Next Article in Journal
Itchy E3 Ubiquitin Ligase-Mediated Ubiquitination of Ferritin Light Chain Contributes to Endothelial Ferroptosis in Atherosclerosis
Previous Article in Journal
Plasma microRNAs Associate Positive, Negative, and Cognitive Symptoms with Inflammation in Schizophrenia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Multifaced Role of Collagen in Cancer Development and Progression

by
Gabriele Lo Buglio
1,2,
Alessandra Lo Cicero
1,*,
Simona Campora
1 and
Giulio Ghersi
1,3,*
1
Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
2
Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
3
Abiel srl, 90128 Palermo, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(24), 13523; https://doi.org/10.3390/ijms252413523
Submission received: 20 November 2024 / Revised: 10 December 2024 / Accepted: 13 December 2024 / Published: 17 December 2024
(This article belongs to the Section Molecular Oncology)

Abstract

:
Collagen is a crucial protein in the extracellular matrix (ECM) essential for preserving tissue architecture and supporting crucial cellular functions like proliferation and differentiation. There are twenty-eight identified types of collagen, which are further divided into different subgroups. This protein plays a critical role in regulating tissue homeostasis. However, in solid tumors, the balance can be disrupted, due to an abundance of collagen in the tumor microenvironment, which significantly affects tumor growth, cell invasion, and metastasis. It is important to investigate the specific types of collagens in cancer ECM and their distinct roles in tumor progression to comprehend their unique contribution to tumor behavior. The diverse pathophysiological functions of different collagen types in cancers illustrate collagen’s dual roles, offering potential therapeutic options and serving as prognostic markers.

1. Introduction

Collagen is the most abundant protein in the human body, accounting for 25–30% of its total protein content and is the main constituent of various tissues, including bones, ligaments, and skin [1]. As the fundamental component of the extracellular matrix (ECM), collagen plays a crucial role in the structural and functional organization of all tissues [2,3]. The ECM is a dynamic three-dimensional network of macromolecules, such as glycoproteins, proteoglycans, fibronectin, and laminin, with functions that are highly dependent on protein compositions. These functions include pH maintaining, regulating cell proliferation and differentiation, and providing support and anchorage for cells [4,5]. Collagen and other ECM components vary in structure and function, thereby regulating tissue homeostasis.
The equilibrium of ECM can be affected by injuries, aging, and various diseases [6]. Recent research studies have focused on understanding the role of ECM and collagen in the formation and progression of solid tumors. Together with tumor vasculature, associated connective tissues, infiltrating immune cells, and cancer-associated fibroblasts (CAFs), the component of ECM constitutes the tumor microenvironment (TME) of solid tumors [7]. Therefore, solid tumors are characterized by ECM rich in collagen, which encapsulates tumor cells, influencing cell accessibility and metabolism, and promoting tumor cell progression, invasion, and migration. This high collagen density leads to a TME stiffness that is significantly stronger compared to healthy tissues [8,9]. This condition is also influenced by the orientation of collagen fibers, which form a random reticle, dynamically remodeled during disease progression [10,11,12]. Excessive deposition and specific orientation of collagen are considered the major pathological features of solid tumors, allowing collagens to play active roles in cancer [13,14]. Increasing evidence suggests that the various types of collagens in the tumor ECM promote tumor proliferation and invasion and create conditions that enhance metastatic dissemination [15].
This review aims to provide an overview of the pathological role of different types of collagens in solid tumors, highlighting how these molecules represent excellent therapeutic targets and reliable prognostic markers. By exploring the diverse functions of collagen within the ECM, it is possible to better understand its impact on tumor biology and identify novel approaches to modulate the ECM for improved cancer treatment outcomes.

2. Collagen: Role and Structure

Along with fibronectin, laminin, and elastin, collagen constitutes the matrisome of the ECM, composed of approximately 300 proteins [16]. Collagen is a complex and dynamic macromolecule, assuming various structures in different tissues, conferring them a range of biological functions. To date, 28 collagen subtypes have been identified, with type I collagen representing over 90%, being the primary component of organs and connective tissues [6]. As a key component of connective tissues, collagen plays an essential role in maintaining their structural and biological integrity [17]. The synthesis and remodeling of collagens are dynamic mechanisms that are closely dependent on the physiological and, in some cases, pathological conditions of the tissue. These processes primarily involve fibroblasts, chondrocytes, osteoblasts, smooth muscle cells (SMCs), epithelial cells, endothelial cells, hepatic stellate cells, and cardiac myocytes [18,19,20,21,22,23,24]. The α chains of various types of collagen are used to build trimeric molecules, which intertwine into a 300 nm long triple helix and subsequently assemble into fibrils. Some types, such as type II collagen, form homotrimeric molecules, while others, such as types I and V, form heterotrimers [25,26]. The collagen components interact in a specific sequence with each other and with other tissue proteins and cells, producing higher-order structures. For example, continuous crosstalk occurs between collagen fibers and resident fibroblasts, allowing for a balance of tensile and recoil energy in a tissue, and modulating mechanotransduction pathways [27]. The modulation of the intracellular signaling pathways also depends on the turnover of the various collagens within the extracellular matrix. This process is highly dependent on the activity of specific proteases, particularly matrix metalloproteinases (MMPs) secreted by resident cells [28,29]. These enzymes modify the different extracellular matrix components, altering the density of the microenvironment and leading to mechanical loads perceived by the cells. Several studies have examined the influence of mechanical force on the extracellular matrix formation. However, the complexity of controlling and assembling collagen fibrils is still not well understood. Some studies suggest both cell-dependent and cell-independent models for collagen organization, highlighting the importance of mechanochemistry in determining fibril arrangement under mechanical stress [30].

3. Biosynthesis

The different types of collagen share the canonical sequence domain Gly-X-Y, where X and Y can vary with a consistent presence of proline and hydroxyproline residues [31]. Hydroxyproline is an amino acid not commonly found in the amino acid sequence of proteins; however, it may account for more than 50% of the total amino acid content of collagen [32]. The high percentage of these residues is critical for reducing the entropic contribution during collagen filament folding [33]. Collagen synthesis is a multi-step process involving several enzymes. After synthesis, pre-collagen undergoes disulfide bond formation at the N- and C-terminal propeptide regions followed by post-translational modifications, mainly on lysines and prolines, such as hydroxylation (via lysyl hydroxylase activity) and glycosylation (through the addition of glucose and galactose oligosaccharides), leading to the formation of pro-collagen α chains [34]. Lysine hydroxylation induces the formation of triple helices, which are then secreted into the extracellular environment via Golgi vesicles [15]. Once secreted, collagen undergoes a combined activity of lysyl oxidases (LOX) and some classes of LOX-like proteins (LOXL) [35], which function as cross-linking agents found both in the intracellular and extracellular environment. They are necessary for collagen’s mechanical properties acquisition, such as tensile strength, contributing to the regulation of ECM stiffness [36,37].

4. Tumoral Extracellular Matrix

Tumorigenesis is a complex and dynamic process involving the cooperation of various elements within the tumor microenvironment (TME) [7]. The TME components play several critical roles in promoting and sustaining tumor progression. These roles include the deregulation of proliferative signals and the evasion of growth suppression control. Consequently, tumor cells acquire several characteristics: resistance to apoptosis, uncontrolled proliferation, promotion of tissue colonization, modulation of cellular metabolism, stimulation of angiogenesis, and evasion from the immune system [38]. The TME is composed of multiple cellular components, such as endothelial cells (ECs), immunocytes, adipocytes, cancer-associated fibroblasts (CAFs), and the ECM [3] (Figure 1).
CAFs are pivotal components of the TME with enhanced functional properties, such as proliferation, migration, and secretion. They persist in a hyperactivated condition and exhibit elevated metabolic activity. Their activity contributes to shaping a thick tumoral extracellular matrix, through synthesis and abnormal deposition of fibronectin, laminin, hyaluronic acid, and collagen types I, III, IV, and V, above all collagen type I [39,40,41].
The ECM plays a key role in the TME, encapsulating tumor cells within an acidic and hypoxic extracellular matrix, which reduces cell accessibility and influences their metabolism. From a structural point of view, the ECM of solid tumors is an envelope that surrounds, protects, and interacts with tumor cells. Moreover, due to its macromolecular composition, the tumor ECM exhibits abnormal rigidity compared to the ECM of healthy tissues. For example, while the stiffness of the ECM in healthy tissues such as the breast, brain, liver, lung, and pancreas is typically around 1000 Pa, it can reach values between 4 and 10 kPa in tumors located in these same sites [8]. This increased stiffness can result from different biomechanical factors, including an imbalance between matrix deposition and degradation, the presence of contractile actomyosin in CAFs, interactions between tumor and normal cells, and especially altered collagen cross-linking [42,43]. Tumor ECM shares several components with healthy tissues ECM, such as collagens, elastin, proteoglycans, and glycoproteins like tenascin-C, laminins, and fibronectins [44]. However, changes in the relative composition of these elements lead to structural and functional alterations that promote tumor development [45]. The ECM components contribute to a fibrotic extracellular environment, modified by multiple factors, such as MMPs. MMPs actively contribute to tumor progression by participating in ECM turnover, which influences tumor cell proliferation, angiogenesis, and invasion, as well as apoptosis inhibition [46].

5. Collagens in Solid Tumors

Collagen is a key component of the ECM in the TME, significantly contributing to tumors’ physical and biochemical properties. The organization and density of collagen fibers can significantly affect the progression and spread of tumors. Collagen provides structural support to tumors, creating a scaffold that aids tumor growth. Increased collagen deposition leads to a stiffer ECM, promoting tumor cell proliferation and survival. Cancer cells interact with collagen through integrins and other receptors, transducing signals that influence tumor cell behavior, leading to cell growth, invasion, resistance to chemotherapy, metabolism variation, invasion, migration, and evasion to the immune system.

5.1. Collagen and Cancer Cell Proliferation

Collagen and other ECM components interact directly with tumor cells, influencing their growth. These molecules play an important role in regulating the cell cycle. Some fibrillar collagens suppress tumor growth by interacting with Discoidin Domain Receptor 2 (DDR2), a transmembrane receptor tyrosine kinase (RTK). Along with Discoidin Domain Receptor 1 (DDR1), DDR2 binds collagen in the extracellular matrix of various tissues. This interaction leads to cell cycle arrest at the G1/S checkpoint [47]. Other collagens, such as type III collagen, induce tumor cells into dormancy via the DDR1/Signal Transducer and Activator of Transcription 1 (STAT1) signaling pathway [48]. However, most of them promote tumor cell proliferation through various pathways. For instance, it has been shown that monomeric collagen can inhibit the expression of p21 in tumor cells in vitro, altering cell cycle restrictions [49]. Moreover, collagen interaction with integrin beta-1 leads to increased tumor cell proliferation, through the activation of β-catenin [50,51] However, collagen regulates tumor cell proliferation through several mechanisms, including diverse metabolic pathways.

5.2. Collagen and Tumor Metabolism

One of the aspects that over the years led to significant attention in the scientific community is tumor glucose metabolism. Tumor cells can absorb more glucose than their healthy tissue counterparts, converting it into lactate [52]. This process, known as the Warburg effect, is regulated by numerous factors, including matrix collagen. It has been demonstrated in vitro that increased collagen density promotes aerobic glycolysis in human breast cancer cell lines, stimulating integrin activation [53]. This activates the Posphoinositide 3-Kinase (PI3K)/Protein Kinase B (Akt) signaling pathway, which positively modulates the activity of various factors upstream, such as Glucose Transporter 1 (GLUT1) and pyruvate kinase M2 (PKM2) [54,55]. Furthermore, these metabolic anomalies modify glutamine uptake and its fate in tumors. In this context, a recent in silico study focused on the increased glutamine uptake in tumor cells exploiting the Warburg effect, noting that this metabolite is directed toward stromal collagen production.
Thus, through positive feedback mechanisms, tumor cells link collagen with glucose and lipid metabolism, promoting tumor progression [56,57,58].

5.3. Collagen and Apoptosis

Apoptosis represents a highly regulated cell death process governed by various endogenous or exogenous stimuli. However, in the tumorigenesis contest, this mechanism is completely dysregulated, promoting tumor growth at the expense of surrounding tissues [59]. The involvement of matrix collagen in anti-apoptotic processes is not fully understood. The presence of collagen in gastric cancer has been shown to upregulate, both in vitro and in vivo, the expression of anti-apoptotic factors, such as B-cell lymphoma 2 (BCL2), increased by the β-catenin pathway [60]. Moreover, DDR1 pathway activation mediated by type I collagen inhibits tumor cell growth, through the pro-apoptotic factor BCL-2 interacting killer (BIK), a member of the BCL2 family [61,62]. Collagen can also suppress tumor growth by negatively modulating certain cell cycle factors, such as p21 and CDKs, and regulating caspase expression [63]. In addition to collagen, some of its peptide derivatives can induce apoptosis, by Bcl-2 and BCL-extra large (BCLxL) pathways [64].
To resume, the dual role of collagen raises many questions about its actual function in solid tumors. Given its pleiotropic effects on malignancy, different factors may play crucial roles, such as tumor type, staging, and the specific type of matrix collagen.

5.4. Collagen and Tumor Cell Invasion

Among the hallmarks of solid tumors, the invasion of tumor cells is one of the most determining factors for patient prognosis since this mechanism plays a crucial role in tumor progression and metastasis formation [65]. For instance, collagen regulates the extension of invadopodia through the integrin αvβ3/Dishevelled-associated activator of morphogenesis 1 (DAAM1)/Ras Homolog family member A (RHOA)-dependent cascade [66], which are small actin-rich protrusions that mediate the invasion of tumor cells [67]. From a biophysical perspective, the increased stromal tension caused by matrix collagen accumulation in breast cancers enhances the integrin β1-mediated signaling pathway, leading to the activation of the Focal Adhesion Kinase (FAK)/Yes-Associated Protein (YAP) pathway [68]. Activation of integrin β1 can be triggered by mutated collagens at one or more sites, which accelerate the tumor cell invasion, as observed in invasive epithelial tumors [69]. However, YAP activation must be corroborated by collagen matrix remodeling, mediated by Lox3 and MMPs, which are regulated by the microenvironment stiffness [70]. On the other hand, collagen degradation, or the loss of ECM stiffness induced by ultraviolet radiation (UVR) against fibroblasts, could surprisingly increase the survival of melanoma patients due to the lack of matrix support for invasiveness [71].

