 25:2735–2742. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ibrahim AM, Roshdy M, Elshorbagy S, Hosny M, Halawa S, Yehia D, Elfawy HA, Eldessouki A, Mohamed F, Ellithy A, et al: An investigation of fibulin-2 in hypertrophic cardiomyopathy. Int J Mol Sci. 21:71762020. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Jiang H, Xing W, Wang S, Zhang Y, Li Y, Mao C, Zeng D, Lan P, Tang D, et al: A clinical diagnostic study: Fibulin-2 is a novel promising biomarker for predicting infection. Infect Dis Ther. 11:1057–1073. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Knittel T, Kobold D, Piscaglia F, Saile B, Neubauer K, Mehde M, Timpl R and Ramadori G: Localization of liver myofibroblasts and hepatic stellate cells in normal and diseased rat livers: Distinct roles of (myo-)fibroblast subpopulations in hepatic tissue repair. Histochem Cell Biol. 112:387–401. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Li SD, Xing W, Wang SC, Li YB, Jiang H, Zheng HX, Li XM, Yang J, Guo DB, Xie XY, et al: Fibulin2: A negative regulator of BMSC osteogenic differentiation in infected bone fracture healing. Exp Mol Med. 55:443–456. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Papon MA, Le Feuvre Y, Barreda-Gómez G, Favereaux A, Farrugia F, Bouali-Benazzouz R, Nagy F, Rodríguez-Puertas R and Landry M: Spinal inhibition of GABAB receptors by the extracellular matrix protein fibulin-2 in neuropathic rats. Front Cell Neurosci. 14:2142020. View Article : Google Scholar : PubMed/NCBI | |
|
Ghorbani S, Li C, Lozinski BM, Moezzi D, D'Mello C, Dong Y, Visser F, Li H, Silva C, Khakpour M, et al: Fibulin-2 is an extracellular matrix inhibitor of oligodendrocytes relevant to multiple sclerosis. J Clin Invest. 134:e1769102024. View Article : Google Scholar : PubMed/NCBI | |
|
Sicot FX, Tsuda T, Markova D, Klement JF, Arita M, Zhang RZ, Pan TC, Mecham RP, Birk DE and Chu ML: Fibulin-2 is dispensable for mouse development and elastic fiber formation. Mol Cell Biol. 28:1061–1067. 2008. View Article : Google Scholar : | |
|
Zhang H, Wu J, Dong H, Khan SA, Chu ML and Tsuda T: Fibulin-2 deficiency attenuates angiotensin II-induced cardiac hypertrophy by reducing transforming growth factor-β signaling. Clin Sci (Lond). 126:275–288. 2014. View Article : Google Scholar | |
|
Tsuda T, Wu J, Gao E, Joyce J, Markova D, Dong H, Liu Y, Zhang H, Zou Y, Gao F, et al: Loss of fibulin-2 protects against progressive ventricular dysfunction after myocardial infarction. J Mol Cell Cardiol. 52:273–282. 2012. View Article : Google Scholar : | |
|
Fässler R, Sasaki T, Timpl R, Chu ML and Werner S: Differential regulation of fibulin, tenascin-C, and nidogen expression during wound healing of normal and glucocorticoid-treated mice. Exp Cell Res. 222:111–116. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Kida Y, Asahina K, Inoue K, Kawada N, Yoshizato K, Wake K and Sato T: Characterization of vitamin A-storing cells in mouse fibrous kidneys using Cygb/STAP as a marker of activated stellate cells. Arch Histol Cytol. 70:95–106. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Piscaglia F, Dudás J, Knittel T, Di Rocco P, Kobold D, Saile B, Zocco MA, Timpl R and Ramadori G: Expression of ECM proteins fibulin-1 and -2 in acute and chronic liver disease and in cultured rat liver cells. Cell Tissue Res. 337:449–462. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Zhang W, Zhang R and Xia Y: Knockdown of FBLN2 suppresses TGF-β1-induced MRC-5 cell migration and fibrosis by downregulating VTN. Tissue Cell. 81:1020052023. View Article : Google Scholar | |
|
Li S, Jiang H, Wang S, Li Y, Guo D, Zhan J, Li Q, Meng H, Chen A, Chen L, et al: Fibulin-2: A potential regulator of immune dysfunction after bone trauma. Immun Inflamm Dis. 11:e8462023. View Article : Google Scholar : PubMed/NCBI | |
|
Radice PD, Mathieu P, Leal MC, Farías MI, Ferrari C, Puntel M, Salibe M, Chernomoretz A and Pitossi FJ: Fibulin-2 is a key mediator of the pro-neurogenic effect of TGF-beta1 on adult neural stem cells. Mol Cell Neurosci. 67:75–83. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Patel MR and Weaver AM: Astrocyte-derived small extracellular vesicles promote synapse formation via fibulin-2-mediated TGF-β signaling. Cell Rep. 34:1088292021. View Article : Google Scholar | |
|
Bois PR and Grosveld GC: FKHR (FOXO1a) is required for myotube fusion of primary mouse myoblasts. EMBO J. 22:1147–1157. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Lehka L and Rędowicz MJ: Mechanisms regulating myoblast fusion: A multilevel interplay. Semin Cell Dev Biol. 104:81–92. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chan CY, Masui O, Krakovska O, Belozerov VE, Voisin S, Ghanny S, Chen J, Moyez D, Zhu P, Evans KR, et al: Identification of differentially regulated secretome components during skeletal myogenesis. Mol Cell Proteomics. 10:M110.0048042011. View Article : Google Scholar : PubMed/NCBI | |