5.5. Collagen and Metastasis

Besides driving tumors to cell invasion, collagen plays a key role in cell migration. The ECM of solid tumors changes the acquisition of metastatic potential [72], and the collagen within it acts as a “molecular binary” to increase the tumor cell’s escape from the primary site [73,74]. However, cancer research has recently focused on the pathways regulated by stromal collagen implicated in this process. For instance, the DDR1 or DDR2 activation mediated by collagen positively regulates the metastatic process involving tumor-associated neutrophils (TANs) and CAFs [75,76]. Upregulation of collagen prolyl-4-hydroxylases (P4HA) of class I and II, whose expression is increased by tissue hypoxia [77], could drive collagen deposition in metastatic ovarian tumors, leading to neo-angiogenesis and tumor cell migration [78].
Thus, metastasis is a complex process involving multiple converging and diverging signaling pathways, often upstream regulated by collagen. Some pathways are even independent, such as versican (VCAN)/Extracellular signal-Regulated Kinase (ERK) and poly [ADP-ribose] polymerase 1 (PARP1)/Zinc finger E-box-Binding homeobox 1 (ZEB1) pathways, which promote the proliferation and migration of colorectal cancer cells, regulated directly or indirectly by collagen [79]. In other cases, collagen can be secreted into exosomes to promote metastasis, as observed in osteosarcomas [80].

5.6. Collagen and Tumor Angiogenesis

Increased collagen deposition and its alignment are characteristics that impact neovascularization [81,82]. More specifically, the high density of collagen and other matrix components creates a hypoxic environment at the tumor core, activating the hypoxia-inducible factor (HIF-1) [83]. HIF-1 is a pivotal transcription factor for vascular endothelial growth factor (VEGF), which stimulates neo-angiogenesis and actively contributes to tumor progression [84]. Additionally, HIF-1 is involved in immune evasion mechanisms through the expression of immunosuppressors, such as those regulating antigen presentation to Natural Killer (NK) cells [85].
Thus, collagen plays a physical role in tumor vascular system formation. However, some studies have also examined the biochemical role of collagen in neo-angiogenesis, investigating the pathways in which this protein is involved. More specifically, some of its peptide derivatives exhibit anti-angiogenic effects involving integrins αvβ3 and α5β1 [86]. Other peptides, including those derived from type IV collagen, inhibit endothelial cell proliferation stimulated by basic fibroblast growth factor (bFGF) or induce apoptosis in endothelial cells via a Fas-dependent mechanism [87], confirming the pleiotropic nature of tumor-associated collagen.

5.7. Collagen and Chemoresistance

Despite the significant advancements in research, chemoresistance remains the main challenge in tumor relapse and therapeutic ineffectiveness [88]. ECM, particularly collagen, plays a crucial role in chemoresistance, acting as a physical barrier against drugs. Additionally, collagen can influence various biochemical processes, such as dormancy, where tumor cells enter a quiescent state and become desensitized to chemotherapeutic agents before re-entering the G1/S phase [89,90]. Another important mechanism of chemoresistance is drug efflux, mediated by the upregulation of the ATP-binding cassette (ABC) superfamily, with P-glycoprotein (ABCB1) being the most well-known factor [91]. Specifically, ABCs can be positively modulated by α2β1-integrin, which is activated by the stromal collagen [92]. Moreover, collagen can also affect the transcription of certain genes to promote chemoresistance. In ovarian carcinoma, collagen can stabilize and activate DNA methyltransferase 1 (DNMT1), which leads to methylation and downregulation of miR-509-3p promoter, an anti-tumor miRNA that also suppresses chemoresistance [93].
Therefore, in addition to physically preventing drug entry, collagen in the tumor matrix can access multiple signaling pathways that reduce therapeutic efficacy.

5.8. Collagen and Immunomodulation

ECM plays an immunomodulatory role in solid tumors. This dense and fibrotic structure, rich in collagen, creates barriers that hinder the infiltration of chemotherapeutics and immune cells. For instance, the ECM can influence leukocyte localization and transmigration, reducing their accessibility to the tumor mass [94,95]. Among the matrix components, collagen regulates immune cell activity, such as T lymphocytes and macrophages, often favoring tumor progression [96,97]. The increased matrix density and collagen alignment suppress the tumor-infiltrating T lymphocyte (TIL) activity [98]. It has been shown in vivo that tumor collagen alignment mediated by the DDR1 extracellular domain binding hinders the infiltration of CD4+ and CD8+ T lymphocytes [99]. Supporting this study, the use of PRTH-101 (monoclonal anti-DDR1 antibodies) in breast cancer mouse models disrupted stromal collagen organization, increasing immune infiltration and, consequently, survival [100]. Beyond DDRs, collagen has receptors on leukocytes, such as leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1). This receptor has an essential physiological role in preventing the immune system’s uncontrolled responses, such as autoimmune reactions [101]. However, when LAIR-1 binds collagen of the tumor microenvironment, the antitumor potential of cytotoxic T lymphocytes is suppressed in an SHP-1-dependent pathway, as seen in gliomas [102]. Therefore, LAIR-1 has recently become a therapeutic target to counteract immune evasion in solid tumors [103,104]. Similarly, collagen fragments generated by MMP1 and MMP9 can bind LAIR-1, leading to immune suppression. LAIR-2, a soluble homolog of LAIR-1, reverses T lymphocyte collagen-mediated suppression through a competitive mechanism [105].
Thus, tumor collagen and the other associated factors, such as lysyl oxidase, represent important therapeutic targets to enhance immune responses against solid tumors [106,107,108,109,110]. However, some cytokines, such as chemokines produced by immune cells, can contribute to tumor matrix formation or even interfere with the antitumor activity of the immune system. Indeed, specific interleukins (IL), such as IL-4, IL-6, and IL-13, induce collagen synthesis, contributing to tissue fibrosis and tumor progression. In addition, other factors, such as IL-10 and Transforming Growth Factor Beta-1 (TGF-β), secreted by M2 macrophages, inhibit Th1 lymphocyte responses and immune cell activation, creating an environment that supports tumor growth and immune evasion [111,112]. Moreover, it has been studied that the collagen I/DDR1 axis activates the Protein Kinase C theta (PKCθ)/Spleen tyrosine kinase (SYK)/Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) cascade, resulting in the upregulation of the chemokine C-X-C motif chemokine 5 (CXCL5). This chemokine leads to the formation of neutrophils extracellular traps (NETs) by TANs, a key mechanism driving the metastatic process [113,114].
Therefore, the relationship between the immune system and the stromal collagen in solid tumors is ambiguous, where even some immune cells can actively participate in the ECM formation and tumor progression. A striking example is tumor-associated macrophages (TAMs). These macrophages acquire specific phenotypic characteristics, polarizing from antitumoral (M1) to protumoral (M2) states through a mechanism induced by IL-4 secreted by CD4+ T cells [75]. These cells influence the tumor progression, enhancing neo-angiogenesis or tumor cell migration [115,116]. Additionally, TAM2s can secrete collagen I to activate the α2β1 integrin/PI3K/Akt axis, supporting tumor progression [117,118].
For this reason, continued studies on the relationship between stromal collagen and the immune system may uncover targeted therapies against solid tumors.

6. Mechanotransduction Mediated by Collagen in Cancer

Mechanotransduction is a complex process in which the mechanical properties of a tissue modulate intracellular signaling pathways, essential for the homeostasis of its constituent cells [119]. Mechanical homeostasis is regulated by the deposition and orientation of matrix components, such as collagens and fibronectin. The balance of stretching and tensile forces determines the elasticity or stiffness of the microenvironment. In this contest, resident cells can perceive the stiffness by acting as mechanosensors [120]. Moreover, focal adhesions are fundamental to the mechanical stimuli reception, mediating cell–matrix contacts in synergy with specific linker proteins, such as integrins and actin cytoskeleton components [121,122]. Additionally, the modulation of mechanical stimuli within the tissue microenvironment can be influenced by the cells, neighboring tissues, or the hydrostatic pressures of surrounding fluids [123]. Within this context, signaling pathways arising from the intricate relationship between a tissue’s biochemistry and physical properties regulate gene expression, modulating the phenotype and primary metabolic activities of resident cells, including proliferation, differentiation, migration, and programmed cell death mechanisms [124]. However, mechanotransduction is also fundamental in the pathology of various diseases. For instance, in tumors, the heterogeneous increase in density and cross-linking of ECM components leads to abnormal stiffness compared to healthy tissue [125]. Generally, receptor tyrosine kinases (RTKs), such as AXL receptor tyrosine kinase (AXL), Receptor Tyrosine Kinase Like Orphan Receptor 2 (ROR2), and ephrin receptors (EPHs), detect matrix contractions across different hotspot regions, working as mechanosensors [126]. For instance, it has been demonstrated that EPHA2, in the absence of its canonical ligand and in cooperation with integrin beta-1, senses the stiffening of the tumor microenvironment. This detection activates the Lck/yes-related protein tyrosine kinase (LYN) signaling pathway, resulting in the phosphorylation and nuclear translocation of the transcription cofactor Twist-related protein 1 (TWIST1). Consequently, the activated pathway triggers the epithelial–mesenchymal transition (EMT) required for tumor cell invasion [127].
The interactions in the cell–matrix interface between cancer cells and collagen can activate the transcriptional coactivator YAP. YAP selectively reprograms tumor cells, promoting the transition of outer basal tumor cells into leader cells with invasive capacities. Furthermore, YAP activation enhances the transcription of ECM remodelers involved in collagen cross-linking, establishing a positive feedback mechanism that amplifies the invasive potential of tumor cells [128,129]. Different studies have highlighted that YAP nuclear translocation is facilitated by morphological modifications of nuclear pores, induced by increased stress fibers in the perinuclear region connected to the tumor ECM [130,131,132].
Mechanical signals from the tumor microenvironment also amplify tumor cell proliferation through positive feedback mechanisms. For instance, excessive collagen deposition and other tumor matrix components are physically sensed by stromal mesenchymal stem cells (MSCs). In response, MSCs start to secrete the protein prosaposin (PSAP), which promotes tumor growth via activation of the Toll-Like Receptor (TLR)/NF-κB signaling pathway [133,134,135]. Additionally, matrix stiffness drives the differentiation of MSCs into cancer-associated fibroblasts (CAFs), which are primary contributors to collagen secretion, further reinforcing overall matrix stiffness in a self-perpetuating cycle [136].
Beyond cell invasiveness and proliferation, continuous remodeling and stiffening of the tumor-associated extracellular collagen activate angiogenic signaling necessary for new vessel development. This process engages the integrin β1/PI3K/Akt signaling pathway, promoting the secretion of factors such as activin A, amphiregulin, artemin, interleukin (IL-1β), persephin, urokinase-type plasminogen activator (uPA), and vascular endothelial growth factor (VEGF) [127,137,138]. Thus, mechanotransduction triggered by matrix components, specifically collagen, underlies cancer pathology by driving processes such as differentiation, proliferation, invasion, and angiogenesis.

7. Collagen Classification

The collagen superfamily is characterized by twenty-eight types of collagens, which are further divided into different subgroups [139]. In vertebrates, collagens are numbered using Roman numerals (I–XXVIII) [140] and are classified into fibrillar and non-fibrillar types. The predominant category is represented by fibrillar collagens, which constitute 90% of the total. These fibrillar collagens have elongated fibril-like structures (rods or bands) and include types I, II, III, V, XI, XXIV, and XXVII. On the other hand, non-fibrillar collagens form various supramolecular structures and are divided into different categories, including fibrils-associated collagens with interrupted triple helices (FACITs) (types IX, XII, XIV, XVI, XIX, XX, XXI, and XXII), network-forming collagens (types IV, VIII, and X), beaded filament-forming collagens (types VI, XXVI, and XXVIII), anchoring fibrils (type VII), transmembrane collagens (types XIII, XVII, XXIII, and XXV), and multiplexins (types XV and XVIII) [141,142]. Various collagens contribute uniquely to the tumor microenvironment, affecting tumor behavior and metastasis through different pathways, such as mechanotransduction (Figure 2).

7.1. Fibrillar Collagens

Fibrillar collagens have a continuous triple-helix structure of approximately 300 nm ø, followed by a chain of 1000 amino acids. This group includes types I, II, III, V, XI, XXIV, and XXVII collagens, which promote the development of solid tumors [143]. Several studies have analyzed collagen fibers’ architecture, characteristic of the solid tumor ECM. While it is well established that tissue fibrosis precedes neoplasms, recent findings increasingly confirm that the stratification and elongation of stromal fibers, particularly their orientation, are statistically associated with a highly aggressive tumor cell phenotype [144]. This is because the orientation of collagen fibers in the tumor ECM differs from that of healthy tissues. Specifically, while in healthy tissues collagen is isotropically oriented, tumor tissues often exhibit an anisotropic orientation, mainly due to an increased ratio of type I to type III collagen [145]. Type I collagen is not only the most abundant protein in the body, but it is also prevalent in solid tumors. Increased deposition of type I collagen (COL1) in the tumor ECM is closely correlated with the aggressiveness of many tumor types, such as triple-negative breast carcinoma, pancreatic ductal adenocarcinoma (PDAC), or gastric carcinoma [146]. For example, COL1 induces cells to spread and reorganize their cytoskeleton in gastric carcinoma, promoting metastasis. This occurs because COL1 decreases adhesion between cells by disassembling the E-cadherin/catenin complex, which is known to be involved in the process of cell adhesion [147,148]. Furthermore, in pancreatic ductal adenocarcinoma, COL1 interacts with Discoidin Domain Receptor 1 (DDR1) [149]. When active, DDRs trigger cellular signals in tumors that affect various functions, including extracellular matrix homeostasis, tumor cell proliferation, and migration [150]. For instance, when DDR1 binds COLI, the proliferation and invasion of tumor cells are promoted through the activation of FAK-related protein tyrosine kinase 2 (PYK2), which consequently downstream activates the Crk-associated substrate (CAS)/Ras-Proximate-1 (RAP1)/c-Jun N-terminal Kinases (JNK1)/c-Jun cascade. Moreover, the cleavage of COL1 by MMPs differently affects the bioenergetics and growth of pancreatic ductal adenocarcinoma via DDR1: matrix-metalloprotease-cleaved COL1 (cCOL1) activates a signaling cascade that promotes tumor growth, while intact COL1 (iCOL1) induces DDR1 degradation, resulting in opposite effects. Patients with enriched iCOL1 exhibit better survival compared to those with tumors enriched in cCOL1 and DDR1, suggesting that targeting the DDR1/NF-κB/Nuclear Factor Erythroid 2-Related factor 2 (NRF2)/Mitochondrial Transcription Factor A (TFAM) pathway could have therapeutic benefits [151]. In addition, COL1 is implicated in the promotion of epithelial–mesenchymal transition (EMT), a key process in embryonic development characterized by the loss of epithelial markers by epithelial cells [152]. Although typically inactive in adults, EMT has been associated with tumor invasion and metastasis. COL1-mediated phosphorylation of IκB is one pathway through which EMT is activated, leading to increased expression of the transcription factors Snail and Lymphoid Enhancer Binding Factor 1 (LEF-1), which decreases the expression of E-cadherin [153]. In addition, COL1 may be a rich source of exploitable metabolic fuel for cancer cells under critical nutrient-deficient conditions [154] and actively involved in tumor progression and metastasis. However, the role of type I collagen produced by CAFs can sometimes be shown to be contradictory since it can also have antitumor activity. The mechanisms underlying these opposing actions remain not entirely clear [139,155].
In contrast to type I collagen, type III collagen (COL3) has been shown to potentially play a tumor growth suppressive role [146]. For instance, in breast cancer patients, COL3 protects the tissues and organs of affected patients, and its reduction enhances the invasiveness of tumor cells. For example, in mice xenografted with breast cancer cells, COL3 inhibited tumor growth [156]. This is because type III collagen is implicated in tumor cells’ quiescence (G0 phase) maintenance. The re-entry of quiescence cancer cells into the cell cycle plays an important role in cancer recurrence. A reduction in COL3 leads not only to cell proliferation but also amplifies the metastatic potential of cancer cells, through modulation of the DDR1/STAT1 pathway [48]. However, the concept of quiescence in cancer remains a matter of debate. Indeed, although a state of quiescence might seemingly be a tumor progression obstacle, some studies claim that in some cases this would represent a defense mechanism from drugs. For example, cancer cells stressed by the presence of drugs enter a long persistence phase, where they can acquire chemoresistance. This drug-tolerance state is reversible as dormant tumor cells can reactivate their proliferation after drug removal; however, this is a process that increases sensitivity [157]. A further study reported that a type III humanized recombinant collagen (rhCOLIII) inhibited breast cancer cell proliferation and invasion in vitro, as well as promoting apoptosis. In addition, rhCOLIII treatment increased DDR1 protein expression, inhibiting autophagy of tumor cells [158]. This is because autophagy regulates cancer stem cell properties and contributes to the maintenance of stemness, development of relapse, and drug resistance [159].
Similar to type I collagen, type V collagen (COL5) created great interest in the scientific community, due to its involvement in tumorigenesis. In particular, type V collagen alpha 2 (COL5A2) has been observed to be associated with gastric cancer progression since this was found to be upregulated in affected patients. COL5A2 has been shown to play a direct role in cell proliferation of gastric cancer cells, where triggering the FAK/PI3K/Akt/mammalian Target Of Rapamycin (mTOR) signaling pathway [160,161]. It is also implicated in epithelial–mesenchymal transition in gastric cancer patients with poor prognosis, by promoting the expression of mesenchymal markers (SNAI1, SNAI2, TWIST, VIM, and MMP2) [162].
Among fibrillar collagens, type XI collagen (COL11) is one of the most involved in the development of several types of cancers, such as papillary thyroid carcinoma, breast cancer, colorectal carcinoma, esophageal cancer, gastric cancer, pancreatic cancer, lung cancer, and ovarian cancer [63,163,164,165,166,167,168,169]. Misregulated secretion of COL11 by CAFs creates a crosstalk between tumor cells and the TME, promoting proliferation, drug resistance, and tumor cell invasion in patients with poor prognosis [170,171,172]. In this context, it has been analyzed that integrin α1β1 and DDR2 act as extracellular Collagen 11 Alpha 1 Chain (COL11A1) receptors, triggering some signaling pathways related to ovarian cancer progression, such as Akt/CCAAT-Enhancer-Binding Proteins (C/EBPβ)/Phosphoinositide-Dependent Kinase-1 (PDK1) [173,174]. Additionally, in mouse models of xenograft with ovarian cancer cells, COL11A1 has been shown to stimulate IL-6 production via induction of TGF-β3, modulating the interaction between CAFs and tumor cells and amplifying tumor invasiveness and progression [172].
A recent study focused on the role of the gene that encodes type XXVII collagen, rather than the role of the collagen itself. Specifically, through an in vitro and in vivo analysis, the study examined the role of a specific miRNA (miRNA-455-3p) located on the gene encoding intron of Alpha Chain 1 of type XXVII collagen (COL271A). Notably, it was observed that in melanoma cells, this miRNA is closely involved in tumor cell dormancy, and its downregulation increased the re-entry of quiescent tumor cells into the cell cycle [175].

7.2. Fibrils-Associated Collagens with Interrupted Triple Helices (FACITs)

Among nonfibrillar collagens, collagens associated with interrupted triple-helix fibrils (FACITs) represent a group of small leucine-rich proteoglycans involved in regulating fibril formation [176]. Unlike fibrillar collagens that form long fibers, FACITs have interruptions in their triple-helix structure. These discontinuities are due to the presence of domains within the collagen molecule that can vary in length and composition, providing binding sites for other components of the extracellular matrix, such as fibrillar collagens [177,178]. Some collagens, such as types XII and XIV, are typically involved in stabilizing the extracellular matrix fibrils, working synergistically with type I. However, the specific organization is disrupted in the tumor microenvironment, resulting in a fibrotic and heterogeneous architecture that plays a functional role in tumorigenesis [59,100,101]. For this reason, several studies have been carried out on the distinct genes encoding for cancer-involved FACITs. For instance, type XII collagen alpha 1 chain (COL12A1) is implicated in cancer cell progression and metastasis [179] and it has been identified as a potential marker of poor prognosis in patients with breast cancer, colorectal cancer, pancreatic adenocarcinoma, and ovarian cancer. Furthermore, recent studies of temporal changes in breast cancer matrisome profile suggest that high levels of collagen XII in primary tumor ECM can facilitate the spread of lung cancer cells [180]. COL12A1 upregulation has also been correlated with metastasis, such as lymph nodes, and reduced patients’ survival with human epidermal growth factor receptor 2 (HER2)-positive breast cancer [181]. Moreover, COL12A1 expression in colorectal cancer has been linked with the regulation of several pathways, such as focal adhesion, and the PI3K-Akt pathway [182]. In patients with poor prognosis pancreatic adenocarcinoma, W. Yao et al. analyzed the relationship between COL12A1 and cytoplasmic poly (A) binding protein-1 (PABPC1), a class of RNA-binding proteins known to modulate tumor progression. PABPC1 acts as an oncogene, accelerating the proliferation and metastasis of human pancreatic cancer cell line BXPC3 through the upregulation of COL12A1 expression [183].
Type XVI Collagen (COL16) has been observed to be upregulated in certain tumor types, such as colorectal cancer (CRC), glioblastoma, and in some dysplastic areas of the mucosal epithelium of patients with oral squamous cell carcinoma [184,185]. In glioblastoma, Col16 expression influences the integrin β1 activation pattern, increasing the invasive phenotype of tumor cells [186].
Vice versa, type XIV (COL14) and type XIX (COL19) collagens exert a suppressive role in tumor progression and cell invasion. Indeed, in MMP14Sf-/- mice, the accumulation of Col14 in peritumoral areas of melanoma is correlated with a reduction in malignancy [187]. Similarly, the NC1 domain of Col19, inhibits tumor cell migration and invasion, as seen on SK-MEL-28 human melanoma cells. NC1 inhibits the FAK/PI3K/Akt/mTOR pathway, reducing the activity of proteins involved in transduction [188].
Information regarding the impact of type XX, XXI, and XXII collagens in tumors is limited. However, recent studies have shown that pancreatic ductal adenocarcinoma patients present elevated levels of circulating type XX and type XXII collagens in their serum, suggesting a possible prognostic value [189]. Another study focused on identifying hub genes and pathways in gastric carcinoma development, found that type XXI collagen alpha chain 1 (Col21a1) might have a direct role in the enrichment of some pathways involved in tumorigenesis, such as DNA repair and KRAS signaling pathway [190].

7.3. Network-Forming Collagens

Among the network-forming collagens, type IV collagen is the most prevalent in basement membranes, is approximately 400 nm in length, and assembles irregularly when forming networks. In contrast, type VIII and X collagens are shorter and form more regular hexagonal networks [191]. The expression mechanisms of network-forming collagens can be regulated by several transcription factors and play a crucial role in the development of solid tumors [143]. Type IV collagen has recently been studied in the ECM of several solid tumors. In mammals, there are six homologous α type IV collagen chains (α1(IV)–α6(IV)) that constitute both the heterotrimeric type IV collagen α1α1α2(IV) and the minor type IV collagens α3α4α5(IV) and α5α5α6(IV). As well as type I collagen, type IV is the most represented in the solid tumor ECM [192]. More specifically, collagen IV reorients within the ECM of various solid tumors, such as hepatocellular carcinoma, breast cancer, and colorectal cancer, conferring tissue stiffness around neoplastic nests. The peritumoral architecture, driven by the dense deposition of collagen IV and the aligned bundling of fibers resulting from the synergistic activation of proteases and lysyl oxidase, creates fibrotic matrix spots with abnormal mechanical properties. These spots attract tumor cells to migrate toward them through a process known as durotaxis [193,194,195]. This, in turn, strengthens the action of integrins and cytoskeletal interactions, which positively modulate the aggressive phenotype of tumor cells [196].
Moreover, during embryonic development, COL IV plays a crucial role in supporting the architecture of the vascular basement membrane, particularly in maintaining the structural integrity of small vessels, mediated by the non-catalytic activity of lysyl oxidase-like protein-2 (LOXL2) [197,198]. However, increased COL IV in TEM has a controversial role, as it may negatively or positively impact tumor angiogenesis [199,200].
For instance, some studies have shown that certain peptides derived from the NC1 domains of COL IV used for cancer therapy, such as Arresten (α1 chain), Canstatin (α2 chain), Tumstatin (α3 chain), and Tetrastatin (α4 chain), suppress vessel formation and cell growth in solid tumor [201,202,203]. These peptides, called collagen-derived antiangiogenic factors (CDAFs), play a critical role in inhibiting angiogenesis by interacting with specific integrin subtypes, influencing neovascularization in different ways [204,205]. Arresten’s activity is regulated by p53, as it controls both its transcription and MMP-dependent remodeling activity [206]. Moreover, the activity of Arresten and Canstatin can directly or indirectly influence the pro-angiogenic effects of VEGF or basic fibroblast growth factor [207]. Canstatin can decrease phosphatidylinositol 3-kinase/Akt signaling activity, downregulating VEGF activity, while Arresten can also sensitize endothelial cells to apoptosis by reducing the levels of the anti-apoptotic protein FADD-like IL-1β-converting enzyme)-inhibitory protein (FLIP) [208,209,210]. However, a recent study indicates that COL4A1 matrix expression and its heightened interaction with integrin beta-1 (ITGB1) in the endothelial cells of the tumor microenvironment is correlated with increased angiogenesis and chemoresistance, raising further questions about COL IV’s role in tumor angiogenesis [211].
Beyond influencing vessel development, type IV collagen is widely recognized for its involvement in various tumor processes. It is one of the most extensively studied collagens in the solid tumor ECM. For instance, in luminal breast carcinoma, COLIVα5 promotes tumor proliferation through the non-integrin receptor DDR1. Through interaction with DDR1, COLIVα5 supports the Warburg effect of tumor cells by influencing c-Myc phosphorylation and regulation of p38 Mitogen-Activated Protein Kinase (MAPK), which are essential for breast cancer progression [212]. Moreover, overexpression of type IV collagen mRNA was found not only in human breast cancer tissue but also in metastatic tissues of poor survival patients [213]. Therefore, this type of collagen could also stimulate prometastatic mechanisms. For this reason, similar to type XX, XXI, and XXII collagens, type IV collagen in the serum of patients with colorectal tumors may be a potential biomarker for liver metastases [214].
Type VIII collagen (COL8) is involved in tumor cell invasion and migration, as well as drug resistance [149]. Its expression is determined by tissue hypoxia, and it correlates with the aggressiveness of several solid tumors, as seen in MDA-MB-231 and Hs578T, two cellular subtypes of triple-negative breast cancer [215]. COLVIII regulates various signaling pathways, including the interaction between DDR1 and COL VIII, which promotes pancreatic ductal adenocarcinoma progression, through PI3K-Akt and FAK-NF-κB upregulation [216]. In this context, inhibiting COLVII expression in vitro and in vivo using lentivirus or siRNA has been found to reduce the metastatic potential and gemcitabine resistance of pancreatic ductal adenocarcinoma cells [217].
Finally, type X collagen is associated with a poor prognosis in patients with several types of solid tumors, such as breast cancer, colorectal cancer, gastric cancer, and lung adenocarcinoma [218,219,220]. In lung adenocarcinoma patients with poor prognosis, COL10A1 is upregulated and could physically interact with DDR2 to maintain a high level of FAK expression [221]. In addition, the direct involvement of COLX in epithelial–mesenchymal transition has been shown by analysis of tissues isolated from patients with aggressive forms of colorectal cancer [219].