|
Ström A, Olin AI, Aspberg A and Hultgårdh-Nilsson A: Fibulin-2 is present in murine vascular lesions and is important for smooth muscle cell migration. Cardiovasc Res. 69:755–763. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Ball K, Dovedi SJ, Vajjah P and Phipps A: Strategies for clinical dose optimization of T cell-engaging therapies in oncology. MAbs. 15:21810162023. View Article : Google Scholar : PubMed/NCBI | |
|
Gueorguieva I, Cleverly AL, Stauber A, Pillay NS, Rodon JA, Miles CP, Yingling JM and Lahn MM: Defining a therapeutic window for the novel TGF-β inhibitor LY2157299 monohydrate based on a pharmacokinetic/pharmacodynamic model. Br J Clin Pharmacol. 77:796–807. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ou FS, Michiels S, Shyr Y, Adjei AA and Oberg AL: Biomarker discovery and validation: Statistical considerations. J Thorac Oncol. 16:537–545. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kraus VB: Biomarkers as drug development tools: Discovery, validation, qualification and use. Nat Rev Rheumatol. 14:354–362. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Wu H, Chong W, Shang L, Jing C and Li L: Liquid biopsy in gastric cancer: Predictive and prognostic biomarkers. Cell Death Dis. 13:9032022. View Article : Google Scholar : PubMed/NCBI | |
|
Galarini R, Diana F, Moretti S, Puppini B, Saluti G and Persic L: Development and validation of a new qualitative ELISA screening for multiresidue detection of sulfonamides in food and feed. Food Control. 35:300–310. 2014. View Article : Google Scholar | |
|
Wang S, Xu B, Zhang Y and He JX: Development of enzyme-linked immunosorbent assay (ELISA) for the detection of neomycin residues in pig muscle, chicken muscle, egg, fish, milk and kidney. Meat Sci. 82:53–58. 2009. View Article : Google Scholar | |
|
Petz M: Recent applications of surface plasmon resonance biosensors for analyzing residues and contaminants in food. Monatshefte für Chemie-Chemical Monthly. 140:953–964. 2009. View Article : Google Scholar | |
|
Ahirwar R, Bhattacharya A and Kumar S: Unveiling the underpinnings of various non-conventional ELISA variants: A review article. Expert Rev Mol Diagn. 22:761–774. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Moreno BH and Ribas A: Anti-programmed cell death protein-1/ligand-1 therapy in different cancers. Br J Cancer. 112:1421–1427. 2015. View Article : Google Scholar | |
|
Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F, Izzeddine H, Marabelle A, Champiat S, Berdelou A, et al: Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 13:473–486. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Moreno V, Hernandez T, de Miguel M, Doger B and Calvo E: Adoptive cell therapy for solid tumors: Chimeric antigen receptor T cells and beyond. Curr Opin Pharmacol. 59:70–84. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Xu J, Wang W, Zhang B, Yu X and Shi S: Epigenetic regulation in the tumor microenvironment: Molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 8:2102023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Han R, Su J, Chen H and Li D: Multi-marker diagnosis method for early hepatocellular carcinoma based on surface plasmon resonance. Clin Chim Acta. 502:9–14. 2020. View Article : Google Scholar | |
|
Yuan W, Tang W, Xie Y, Wang S, Chen Y, Qi J, Qiao Y and Ma J: New combined microRNA and protein plasmatic biomarker panel for pancreatic cancer. Oncotarget. 7:80033–80045. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ginghina O, Hudita A, Zamfir M, Spanu A, Mardare M, Bondoc I, Buburuzan L, Georgescu SE, Costache M, Negrei C, et al: Liquid biopsy and artificial intelligence as tools to detect signatures of colorectal malignancies: A modern approach in patient's stratification. Front Oncol. 12:8565752022. View Article : Google Scholar : PubMed/NCBI | |
|
Weng X, Gaur G and Neethirajan S: Rapid detection of food allergens by microfluidics ELISA-based optical sensor. Biosensors (Basel). 6:242016. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng CM, Martinez AW, Gong J, Mace CR, Phillips ST, Carrilho E, Mirica KA and Whitesides GM: Paper-based ELISA. Angew Chem Int Ed Engl. 49:4771–4774. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Kudłak B and Wieczerzak M: Aptamer based tools for environmental and therapeutic monitoring: A review of developments, applications, future perspectives. Crit Rev Environ Sci Technol. 50:816–867. 2020. View Article : Google Scholar | |
|
Bauer M, Strom M, Hammond DS and Shigdar S: Anything you can do, I can do better: Can aptamers replace antibodies in clinical diagnostic applications? Molecules. 24:43772019. View Article : Google Scholar : PubMed/NCBI |