7.4. Beaded Filament-Forming Collagens

Beaded filament-forming collagens are a family that includes type VI, XXVI, and XXVIII collagens. The beaded filaments appear as flexible, unbranched structures with a width of approximately 3 nm and a length of at least 2 microns [222]. Among them, type VI collagen (COL6) is a crucial component of the extracellular matrix, located in the basal membrane. Its main isoforms include the chains α1 (VI), α2 (VI), and α3 (VI). Moreover, there are three additional subunits (COL6A4, COL6A5, and COL6A6) that have been identified subsequently [223]. COL6 forms a microfilamentous network that structurally supports cells and interacts with other ECM molecules, affecting their biological functions, such as tumor response. In solid tumors, acting as a scaffold between different types of collagens and cellular receptors such as integrins, COL6 is involved in the formation of new blood vessels through the assembly of the basement membrane, a unique feature among collagens and a fundamental process in tumorigenesis. The expression levels of collagen VI vary in different cancers such as glioblastoma, ovarian cancer, and pancreatic ductal adenocarcinoma, of which different expression is probably cancer-dependent [224,225]. For example, the increased expression of COL6 is related to the aggressiveness of tumor cells, as seen in bladder cancer of high-grade T1 patients [226]. COL6, mainly produced by fibroblasts and adipocytes, supports tumor stability, and transcriptional program maintenance is related to the aggressiveness of glioblastoma cells. For this reason, its silencing in glioblastoma cells causes a downregulation of genes related to tumor progression and DNA repair mechanisms, including PI3K/AKT, Insulin-like Growth Factor 1(IGF1), Fms-like Tyrosine kinase 3 (FLT3), Platelet-Derived Growth Factor (PDGF), and MAPK [227]. In addition, pancreatic ductal adenocarcinoma cells isolated from areas of low-rigidity tumor tissue have been seen to increase COL6 expression, which has promoted in vitro motility and in vivo metastatic potential of the tumor cells [228]. Similarly, in lung sarcoma metastases, a high expression of COL6 has been detected, suggesting that it is critical for the development of metastatic niches [229].
To date, the roles of cancer of type XXVI collagen (COL26) and type XXVIII collagen (COL28) in cancer remain poorly understood. A recent study showed that in patients with poor prognosis thyroid carcinoma, there was an overexpression of the alpha 1 chain of type XXVI collagen (COL26A1). However, the signaling pathways in which it is involved remain unclear [230]. Moreover, the serum collagen levels of type XXVIII were found to be high in patients with lung cancer, but also in this case the roles are not known yet [231].

7.5. Anchoring Fibrils Collagens

Anchoring fibrils collagens are fibrous structures located in the lamina of different external epithelia that allow the attachment of the epidermal basement membrane to the dermal extracellular matrix [216]. Type VII collagen (COL7) is the only component of this group and its role in tumorigenesis is little understood [232]. Some studies show that hyperexpression promotes tumor progression since this type of collagen creates a scaffold between stroma and tumor epithelium [233]. However, it has been shown that its reduction during the advanced stages of the disease is associated with greater invasiveness of various solid tumors. For example, immunohistochemical studies have demonstrated that COL7A1 is highly expressed in the stroma surrounding tumor cells in solid tumors, such as moderately and poorly differentiated gastric adenocarcinomas, thereby promoting neoplastic processes [234,235]. Moreover, hypomorphic mice with reduced Col7 expression exhibited heightened tumor invasiveness in oral squamous cell carcinoma, suggesting a possible causal link. In addition, the COL7 absence in keratinocytes increases the expression of lysosomal proteases and matrix metalloproteinases, promoting cell invasion [236]. Moreover, in gastric carcinoma immunohistochemistry, it was observed that expression of type VII collagen alpha chain (COL7A1) was higher in tumor tissue than in healthy tissue, mainly localized in the extracellular matrix [234]. An increase in COL7 in the initial stages of the disease may be functional to the progression. On the other hand, its reduction during the more advanced phases can increase the invasive and metastatic potential of the cancer cells of some solid tumors, such as oral carcinoma.

7.6. Transmembrane Collagens

Transmembrane collagen proteins are trimers of α chains and exhibit a characteristic type II membrane protein structure. They are involved in various functions, including cell adhesion and epithelial–mesenchymal interactions [237]. This subgroup includes type XIII, XVII, XXIII, and XXV collagens. Among these, the role of type XVII collagen (COL17), a component of hemidesmosome, is particularly controversial. This is because its presence may be associated with a risk factor for patients, depending on the cancer type. In particular, the transmembrane COL17 plays a crucial role in anchoring undifferentiated keratinocytes to hemidesmosomes, while its expression is reduced in mature keratinocytes [238,239]. However, its expression significantly increases in melanomas and squamous cell carcinomas, where its deposition at the vertical fronts of the tumor is statistically associated with tumor cell invasiveness [152,153]. Moreover, a high expression of type 17 collagen alpha 1 chain (COL17A1) may be associated with a positive course for breast cancer patients, while it could be an unfavorable prognostic marker for pancreatic cancer patients [240,241]. A recent study showed how COL17 negatively affects the growth and proliferation of breast cancer cells. In particular, the tumor progression was inversely proportional to the expression of COL17, both in MCF7 and MDA-MB-231 cells and in xenotransplanted mice with overexpressed COL17 cells. COL17 inhibited the AKT/mTOR pathway and consequently reduced activation of downstream effectors such as p70S6K and 4EBP1, essential for tumor progression [242]. Furthermore, its role in colorectal cancer has also been analyzed. The absence of COL17A1 positively affected the cell cycle of dormant colorectal cancer stem cells (LGR5p27), leading to a poor prognosis for patients. More specifically, chemotherapy exposure caused a remodeling of the ECM involving COL17A1 degradation. In this way, the absence of COL17A1 promotes the FAK-YAP pathway, restoring the proliferation of dormant LGR5p27 [243]. Finally, COL17A1 can be used as a tumor biomarker, since serum COL17 levels were higher in patients with different solid tumors, including pancreatic cancer, compared to healthy controls [244].
The involvement of type XIII, type XXIII, and type XXV collagens in cancer ECM has been marginally analyzed. However, most recent studies analyze the involvement of type XIII collagen (COL13) in the invasion, progression, and migration of triple-negative breast cancer cells (MDA-MB-231). COL13 interacts with the integrin β1 and active downstream TGF-β contributing to tumor progression, as seen also in the mammosphere. In addition, COL13 boosts staminality and anoikis resistance of breast cancer cells, during the migration process [245]. Anoikis is apoptosis induced by the detachment of cells from the origin environment, through the disassembly of integrin–ECM and cadherin–cell [246]. Resistance to this mechanism plays a fundamental role in the circulating survival of cancer cells with migratory abilities [247].
Recent research has investigated the role of the α1 chain of collagen XXIII (COL23A1) in clear cell renal cell carcinoma (ccRCC). Analysis of tissues from patients with ccRCC with poor prognosis showed that this molecule enhances the progression, adhesion, and migration of cancer cells. Furthermore, knockdown underregulated the adhesion, proliferation, and migration of ccRCC cells, suggesting that COL23A1 plays a crucial role in this context [248].
Finally, the role of type XXV collagen in tumors is not well understood. There is only one study (2022) that identified for the first time the presence of alpha-1 collagen chain type XXV (Col25a1) in the ECM of murine breast tumors, while its expression was not found in the corresponding healthy tissue and human breast cancer tissue [249]. Consequently, its association with healthy and tumor ECM remains unclear.

7.7. Multiplexine

Multiplexins are basement membrane-associated collagens, including type XV (COL15) and type XVIII (COL18) collagens [250]. Despite the structural similarity and localization in the basal membrane, type XV and type XVIII collagens have distinct biological functions. COL15 is involved in the development of muscles and microvessels, while COL18 is in the development of the brain and eyes [251]. Their functions in tumorigenesis have been poorly understood. However, in the past decade, a possible role of COL15 has been identified as a cervical cancer suppressor [252]. Moreover, recent studies have shown its direct involvement in the suppression of hepato-carcinoma cells. More specifically, the silencing of the type XV collagen alpha chain (COL15A1), obtained by small interfering RNA, significantly enhanced the growth, invasion, and migration of hepato-carcinoma cells induced by the hepatitis B virus [253]. In addition, it has been shown that COL15 could impede hepato-carcinoma cell migration by underregulating the transcription factor Snail/Slug-mediated EMT [254].
In contrast to COL15, COL18 seems to have a positive role in the growth of solid tumors, such as in clear cell renal cell carcinoma and breast cancer [255]. COL18 interacts with the receptor tyrosine kinase of epidermal growth factor and with integrin α6 in breast cancer. The TSP-1 N-terminal domain of COL18 interacts with its receptors, triggering the downstream activation of MAPK/ERK and PI3/AKT pathways. In this way, COLXVIII supports the proliferation and migration of clear cell renal cell carcinoma [256].
Finally, the literature currently lacks the role of type II, IX, and type XXIV collagen in tumor ECM.
The various roles of specific tumor collagens are listed in Table 1.

8. Conclusions

Collagen is the most common protein in mammals and is the main component of the extracellular matrix. In normal tissues, a dynamic biophysical and biochemical interaction is established between collagen fibers and resident fibroblasts, where tensile and rebound energy in tissue is translated into the activation of appropriate mechanical transduction pathways [27,120,258]. Therefore, the structural changes that collagen undergoes are not only limited to the physical concept of the tissue but also modulate the biochemistry of the cells that compose it. However, these mechanisms are also critical for the development of various diseases, such as cancer [12,259].
In this context, a continuous crosstalk is established between collagen fibers within the tumor microenvironment and cancer cells. Structural modifications and variations in matrix collagens are perceived by cancer cells, activating mechanotransduction pathways, which modulate tumor progression, invasion, and migration.
For instance, type I collagen promotes epithelial–mesenchymal transition, thereby enhancing cancer cell invasion. Other collagens, such as types V, VI, VIII, and XII, facilitate tumor progression by upregulating the PI3K/Akt signaling pathway. In addition, collagen can modulate the immune system activity, contributing to tumor progression [94], acting as a physical barrier to the infiltration of the immune cells. However, some collagens can suppress these mechanisms in tumors. For instance, a reduction in type III collagen in ECM breast cancer is associated with a poor prognosis. Type XV collagen has also been identified as a tumor suppressor. This is due to its action upstream of EMT-related signaling pathways by downregulating transcription factors Snail/Slug. Furthermore, some peptide fragments derived from collagen type IV can inhibit neo-angiogenesis in tumors, with possible therapeutic applications [204].
Unfortunately, significant data on the role of type II, IX, and XXIV collagens in the extracellular matrix of tumors are still lacking in current scientific research. While other collagen types have been studied over the years, their impact on solid tumors remains uncertain. This review provides an overview of the roles of various collagen types in the extracellular matrix of solid tumors. A deeper understanding of these molecules could lead to significant advancements in the diagnosis, prognosis, and treatment of solid tumors. In recent decades, the use of collagenases in therapeutic applications—both as free enzymes and when conjugated to nanoparticles—has gained attention in the scientific community. These enzymes have the potential to break down the collagen-rich extracellular matrix of tumors, facilitating their treatment. The proteolytic activity of collagenases can improve the permeability of tumors to chemotherapeutic agents and immune cells, resulting in enhanced effectiveness of various cancer therapies. In summary, these innovative approaches represent a new frontier in the treatment of solid tumors, with collagen serving as a primary therapeutic target [260,261,262].

Author Contributions

G.L.B. and A.L.C., writing—original draft preparation; S.C. and G.G., writing—review and editing; G.G., supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Italian Ministry of University and Research (MUR) for the PON project on Research and Innovation 2014–2020 (Azione IV.4—Contratti di ricerca su tematiche dell’Innovazione—B75F21002190001). This work has received funding from the European Union-NextGenerationEU through the Italian Ministry of University and Research under PNRR–MUR, PNRR-M4C2, ECS_00000022-SiciliAn MicronanOTecH Research And Innovation CEnter “SAMOTHRACE”—CUP: B73C22000810001. The views and opinions expressed are those of the authors only and do not necessarily reflect those of the European Union or the European Commission. Neither the European Union nor the European Commission can be held responsible for them.

Data Availability Statement

Not applicable.

Conflicts of Interest

Author Giulio Ghersi was employed by the company Abiel srl. The authors declare that this research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Holwerda, A.M.; van Loon, L.J.C. The Impact of Collagen Protein Ingestion on Musculoskeletal Connective Tissue Remodeling: A Narrative Review. Nutr. Rev. 2022, 80, 1497–1514. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, Q.; Sioud, M. Tumor-Associated Macrophage Subsets: Shaping Polarization and Targeting. Int. J. Mol. Sci. 2023, 24, 7493. [Google Scholar] [CrossRef] [PubMed]
  3. Yuan, Z.; Li, Y.; Zhang, S.; Wang, X.; Dou, H.; Yu, X.; Zhang, Z.; Yang, S.; Xiao, M. Extracellular Matrix Remodeling in Tumor Progression and Immune Escape: From Mechanisms to Treatments. Mol. Cancer 2023, 22, 48. [Google Scholar] [CrossRef] [PubMed]
  4. Prasad, N.; Topping, R.S.; Decker, S.J. Src Family Tyrosine Kinases Regulate Adhesion-Dependent Tyrosine Phosphorylation of 5′-Inositol Phosphatase SHIP2 during Cell Attachment and Spreading on Collagen I. J. Cell Sci. 2002, 115, 3807–3815. [Google Scholar] [CrossRef]
  5. Theocharis, A.D.; Skandalis, S.S.; Gialeli, C.; Karamanos, N.K. Extracellular Matrix Structure. Adv. Drug Deliv. Rev. 2016, 97, 4–27. [Google Scholar] [CrossRef]
  6. Sun, B. The Mechanics of Fibrillar Collagen Extracellular Matrix. Cell Rep. Phys. Sci. 2021, 2, 100515. [Google Scholar] [CrossRef]
  7. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  8. Piersma, B.; Hayward, M.-K.; Weaver, V.M. Fibrosis and Cancer: A Strained Relationship. Biochim. Biophys. Acta—Rev. Cancer 2020, 1873, 188356. [Google Scholar] [CrossRef]
  9. Conklin, M.W.; Eickhoff, J.C.; Riching, K.M.; Pehlke, C.A.; Eliceiri, K.W.; Provenzano, P.P.; Friedl, A.; Keely, P.J. Aligned Collagen Is a Prognostic Signature for Survival in Human Breast Carcinoma. Am. J. Pathol. 2011, 178, 1221–1232. [Google Scholar] [CrossRef]
  10. Sarwar, M.; Sykes, P.H.; Chitcholtan, K.; Evans, J.J. Collagen I Dysregulation Is Pivotal for Ovarian Cancer Progression. Tissue Cell 2022, 74, 101704. [Google Scholar] [CrossRef]
  11. Nelson, M.S.; Liu, Y.; Wilson, H.M.; Li, B.; Rosado-Mendez, I.M.; Rogers, J.D.; Block, W.F.; Eliceiri, K.W. Multiscale Label-Free Imaging of Fibrillar Collagen in the Tumor Microenvironment; Springer: Berlin/Heidelberg, Germany, 2023; pp. 187–235. [Google Scholar]
  12. Xu, S.; Xu, H.; Wang, W.; Li, S.; Li, H.; Li, T.; Zhang, W.; Yu, X.; Liu, L. The Role of Collagen in Cancer: From Bench to Bedside. J. Transl. Med. 2019, 17, 309. [Google Scholar] [CrossRef] [PubMed]
  13. Du, H.; Pang, M.; Hou, X.; Yuan, S.; Sun, L. PLOD2 in Cancer Research. Biomed. Pharmacother. 2017, 90, 670–676. [Google Scholar] [CrossRef] [PubMed]
  14. Poonja, S.; Forero Pinto, A.; Lloyd, M.C.; Damaghi, M.; Rejniak, K.A. Dynamics of Fibril Collagen Remodeling by Tumor Cells: A Model of Tumor-Associated Collagen Signatures. Cells 2023, 12, 2688. [Google Scholar] [CrossRef] [PubMed]
  15. Necula, L.; Matei, L.; Dragu, D.; Pitica, I.; Neagu, A.; Bleotu, C.; Diaconu, C.C.; Chivu-Economescu, M. Collagen Family as Promising Biomarkers and Therapeutic Targets in Cancer. Int. J. Mol. Sci. 2022, 23, 12415. [Google Scholar] [CrossRef]
  16. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the Extracellular Matrix in Development and Disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  17. Sorushanova, A.; Delgado, L.M.; Wu, Z.; Shologu, N.; Kshirsagar, A.; Raghunath, R.; Mullen, A.M.; Bayon, Y.; Pandit, A.; Raghunath, M.; et al. The Collagen Suprafamily: From Biosynthesis to Advanced Biomaterial Development. Adv. Mater. 2019, 31, 1801651. [Google Scholar] [CrossRef]
  18. Hinz, B. Myofibroblasts. Exp. Eye Res. 2016, 142, 56–70. [Google Scholar] [CrossRef]
  19. Zhang, M.; Serna-Salas, S.; Damba, T.; Borghesan, M.; Demaria, M.; Moshage, H. Hepatic Stellate Cell Senescence in Liver Fibrosis: Characteristics, Mechanisms and Perspectives. Mech. Ageing Dev. 2021, 199, 111572. [Google Scholar] [CrossRef]
  20. Goldmann, T.; Zissel, G.; Watz, H.; Drömann, D.; Reck, M.; Kugler, C.; Rabe, K.F.; Marwitz, S. Human Alveolar Epithelial Cells Type II Are Capable of TGFβ-Dependent Epithelial-Mesenchymal-Transition and Collagen-Synthesis. Respir. Res. 2018, 19, 138. [Google Scholar] [CrossRef]
  21. van den Akker, J.; Tuna, B.G.; Pistea, A.; Sleutel, A.J.J.; Bakker, E.N.T.P.; van Bavel, E. Vascular Smooth Muscle Cells Remodel Collagen Matrices by Long-Distance Action and Anisotropic Interaction. Med. Biol. Eng. Comput. 2012, 50, 701–715. [Google Scholar] [CrossRef]
  22. Zunich, S.M.; Valdovinos, M.; Douglas, T.; Walterhouse, D.; Iannaccone, P.; Lamm, M.L.G. Osteoblast-Secreted Collagen Upregulates Paracrine Sonic Hedgehog Signaling by Prostate Cancer Cells and Enhances Osteoblast Differentiation. Mol. Cancer 2012, 11, 30. [Google Scholar] [CrossRef] [PubMed]
  23. RHEE, S.; GRINNELL, F. Fibroblast Mechanics in 3D Collagen Matrices. Adv. Drug Deliv. Rev. 2007, 59, 1299–1305. [Google Scholar] [CrossRef] [PubMed]
  24. Sobierajska, K.; Ciszewski, W.M.; Sacewicz-Hofman, I.; Niewiarowska, J. Endothelial Cells in the Tumor Microenvironment; Springer: Berlin/Heidelberg, Germany, 2020; pp. 71–86. [Google Scholar]
  25. Varma, S.; Orgel, J.P.R.O.; Schieber, J.D. Nanomechanics of Type I Collagen. Biophys. J. 2016, 111, 50–56. [Google Scholar] [CrossRef] [PubMed]
  26. Gordon, M.K.; Hahn, R.A. Collagens. Cell Tissue Res. 2010, 339, 247–257. [Google Scholar] [CrossRef]
  27. Silver, F.H.; Kelkar, N.; Deshmukh, T. Molecular Basis for Mechanical Properties of ECMs: Proposed Role of Fibrillar Collagen and Proteoglycans in Tissue Biomechanics. Biomolecules 2021, 11, 1018. [Google Scholar] [CrossRef]
  28. Costa, S.; Ragusa, M.A.; Lo Buglio, G.; Scilabra, S.D.; Nicosia, A. The Repertoire of Tissue Inhibitors of Metalloproteases: Evolution, Regulation of Extracellular Matrix Proteolysis, Engineering and Therapeutic Challenges. Life 2022, 12, 1145. [Google Scholar] [CrossRef]
  29. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular Matrix Degradation and Remodeling in Development and Disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  30. Siadat, S.M.; Ruberti, J.W. Mechanochemistry of Collagen. Acta Biomater. 2023, 163, 50–62. [Google Scholar] [CrossRef]
  31. Fang, M.; Yuan, J.; Peng, C.; Li, Y. Collagen as a Double-Edged Sword in Tumor Progression. Tumor Biol. 2014, 35, 2871–2882. [Google Scholar] [CrossRef]
  32. Ferreira, A.M.; Gentile, P.; Chiono, V.; Ciardelli, G. Collagen for Bone Tissue Regeneration. Acta Biomater. 2012, 8, 3191–3200. [Google Scholar] [CrossRef]
  33. Cram, D.J. The Design of Molecular Hosts, Guests, and Their Complexes. Science 1988, 240, 760–767. [Google Scholar] [CrossRef] [PubMed]
  34. Yamauchi, M.; Sricholpech, M. Lysine Post-Translational Modifications of Collagen. Essays Biochem. 2012, 52, 113–133. [Google Scholar] [CrossRef] [PubMed]
  35. Vallet, S.D.; Ricard-Blum, S. Lysyl Oxidases: From Enzyme Activity to Extracellular Matrix Cross-Links. Essays Biochem. 2019, 63, 349–364. [Google Scholar] [CrossRef] [PubMed]
  36. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of Extracellular Matrix Remodelling in Tumour Progression and Metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef]
  37. Zhang, Q.; An, Z.-Y.; Jiang, W.; Jin, W.-L.; He, X.-Y. Collagen Code in Tumor Microenvironment: Functions, Molecular Mechanisms, and Therapeutic Implications. Biomed. Pharmacother. 2023, 166, 115390. [Google Scholar] [CrossRef]
  38. Belli, C.; Trapani, D.; Viale, G.; D’Amico, P.; Duso, B.A.; Della Vigna, P.; Orsi, F.; Curigliano, G. Targeting the Microenvironment in Solid Tumors. Cancer Treat. Rev. 2018, 65, 22–32. [Google Scholar] [CrossRef]
  39. Xing, F. Cancer Associated Fibroblasts (CAFs) in Tumor Microenvironment. Front. Biosci. 2010, 15, 166. [Google Scholar] [CrossRef]
  40. Chen, Y.; McAndrews, K.M.; Kalluri, R. Clinical and Therapeutic Relevance of Cancer-Associated Fibroblasts. Nat. Rev. Clin. Oncol. 2021, 18, 792–804. [Google Scholar] [CrossRef]
  41. Hu, D.; Li, Z.; Zheng, B.; Lin, X.; Pan, Y.; Gong, P.; Zhuo, W.; Hu, Y.; Chen, C.; Chen, L.; et al. Cancer-associated Fibroblasts in Breast Cancer: Challenges and Opportunities. Cancer Commun. 2022, 42, 401–434. [Google Scholar] [CrossRef]
  42. Tian, H.; Shi, H.; Yu, J.; Ge, S.; Ruan, J. Biophysics Role and Biomimetic Culture Systems of ECM Stiffness in Cancer EMT. Glob. Chall. 2022, 6, 2100094. [Google Scholar] [CrossRef]
  43. Paszek, M.J.; Weaver, V.M. The Tension Mounts: Mechanics Meets Morphogenesis and Malignancy. J. Mammary Gland Biol. Neoplasia 2004, 9, 325–342. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, Y.; Zhang, H.; Wang, J.; Liu, Y.; Luo, T.; Hua, H. Targeting Extracellular Matrix Stiffness and Mechanotransducers to Improve Cancer Therapy. J. Hematol. Oncol. 2022, 15, 34. [Google Scholar] [CrossRef] [PubMed]
  45. Xiong, J.; Yan, L.; Zou, C.; Wang, K.; Chen, M.; Xu, B.; Zhou, Z.; Zhang, D. Integrins Regulate Stemness in Solid Tumor: An Emerging Therapeutic Target. J. Hematol. Oncol. 2021, 14, 177. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, X.; Huang, S.; Guo, J.; Zhou, L.; You, L.; Zhang, T.; Zhao, Y. Insights into the Distinct Roles of MMP-11 in Tumor Biology and Future Therapeutics (Review). Int. J. Oncol. 2016, 48, 1783–1793. [Google Scholar] [CrossRef]
  47. Wall, S.J.; Werner, E.; Werb, Z.; DeClerck, Y.A. Discoidin Domain Receptor 2 Mediates Tumor Cell Cycle Arrest Induced by Fibrillar Collagen. J. Biol. Chem. 2005, 280, 40187–40194. [Google Scholar] [CrossRef]
  48. Di Martino, J.S.; Nobre, A.R.; Mondal, C.; Taha, I.; Farias, E.F.; Fertig, E.J.; Naba, A.; Aguirre-Ghiso, J.A.; Bravo-Cordero, J.J. A Tumor-Derived Type III Collagen-Rich ECM Niche Regulates Tumor Cell Dormancy. Nat. Cancer 2021, 3, 90–107. [Google Scholar] [CrossRef]
  49. Koohestani, F.; Braundmeier, A.G.; Mahdian, A.; Seo, J.; Bi, J.; Nowak, R.A. Extracellular Matrix Collagen Alters Cell Proliferation and Cell Cycle Progression of Human Uterine Leiomyoma Smooth Muscle Cells. PLoS ONE 2013, 8, e75844. [Google Scholar] [CrossRef]
  50. Deka, J.; Wiedemann, N.; Anderle, P.; Murphy-Seiler, F.; Bultinck, J.; Eyckerman, S.; Stehle, J.-C.; André, S.; Vilain, N.; Zilian, O.; et al. Bcl9/Bcl9l Are Critical for Wnt-Mediated Regulation of Stem Cell Traits in Colon Epithelium and Adenocarcinomas. Cancer Res. 2010, 70, 6619–6628. [Google Scholar] [CrossRef]
  51. Hou, S.; Isaji, T.; Hang, Q.; Im, S.; Fukuda, T.; Gu, J. Distinct Effects of Β1 Integrin on Cell Proliferation and Cellular Signaling in MDA-MB-231 Breast Cancer Cells. Sci. Rep. 2016, 6, 18430. [Google Scholar] [CrossRef]
  52. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does It Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef]
  53. Mah, E.J.; Lefebvre, A.E.Y.T.; McGahey, G.E.; Yee, A.F.; Digman, M.A. Collagen Density Modulates Triple-Negative Breast Cancer Cell Metabolism through Adhesion-Mediated Contractility. Sci. Rep. 2018, 8, 17094. [Google Scholar] [CrossRef] [PubMed]
  54. Fontana, F.; Giannitti, G.; Marchesi, S.; Limonta, P. The PI3K/Akt Pathway and Glucose Metabolism: A Dangerous Liaison in Cancer. Int. J. Biol. Sci. 2024, 20, 3113–3125. [Google Scholar] [CrossRef] [PubMed]
  55. Yang, L.; Hou, Y.; Yuan, J.; Tang, S.; Zhang, H.; Zhu, Q.; Du, Y.; Zhou, M.; Wen, S.; Xu, L.; et al. Twist Promotes Reprogramming of Glucose Metabolism in Breast Cancer Cells through PI3K/AKT and P53 Signaling Pathways. Oncotarget 2015, 6, 25755–25769. [Google Scholar] [CrossRef] [PubMed]
  56. Mahout, M.; Schwartz, L.; Attal, R.; Bakkar, A.; Peres, S. Metabolic Modelling Links Warburg Effect to Collagen Formation, Angiogenesis and Inflammation in the Tumoral Stroma. PLoS ONE 2024, 19, e0313962. [Google Scholar] [CrossRef] [PubMed]
  57. Zheng, Y.; Zhou, R.; Cai, J.; Yang, N.; Wen, Z.; Zhang, Z.; Sun, H.; Huang, G.; Guan, Y.; Huang, N.; et al. Matrix Stiffness Triggers Lipid Metabolic Cross-Talk between Tumor and Stromal Cells to Mediate Bevacizumab Resistance in Colorectal Cancer Liver Metastases. Cancer Res. 2023, 83, 3577–3592. [Google Scholar] [CrossRef]
  58. Cai, J.; Chen, T.; Jiang, Z.; Yan, J.; Ye, Z.; Ruan, Y.; Tao, L.; Shen, Z.; Liang, X.; Wang, Y.; et al. Bulk and Single-Cell Transcriptome Profiling Reveal Extracellular Matrix Mechanical Regulation of Lipid Metabolism Reprograming through YAP/TEAD4/ACADL Axis in Hepatocellular Carcinoma. Int. J. Biol. Sci. 2023, 19, 2114–2131. [Google Scholar] [CrossRef]
  59. Morana, O.; Wood, W.; Gregory, C.D. The Apoptosis Paradox in Cancer. Int. J. Mol. Sci. 2022, 23, 1328. [Google Scholar] [CrossRef]
  60. Lv, Y.; Shan, Y.; Song, L.; Zhao, Y.; Lai, R.; Su, J.; Zhang, X. Type I Collagen Promotes Tumor Progression of Integrin Β1 Positive Gastric Cancer through a BCL9L/β-Catenin Signaling Pathway. Aging 2021, 13, 19064–19076. [Google Scholar] [CrossRef]
  61. Saby, C.; Rammal, H.; Magnien, K.; Buache, E.; Brassart-Pasco, S.; Van-Gulick, L.; Jeannesson, P.; Maquoi, E.; Morjani, H. Age-Related Modifications of Type I Collagen Impair DDR1-Induced Apoptosis in Non-Invasive Breast Carcinoma Cells. Cell Adh. Migr. 2018, 12, 335–347. [Google Scholar] [CrossRef]
  62. Maquoi, E.; Assent, D.; Detilleux, J.; Pequeux, C.; Foidart, J.-M.; Noël, A. MT1-MMP Protects Breast Carcinoma Cells against Type I Collagen-Induced Apoptosis. Oncogene 2012, 31, 480–493. [Google Scholar] [CrossRef]
  63. Tu, H.; Li, J.; Lin, L.; Wang, L. COL11A1 Was Involved in Cell Proliferation, Apoptosis and Migration in Non-Small Cell Lung Cancer Cells. J. Investig. Surg. 2021, 34, 664–669. [Google Scholar] [CrossRef] [PubMed]
  64. Nyberg, P.; Xie, L.; Sugimoto, H.; Colorado, P.; Sund, M.; Holthaus, K.; Sudhakar, A.; Salo, T.; Kalluri, R. Characterization of the Anti-Angiogenic Properties of Arresten, an A1β1 Integrin-Dependent Collagen-Derived Tumor Suppressor. Exp. Cell Res. 2008, 314, 3292–3305. [Google Scholar] [CrossRef] [PubMed]
  65. Wu, J.; Jiang, J.; Chen, B.; Wang, K.; Tang, Y.; Liang, X. Plasticity of Cancer Cell Invasion: Patterns and Mechanisms. Transl. Oncol. 2021, 14, 100899. [Google Scholar] [CrossRef] [PubMed]
  66. Yan, T.; Zhang, A.; Shi, F.; Chang, F.; Mei, J.; Liu, Y.; Zhu, Y. Integrin Avβ3–Associated DAAM1 Is Essential for Collagen-Induced Invadopodia Extension and Cell Haptotaxis in Breast Cancer Cells. J. Biol. Chem. 2018, 293, 10172–10185. [Google Scholar] [CrossRef]
  67. Artym, V.V.; Swatkoski, S.; Matsumoto, K.; Campbell, C.B.; Petrie, R.J.; Dimitriadis, E.K.; Li, X.; Mueller, S.C.; Bugge, T.H.; Gucek, M.; et al. Dense Fibrillar Collagen Is a Potent Inducer of Invadopodia via a Specific Signaling Network. J. Cell Biol. 2015, 208, 331–350. [Google Scholar] [CrossRef]
  68. Han, B.; Guan, X.; Ma, M.; Liang, B.; Ren, L.; Liu, Y.; Du, Y.; Jiang, S.-H.; Song, D. Stiffened Tumor Microenvironment Enhances Perineural Invasion in Breast Cancer via Integrin Signaling. Cell. Oncol. 2024, 47, 867–882. [Google Scholar] [CrossRef]
  69. Lee, C.S.; Siprashvili, Z.; Mah, A.; Bencomo, T.; Elcavage, L.E.; Che, Y.; Shenoy, R.M.; Aasi, S.Z.; Khavari, P.A. Mutant Collagen COL11A1 Enhances Cancerous Invasion. Oncogene 2021, 40, 6299–6307. [Google Scholar] [CrossRef]
  70. Koorman, T.; Jansen, K.A.; Khalil, A.; Haughton, P.D.; Visser, D.; Rätze, M.A.K.; Haakma, W.E.; Sakalauskaitè, G.; van Diest, P.J.; de Rooij, J.; et al. Spatial Collagen Stiffening Promotes Collective Breast Cancer Cell Invasion by Reinforcing Extracellular Matrix Alignment. Oncogene 2022, 41, 2458–2469. [Google Scholar] [CrossRef]
  71. Budden, T.; Gaudy-Marqueste, C.; Porter, A.; Kay, E.; Gurung, S.; Earnshaw, C.H.; Roeck, K.; Craig, S.; Traves, V.; Krutmann, J.; et al. Ultraviolet Light-Induced Collagen Degradation Inhibits Melanoma Invasion. Nat. Commun. 2021, 12, 2742. [Google Scholar] [CrossRef]
  72. Naba, A.; Clauser, K.R.; Lamar, J.M.; Carr, S.A.; Hynes, R.O. Extracellular Matrix Signatures of Human Mammary Carcinoma Identify Novel Metastasis Promoters. eLife 2014, 3, e01308. [Google Scholar] [CrossRef]
  73. Sahai, E.; Wyckoff, J.; Philippar, U.; Segall, J.E.; Gertler, F.; Condeelis, J. Simultaneous Imaging of GFP, CFP and Collagen in Tumors in Vivousing Multiphoton Microscopy. BMC Biotechnol. 2005, 5, 14. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, D.; Liu, Z.; Liu, W.; Fu, M.; Jiang, W.; Xu, S.; Wang, G.; Chen, F.; Lu, J.; Chen, H.; et al. Predicting Postoperative Peritoneal Metastasis in Gastric Cancer with Serosal Invasion Using a Collagen Nomogram. Nat. Commun. 2021, 12, 179. [Google Scholar] [CrossRef] [PubMed]
  75. Deng, J.; Kang, Y.; Cheng, C.-C.; Li, X.; Dai, B.; Katz, M.H.; Men, T.; Kim, M.P.; Koay, E.A.; Huang, H.; et al. DDR1-Induced Neutrophil Extracellular Traps Drive Pancreatic Cancer Metastasis. JCI Insight 2021, 6, e146133. [Google Scholar] [CrossRef] [PubMed]
  76. Akinjiyan, F.A.; Ibitoye, Z.; Zhao, P.; Shriver, L.P.; Patti, G.J.; Longmore, G.D.; Fuh, K.C. DDR2-Regulated Arginase Activity in Ovarian Cancer-Associated Fibroblasts Promotes Collagen Production and Tumor Progression. Oncogene 2024, 43, 189–201. [Google Scholar] [CrossRef]
  77. Song, M.; Schnettler, E.; Venkatachalam, A.; Wang, Y.; Feldman, L.; Argenta, P.; Rodriguez-Rodriguez, L.; Ramakrishnan, S. Increased Expression of Collagen Prolyl Hydroxylases in Ovarian Cancer Is Associated with Cancer Growth and Metastasis. Am. Cancer Res. 2023, 13, 6051. [Google Scholar]
  78. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The Role of Hypoxia in Cancer Progression, Angiogenesis, Metastasis, and Resistance to Therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef]
  79. Lin, K.; Zhao, Y.; Tang, Y.; Chen, Y.; Lin, M.; He, L. Collagen I-Induced VCAN/ERK Signaling and PARP1/ZEB1-Mediated Metastasis Facilitate OSBPL2 Defect to Promote Colorectal Cancer Progression. Cell Death Dis. 2024, 15, 85. [Google Scholar] [CrossRef]
  80. Zhang, Y.; Liu, Z.; Yang, X.; Lu, W.; Chen, Y.; Lin, Y.; Wang, J.; Lin, S.; Yun, J.-P. H3K27 Acetylation Activated-COL6A1 Promotes Osteosarcoma Lung Metastasis by Repressing STAT1 and Activating Pulmonary Cancer-Associated Fibroblasts. Theranostics 2021, 11, 1473–1492. [Google Scholar] [CrossRef]
  81. Edgar, L.T.; Underwood, C.J.; Guilkey, J.E.; Hoying, J.B.; Weiss, J.A. Extracellular Matrix Density Regulates the Rate of Neovessel Growth and Branching in Sprouting Angiogenesis. PLoS ONE 2014, 9, e85178. [Google Scholar] [CrossRef]
  82. Balcioglu, H.E.; van de Water, B.; Danen, E.H.J. Tumor-Induced Remote ECM Network Orientation Steers Angiogenesis. Sci. Rep. 2016, 6, 22580. [Google Scholar] [CrossRef]
  83. Chen, M.-C.; Hsu, W.-L.; Hwang, P.-A.; Chou, T.-C. Low Molecular Weight Fucoidan Inhibits Tumor Angiogenesis through Downregulation of HIF-1/VEGF Signaling under Hypoxia. Mar. Drugs 2015, 13, 4436–4451. [Google Scholar] [CrossRef] [PubMed]
  84. Ahluwalia, A.; Tarnawski, A.S. Critical Role of Hypoxia Sensor-HIF-1α in VEGF Gene Activation. Implications for Angiogenesis and Tissue Injury Healing. Curr. Med. Chem. 2012, 19, 90–97. [Google Scholar] [CrossRef] [PubMed]
  85. You, L.; Wu, W.; Wang, X.; Fang, L.; Adam, V.; Nepovimova, E.; Wu, Q.; Kuca, K. The Role of Hypoxia-inducible Factor 1 in Tumor Immune Evasion. Med. Res. Rev. 2021, 41, 1622–1643. [Google Scholar] [CrossRef] [PubMed]
  86. Oudart, J.-B.; Villemin, M.; Brassart, B.; Sellier, C.; Terryn, C.; Dupont-Deshorgue, A.; Monboisse, J.C.; Maquart, F.-X.; Ramont, L.; Brassart-Pasco, S. F4, a Collagen XIX-Derived Peptide, Inhibits Tumor Angiogenesis through Avβ3 and A5β1 Integrin Interaction. Cell Adh. Migr. 2021, 15, 215–223. [Google Scholar] [CrossRef] [PubMed]
  87. Mundel, T.M.; Kalluri, R. Type IV Collagen-Derived Angiogenesis Inhibitors. Microvasc. Res. 2007, 74, 85–89. [Google Scholar] [CrossRef]
  88. Ramos, A.; Sadeghi, S.; Tabatabaeian, H. Battling Chemoresistance in Cancer: Root Causes and Strategies to Uproot Them. Int. J. Mol. Sci. 2021, 22, 9451. [Google Scholar] [CrossRef]
  89. Lin, J.; Zou, B.; Li, H.; Wang, J.; Li, S.; Cao, J.; Xie, D.; Wang, F. Collagen XVII Promotes Dormancy of Colorectal Cancer Cells by Activating MTORC2 Signaling. Cell Signal. 2024, 120, 111234. [Google Scholar] [CrossRef]
  90. Saatci, O.; Kaymak, A.; Raza, U.; Ersan, P.G.; Akbulut, O.; Banister, C.E.; Sikirzhytski, V.; Tokat, U.M.; Aykut, G.; Ansari, S.A.; et al. Targeting Lysyl Oxidase (LOX) Overcomes Chemotherapy Resistance in Triple Negative Breast Cancer. Nat. Commun. 2020, 11, 2416. [Google Scholar] [CrossRef]
  91. Sharom, F.J. ABC Multidrug Transporters: Structure, Function and Role in Chemoresistance. Pharmacogenomics 2008, 9, 105–127. [Google Scholar] [CrossRef]
  92. Baltes, F.; Pfeifer, V.; Silbermann, K.; Caspers, J.; Wantoch von Rekowski, K.; Schlesinger, M.; Bendas, G. B1-Integrin Binding to Collagen Type 1 Transmits Breast Cancer Cells into Chemoresistance by Activating ABC Efflux Transporters. Biochim. Biophys. Acta—Mol. Cell Res. 2020, 1867, 118663. [Google Scholar] [CrossRef]
  93. Wu, Y.-H.; Huang, Y.-F.; Wu, P.-Y.; Chang, T.-H.; Huang, S.-C.; Chou, C.-Y. The Downregulation of MiR-509-3p Expression by Collagen Type XI Alpha 1-Regulated Hypermethylation Facilitates Cancer Progression and Chemoresistance via the DNA Methyltransferase 1/Small Ubiquitin-like Modifier-3 Axis in Ovarian Cancer Cells. J. Ovarian. Res. 2023, 16, 124. [Google Scholar] [CrossRef] [PubMed]
  94. Collado, J.; Boland, L.; Ahrendsen, J.T.; Miska, J.; Lee-Chang, C. Understanding the Glioblastoma Tumor Microenvironment: Leveraging the Extracellular Matrix to Increase Immunotherapy Efficacy. Front. Immunol. 2024, 15, 1336476. [Google Scholar] [CrossRef] [PubMed]
  95. Peng, D.H.; Rodriguez, B.L.; Diao, L.; Chen, L.; Wang, J.; Byers, L.A.; Wei, Y.; Chapman, H.A.; Yamauchi, M.; Behrens, C.; et al. Collagen Promotes Anti-PD-1/PD-L1 Resistance in Cancer through LAIR1-Dependent CD8+ T Cell Exhaustion. Nat. Commun. 2020, 11, 4520. [Google Scholar] [CrossRef] [PubMed]
  96. Rømer, A.M.A.; Thorseth, M.-L.; Madsen, D.H. Immune Modulatory Properties of Collagen in Cancer. Front. Immunol. 2021, 12, 791453. [Google Scholar] [CrossRef]
  97. Salmon, H.; Franciszkiewicz, K.; Damotte, D.; Dieu-Nosjean, M.-C.; Validire, P.; Trautmann, A.; Mami-Chouaib, F.; Donnadieu, E. Matrix Architecture Defines the Preferential Localization and Migration of T Cells into the Stroma of Human Lung Tumors. J. Clin. Investig. 2012, 122, 899–910. [Google Scholar] [CrossRef]
  98. Ohno, S.; Tachibana, M.; Fujii, T.; Ueda, S.; Kubota, H.; Nagasue, N. Role of Stromal Collagen in Immunomodulation and Prognosis of Advanced Gastric Carcinoma. Int. J. Cancer 2002, 97, 770–774. [Google Scholar] [CrossRef]
  99. Kuczek, D.E.; Larsen, A.M.H.; Thorseth, M.-L.; Carretta, M.; Kalvisa, A.; Siersbæk, M.S.; Simões, A.M.C.; Roslind, A.; Engelholm, L.H.; Noessner, E.; et al. Collagen Density Regulates the Activity of Tumor-Infiltrating T Cells. J. Immunother. Cancer 2019, 7, 68. [Google Scholar] [CrossRef]
  100. Sun, X.; Wu, B.; Chiang, H.-C.; Deng, H.; Zhang, X.; Xiong, W.; Liu, J.; Rozeboom, A.M.; Harris, B.T.; Blommaert, E.; et al. Tumour DDR1 Promotes Collagen Fibre Alignment to Instigate Immune Exclusion. Nature 2021, 599, 673–678. [Google Scholar] [CrossRef]
  101. Liu, J.; Chiang, H.-C.; Xiong, W.; Laurent, V.; Griffiths, S.C.; Dülfer, J.; Deng, H.; Sun, X.; Yin, Y.W.; Li, W.; et al. A Highly Selective Humanized DDR1 MAb Reverses Immune Exclusion by Disrupting Collagen Fiber Alignment in Breast Cancer. J. Immunother. Cancer 2023, 11, e006720. [Google Scholar] [CrossRef]
  102. Tripathi, S.; Najem, H.; Dussold, C.; Pacheco, S.; Miska, J.; McCortney, K.; Steffens, A.; Walshon, J.; Winkowski, D.; Cloney, M.; et al. Cancer-Associated Fibroblast–Secreted Collagen Is Associated with Immune Inhibitor Receptor LAIR1 in Gliomas. J. Clin. Investig. 2024, 134, e176613. [Google Scholar] [CrossRef]
  103. Rodriguez, B.L.; Huang, J.; Gibson, L.; Fradette, J.J.; Chen, H.-I.H.; Koyano, K.; Cortez, C.; Li, B.; Ho, C.; Ashique, A.M.; et al. Antitumor Activity of a Novel LAIR1 Antagonist in Combination with Anti-PD1 to Treat Collagen-Rich Solid Tumors. Mol. Cancer Ther. 2024, 23, 1144–1158. [Google Scholar] [CrossRef] [PubMed]
  104. Xie, J.; Gui, X.; Deng, M.; Chen, H.; Chen, Y.; Liu, X.; Ku, Z.; Tan, L.; Huang, R.; He, Y.; et al. Blocking LAIR1 Signaling in Immune Cells Inhibits Tumor Development. Front. Immunol. 2022, 13, 996026. [Google Scholar] [CrossRef] [PubMed]
  105. Vijver, S.V.; Singh, A.; Mommers-Elshof, E.T.A.M.; Meeldijk, J.; Copeland, R.; Boon, L.; Langermann, S.; Flies, D.; Meyaard, L.; Ramos, M.I.P. Collagen Fragments Produced in Cancer Mediate T Cell Suppression Through Leukocyte-Associated Immunoglobulin-Like Receptor 1. Front. Immunol. 2021, 12, 733561. [Google Scholar] [CrossRef] [PubMed]
  106. Nicolas-Boluda, A.; Vaquero, J.; Vimeux, L.; Guilbert, T.; Barrin, S.; Kantari-Mimoun, C.; Ponzo, M.; Renault, G.; Deptula, P.; Pogoda, K.; et al. Tumor Stiffening Reversion Through Collagen Crosslinking Inhibition Improves T Cell Migration and Anti-PD-1 Treatment. eLife 2021, 10, e58688. [Google Scholar] [CrossRef]
  107. Trackman, P.C. Lysyl Oxidase Isoforms and Potential Therapeutic Opportunities for Fibrosis and Cancer. Expert Opin. Ther. Targets 2016, 20, 935–945. [Google Scholar] [CrossRef]
  108. Li, Z.; Mo, F.; Guo, K.; Ren, S.; Wang, Y.; Chen, Y.; Schwartz, P.B.; Richmond, N.; Liu, F.; Ronnekleiv-Kelly, S.M.; et al. Nanodrug-Bacteria Conjugates-Mediated Oncogenic Collagen Depletion Enhances Immune Checkpoint Blockade Therapy against Pancreatic Cancer. Med 2024, 5, 348–367.e7. [Google Scholar] [CrossRef]
  109. Fukuda, Y.; Kim, S.-H.; Bustos, M.A.; Cho, S.-N.; Roszik, J.; Burks, J.K.; Kim, H.; Hoon, D.S.B.; Grimm, E.A.; Ekmekcioglu, S. Inhibition of Microsomal Prostaglandin E2 Synthase Reduces Collagen Deposition in Melanoma Tumors and May Improve Immunotherapy Efficacy by Reducing T-Cell Exhaustion. Cancer Res. Commun. 2023, 3, 1397–1408. [Google Scholar] [CrossRef]
  110. Zhang, X.; Liu, J.; Yang, X.; Jiao, W.; Shen, C.; Zhao, X.; Wang, Y. High Expression of COL6A1 Predicts Poor Prognosis and Response to Immunotherapy in Bladder Cancer. Cell Cycle 2023, 22, 610–618. [Google Scholar] [CrossRef]
  111. Chen, Y.; Yang, S.; Tavormina, J.; Tampe, D.; Zeisberg, M.; Wang, H.; Mahadevan, K.K.; Wu, C.-J.; Sugimoto, H.; Chang, C.-C.; et al. Oncogenic Collagen I Homotrimers from Cancer Cells Bind to A3β1 Integrin and Impact Tumor Microbiome and Immunity to Promote Pancreatic Cancer. Cancer Cell 2022, 40, 818–834.e9. [Google Scholar] [CrossRef]
  112. Saad, E.E.; Michel, R.; Borahay, M.A. Immunosuppressive Tumor Microenvironment and Uterine Fibroids: Role in Collagen Synthesis. Cytokine Growth Factor Rev. 2024, 75, 93–100. [Google Scholar] [CrossRef]
  113. Saad, E.E.; Michel, R.; Borahay, M.A. Senescence-Associated Secretory Phenotype (SASP) and Uterine Fibroids: Association with PD-L1 Activation and Collagen Deposition. Ageing Res. Rev. 2024, 97, 102314. [Google Scholar] [CrossRef] [PubMed]
  114. Tohme, S.; Yazdani, H.O.; Al-Khafaji, A.B.; Chidi, A.P.; Loughran, P.; Mowen, K.; Wang, Y.; Simmons, R.L.; Huang, H.; Tsung, A. Neutrophil Extracellular Traps Promote the Development and Progression of Liver Metastases after Surgical Stress. Cancer Res. 2016, 76, 1367–1380. [Google Scholar] [CrossRef] [PubMed]
  115. Sica, A.; Mantovani, A. Macrophage Plasticity and Polarization: In Vivo Veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef] [PubMed]
  116. Yao, R.-R.; Li, J.-H.; Zhang, R.; Chen, R.-X.; Wang, Y.-H. M2-Polarized Tumor-Associated Macrophages Facilitated Migration and Epithelial-Mesenchymal Transition of HCC Cells via the TLR4/STAT3 Signaling Pathway. World. J. Surg. Oncol. 2018, 16, 9. [Google Scholar] [CrossRef]
  117. Santoni, M.; Massari, F.; Amantini, C.; Nabissi, M.; Maines, F.; Burattini, L.; Berardi, R.; Santoni, G.; Montironi, R.; Tortora, G.; et al. Emerging Role of Tumor-Associated Macrophages as Therapeutic Targets in Patients with Metastatic Renal Cell Carcinoma. Cancer Immunol. Immunother. 2013, 62, 1757–1768. [Google Scholar] [CrossRef]
  118. Qiu, S.; Deng, L.; Liao, X.; Nie, L.; Qi, F.; Jin, K.; Tu, X.; Zheng, X.; Li, J.; Liu, L.; et al. Tumor-associated Macrophages Promote Bladder Tumor Growth through PI3K/AKT Signal Induced by Collagen. Cancer Sci. 2019, 110, 2110–2118. [Google Scholar] [CrossRef]
  119. Di, X.; Gao, X.; Peng, L.; Ai, J.; Jin, X.; Qi, S.; Li, H.; Wang, K.; Luo, D. Cellular Mechanotransduction in Health and Diseases: From Molecular Mechanism to Therapeutic Targets. Signal. Transduct. Target Ther. 2023, 8, 282. [Google Scholar] [CrossRef]
  120. Humphrey, J.D.; Dufresne, E.R.; Schwartz, M.A. Mechanotransduction and Extracellular Matrix Homeostasis. Nat. Rev. Mol. Cell Biol. 2014, 15, 802–812. [Google Scholar] [CrossRef]
  121. Martino, F.; Perestrelo, A.R.; Vinarský, V.; Pagliari, S.; Forte, G. Cellular Mechanotransduction: From Tension to Function. Front. Physiol. 2018, 9, 824. [Google Scholar] [CrossRef]
  122. Haller, S.J.; Dudley, A.T. Extracellular Mechanotransduction. J. Gen. Physiol. 2022, 154, e202113026. [Google Scholar] [CrossRef]
  123. Mousavizadeh, R.; Hojabrpour, P.; Eltit, F.; McDonald, P.C.; Dedhar, S.; McCormack, R.G.; Duronio, V.; Jafarnejad, S.M.; Scott, A. Β1 Integrin, ILK and MTOR Regulate Collagen Synthesis in Mechanically Loaded Tendon Cells. Sci. Rep. 2020, 10, 12644. [Google Scholar] [CrossRef] [PubMed]
  124. Romani, P.; Valcarcel-Jimenez, L.; Frezza, C.; Dupont, S. Crosstalk between Mechanotransduction and Metabolism. Nat. Rev. Mol. Cell. Biol. 2021, 22, 22–38. [Google Scholar] [CrossRef] [PubMed]
  125. Xin, Y.; Li, K.; Huang, M.; Liang, C.; Siemann, D.; Wu, L.; Tan, Y.; Tang, X. Biophysics in Tumor Growth and Progression: From Single Mechano-Sensitive Molecules to Mechanomedicine. Oncogene 2023, 42, 3457–3490. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, B.; Lieu, Z.Z.; Wolfenson, H.; Hameed, F.M.; Bershadsky, A.D.; Sheetz, M.P. Mechanosensing Controlled Directly by Tyrosine Kinases. Nano Lett. 2016, 16, 5951–5961. [Google Scholar] [CrossRef]
  127. Fattet, L.; Jung, H.-Y.; Matsumoto, M.W.; Aubol, B.E.; Kumar, A.; Adams, J.A.; Chen, A.C.; Sah, R.L.; Engler, A.J.; Pasquale, E.B.; et al. Matrix Rigidity Controls Epithelial-Mesenchymal Plasticity and Tumor Metastasis via a Mechanoresponsive EPHA2/LYN Complex. Dev. Cell 2020, 54, 302–316.e7. [Google Scholar] [CrossRef]
  128. Khalil, A.A.; Smits, D.; Haughton, P.D.; Koorman, T.; Jansen, K.A.; Verhagen, M.P.; van der Net, M.; van Zwieten, K.; Enserink, L.; Jansen, L.; et al. A YAP-Centered Mechanotransduction Loop Drives Collective Breast Cancer Cell Invasion. Nat. Commun. 2024, 15, 4866. [Google Scholar] [CrossRef]
  129. Wu, T.; Xiong, S.; Chen, M.; Tam, B.T.; Chen, W.; Dong, K.; Ma, Z.; Wang, Z.; Ouyang, G. Matrix Stiffening Facilitates the Collective Invasion of Breast Cancer through the Periostin-Integrin Mechanotransduction Pathway. Matrix Biol. 2023, 121, 22–40. [Google Scholar] [CrossRef]
  130. Shiu, J.-Y.; Aires, L.; Lin, Z.; Vogel, V. Nanopillar Force Measurements Reveal Actin-Cap-Mediated YAP Mechanotransduction. Nat. Cell Biol. 2018, 20, 262–271. [Google Scholar] [CrossRef]
  131. Elosegui-Artola, A.; Andreu, I.; Beedle, A.E.M.; Lezamiz, A.; Uroz, M.; Kosmalska, A.J.; Oria, R.; Kechagia, J.Z.; Rico-Lastres, P.; Le Roux, A.-L.; et al. Force Triggers YAP Nuclear Entry by Regulating Transport across Nuclear Pores. Cell 2017, 171, 1397–1410.e14. [Google Scholar] [CrossRef]
  132. Lee, J.Y.; Chang, J.K.; Dominguez, A.A.; Lee, H.; Nam, S.; Chang, J.; Varma, S.; Qi, L.S.; West, R.B.; Chaudhuri, O. YAP-Independent Mechanotransduction Drives Breast Cancer Progression. Nat. Commun. 2019, 10, 1848. [Google Scholar] [CrossRef]
  133. Miyahara, Y.; Takano, S.; Sogawa, K.; Tomizawa, S.; Furukawa, K.; Takayashiki, T.; Kuboki, S.; Ohtsuka, M. Prosaposin, Tumor-secreted Protein, Promotes Pancreatic Cancer Progression by Decreasing Tumor-infiltrating Lymphocytes. Cancer Sci. 2022, 113, 2548–2559. [Google Scholar] [CrossRef] [PubMed]
  134. Wu, Y.; Sun, L.; Zou, W.; Xu, J.; Liu, H.; Wang, W.; Yun, X.; Gu, J. Prosaposin, a Regulator of Estrogen Receptor Alpha, Promotes Breast Cancer Growth. Cancer Sci. 2012, 103, 1820–1825. [Google Scholar] [CrossRef] [PubMed]
  135. Jiang, Y.; Zhou, J.; Luo, P.; Gao, H.; Ma, Y.; Chen, Y.-S.; Li, L.; Zou, D.; Zhang, Y.; Jing, Z. Prosaposin Promotes the Proliferation and Tumorigenesis of Glioma through Toll-like Receptor 4 (TLR4)-Mediated NF-ΚB Signaling Pathway. EBioMedicine 2018, 37, 78–90. [Google Scholar] [CrossRef] [PubMed]
  136. Ishihara, S.; Inman, D.R.; Li, W.-J.; Ponik, S.M.; Keely, P.J. Mechano-Signal Transduction in Mesenchymal Stem Cells Induces Prosaposin Secretion to Drive the Proliferation of Breast Cancer Cells. Cancer Res. 2017, 77, 6179–6189. [Google Scholar] [CrossRef] [PubMed]
  137. Nasser, M.; Wu, Y.; Danaoui, Y.; Ghosh, G. Engineering Microenvironments towards Harnessing Pro-Angiogenic Potential of Mesenchymal Stem Cells. Mater. Sci. Eng. C 2019, 102, 75–84. [Google Scholar] [CrossRef]
  138. Dong, Y.; Xie, X.; Wang, Z.; Hu, C.; Zheng, Q.; Wang, Y.; Chen, R.; Xue, T.; Chen, J.; Gao, D.; et al. Increasing Matrix Stiffness Upregulates Vascular Endothelial Growth Factor Expression in Hepatocellular Carcinoma Cells Mediated by Integrin Β1. Biochem. Biophys. Res. Commun. 2014, 444, 427–432. [Google Scholar] [CrossRef]
  139. Thorlacius-Ussing, J.; Jensen, C.; Nissen, N.I.; Cox, T.R.; Kalluri, R.; Karsdal, M.; Willumsen, N. The Collagen Landscape in Cancer: Profiling Collagens in Tumors and in Circulation Reveals Novel Markers of Cancer-associated Fibroblast Subtypes. J. Pathol. 2024, 262, 22–36. [Google Scholar] [CrossRef]
  140. Ricard-Blum, S. The Collagen Family. Cold Spring Harb. Perspect. Biol. 2011, 3, a004978. [Google Scholar] [CrossRef]
  141. van der Rest, M.; Dublet, B.; Champliaud, M.F. Fibril-Associated Collagens. Biomaterials 1990, 11, 28–31. [Google Scholar]
  142. Song, K.; Yu, Z.; Zu, X.; Li, G.; Hu, Z.; Xue, Y. Collagen Remodeling along Cancer Progression Providing a Novel Opportunity for Cancer Diagnosis and Treatment. Int. J. Mol. Sci. 2022, 23, 10509. [Google Scholar] [CrossRef]
  143. Zhang, J.; Liu, J.; Zhang, H.; Wang, J.; Hua, H.; Jiang, Y. The Role of Network-forming Collagens in Cancer Progression. Int. J. Cancer 2022, 151, 833–842. [Google Scholar] [CrossRef] [PubMed]
  144. Hanley, C.J.; Noble, F.; Ward, M.; Bullock, M.; Drifka, C.; Mellone, M.; Manousopoulou, A.; Johnston, H.E.; Hayden, A.; Thirdborough, S.; et al. A Subset of Myofibroblastic Cancer-Associated Fibroblasts Regulate Collagen Fiber Elongation, Which Is Prognostic in Multiple Cancers. Oncotarget 2016, 7, 6159–6174. [Google Scholar] [CrossRef] [PubMed]
  145. Stylianopoulos, T.; Diop-Frimpong, B.; Munn, L.L.; Jain, R.K. Diffusion Anisotropy in Collagen Gels and Tumors: The Effect of Fiber Network Orientation. Biophys. J. 2010, 99, 3119–3128. [Google Scholar] [CrossRef] [PubMed]
  146. Brisson, B.K.; Mauldin, E.A.; Lei, W.; Vogel, L.K.; Power, A.M.; Lo, A.; Dopkin, D.; Khanna, C.; Wells, R.G.; Puré, E.; et al. Type III Collagen Directs Stromal Organization and Limits Metastasis in a Murine Model of Breast Cancer. Am. J. Pathol. 2015, 185, 1471–1486. [Google Scholar] [CrossRef] [PubMed]
  147. Li, A.; Zhou, T.; Guo, L.; Si, J. Collagen Type I Regulates β-Catenin Tyrosine Phosphorylation and Nuclear Translocation to Promote Migration and Proliferation of Gastric Carcinoma Cells. Oncol. Rep. 2010, 23, 1247–1255. [Google Scholar] [CrossRef]
  148. Mendonsa, A.M.; Na, T.-Y.; Gumbiner, B.M. E-Cadherin in Contact Inhibition and Cancer. Oncogene 2018, 37, 4769–4780. [Google Scholar] [CrossRef]
  149. Su, H.; Karin, M. Multifaceted Collagen-DDR1 Signaling in Cancer. Trends Cell Biol. 2024, 34, 406–415. [Google Scholar] [CrossRef]
  150. Kothiwale, S.; Borza, C.M.; Lowe, E.W.; Pozzi, A.; Meiler, J. Discoidin Domain Receptor 1 (DDR1) Kinase as Target for Structure-Based Drug Discovery. Drug Discov. Today 2015, 20, 255–261. [Google Scholar] [CrossRef]
  151. Su, H.; Yang, F.; Fu, R.; Trinh, B.; Sun, N.; Liu, J.; Kumar, A.; Baglieri, J.; Siruno, J.; Le, M.; et al. Collagenolysis-Dependent DDR1 Signalling Dictates Pancreatic Cancer Outcome. Nature 2022, 610, 366–372. [Google Scholar] [CrossRef]
  152. Chen, T.; You, Y.; Jiang, H.; Wang, Z.Z. Epithelial–Mesenchymal Transition (EMT): A Biological Process in the Development, Stem Cell Differentiation, and Tumorigenesis. J. Cell Physiol. 2017, 232, 3261–3272. [Google Scholar] [CrossRef]
  153. Medici, D.; Nawshad, A. Type I Collagen Promotes Epithelial–Mesenchymal Transition through ILK-Dependent Activation of NF-ΚB and LEF-1. Matrix Biol. 2010, 29, 161–165. [Google Scholar] [CrossRef] [PubMed]
  154. Hsu, K.-S.; Dunleavey, J.M.; Szot, C.; Yang, L.; Hilton, M.B.; Morris, K.; Seaman, S.; Feng, Y.; Lutz, E.M.; Koogle, R.; et al. Cancer Cell Survival Depends on Collagen Uptake into Tumor-Associated Stroma. Nat. Commun. 2022, 13, 7078. [Google Scholar] [CrossRef] [PubMed]
  155. Bhattacharjee, S.; Hamberger, F.; Ravichandra, A.; Miller, M.; Nair, A.; Affo, S.; Filliol, A.; Chin, L.; Savage, T.M.; Yin, D.; et al. Tumor Restriction by Type I Collagen Opposes Tumor-Promoting Effects of Cancer-Associated Fibroblasts. J. Clin. Investig. 2021, 131, e146987. [Google Scholar] [CrossRef] [PubMed]
  156. Brisson, B.K.; Stewart, D.C.; Burgwin, C.; Chenoweth, D.; Wells, R.G.; Adams, S.L.; Volk, S.W. Cysteine-Rich Domain of Type III Collagen N-Propeptide Inhibits Fibroblast Activation by Attenuating TGFβ Signaling. Matrix Biol. 2022, 109, 19–33. [Google Scholar] [CrossRef]
  157. Ramirez, M.; Rajaram, S.; Steininger, R.J.; Osipchuk, D.; Roth, M.A.; Morinishi, L.S.; Evans, L.; Ji, W.; Hsu, C.-H.; Thurley, K.; et al. Diverse Drug-Resistance Mechanisms Can Emerge from Drug-Tolerant Cancer Persister Cells. Nat. Commun. 2016, 7, 10690. [Google Scholar] [CrossRef]
  158. Liu, X.; Li, H.; Wang, T.; Yang, T.; Yang, X.; Guo, K.; Hu, L.; Ming, J. Recombinant Humanized Collagen Type III with High Antitumor Activity Inhibits Breast Cancer Cells Autophagy, Proliferation, and Migration through DDR1. Int. J. Biol. Macromol. 2023, 243, 125130. [Google Scholar] [CrossRef]
  159. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef]
  160. Guo, X.; Bu, X.; Yuan, L.; Ji, L. Collagen Type V Alpha 2 Promotes the Development of Gastric Cancer via M2 Macrophage Polarization. Chin. J. Physiol. 2023, 66, 93. [Google Scholar] [CrossRef]
  161. Lee, B.Y.; Timpson, P.; Horvath, L.G.; Daly, R.J. FAK Signaling in Human Cancer as a Target for Therapeutics. Pharmacol. Ther. 2015, 146, 132–149. [Google Scholar] [CrossRef]
  162. Jin, Y.; Song, X.; Sun, X.; Ding, Y. Up-Regulation of Collagen Type V Alpha 2 (COL5A2) Promotes Malignant Phenotypes in Gastric Cancer Cell via Inducing Epithelial–Mesenchymal Transition (EMT). Open Med. 2023, 18, 20220593. [Google Scholar] [CrossRef]
  163. Arolt, C.; Meyer, M.; Hoffmann, F.; Wagener-Ryczek, S.; Schwarz, D.; Nachtsheim, L.; Beutner, D.; Odenthal, M.; Guntinas-Lichius, O.; Buettner, R.; et al. Expression Profiling of Extracellular Matrix Genes Reveals Global and Entity-Specific Characteristics in Adenoid Cystic, Mucoepidermoid and Salivary Duct Carcinomas. Cancers 2020, 12, 2466. [Google Scholar] [CrossRef] [PubMed]
  164. Ellsworth, R.E.; Seebach, J.; Field, L.A.; Heckman, C.; Kane, J.; Hooke, J.A.; Love, B.; Shriver, C.D. A Gene Expression Signature That Defines Breast Cancer Metastases. Clin. Exp. Metastasis 2009, 26, 205–213. [Google Scholar] [CrossRef] [PubMed]
  165. Galván, J.A.; García-Martínez, J.; Vázquez-Villa, F.; García-Ocaña, M.; García-Pravia, C.; Menéndez-Rodríguez, P.; González-del Rey, C.; Barneo-Serra, L.; de los Toyos, J.R. Validation of COL11A1/Procollagen 11A1 Expression in TGF-Β1-Activated Immortalised Human Mesenchymal Cells and in Stromal Cells of Human Colon Adenocarcinoma. BMC Cancer 2014, 14, 867. [Google Scholar] [CrossRef] [PubMed]
  166. Chattopadhyay, I.; Singh, A.; Phukan, R.; Purkayastha, J.; Kataki, A.; Mahanta, J.; Saxena, S.; Kapur, S. Genome-Wide Analysis of Chromosomal Alterations in Patients with Esophageal Squamous Cell Carcinoma Exposed to Tobacco and Betel Quid from High-Risk Area in India. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2010, 696, 130–138. [Google Scholar] [CrossRef]
  167. Hao, S.; Lv, J.; Yang, Q.; Wang, A.; Li, Z.; Guo, Y.; Zhang, G. Identification of Key Genes and Circular RNAs in Human Gastric Cancer. Med. Sci. Monit. 2019, 25, 2488–2504. [Google Scholar] [CrossRef]
  168. Nizri, E.; Bar-David, S.; Aizic, A.; Sternbach, N.; Lahat, G.; Wolf, I.; Klausner, J. Desmoplasia in Lymph Node Metastasis of Pancreatic Adenocarcinoma Reveals Activation of Cancer-Associated Fibroblasts Pattern and T-Helper 2 Immune Cell Infiltration. Pancreas 2019, 48, 367–373. [Google Scholar] [CrossRef]
  169. Nallanthighal, S.; Rada, M.; Heiserman, J.P.; Cha, J.; Sage, J.; Zhou, B.; Yang, W.; Hu, Y.; Korgaonkar, C.; Hanos, C.T.; et al. Inhibition of Collagen XI Alpha 1-Induced Fatty Acid Oxidation Triggers Apoptotic Cell Death in Cisplatin-Resistant Ovarian Cancer. Cell Death Dis. 2020, 11, 258. [Google Scholar] [CrossRef]
  170. Vázquez-Villa, F.; García-Ocaña, M.; Galván, J.A.; García-Martínez, J.; García-Pravia, C.; Menéndez-Rodríguez, P.; Rey, C.G.; Barneo-Serra, L.; de los Toyos, J.R. COL11A1/(pro)Collagen 11A1 Expression Is a Remarkable Biomarker of Human Invasive Carcinoma-Associated Stromal Cells and Carcinoma Progression. Tumor Biol. 2015, 36, 2213–2222. [Google Scholar] [CrossRef]
  171. Jia, D.; Liu, Z.; Deng, N.; Tan, T.Z.; Huang, R.Y.-J.; Taylor-Harding, B.; Cheon, D.-J.; Lawrenson, K.; Wiedemeyer, W.R.; Walts, A.E.; et al. A COL11A1-Correlated Pan-Cancer Gene Signature of Activated Fibroblasts for the Prioritization of Therapeutic Targets. Cancer Lett. 2016, 382, 203–214. [Google Scholar] [CrossRef]
  172. Wu, Y.-H.; Chou, C.-Y. Collagen XI Alpha 1 Chain, a Novel Therapeutic Target for Cancer Treatment. Front. Oncol. 2022, 12, 925165. [Google Scholar] [CrossRef]
  173. Rada, M.; Nallanthighal, S.; Cha, J.; Ryan, K.; Sage, J.; Eldred, C.; Ullo, M.; Orsulic, S.; Cheon, D.-J. Inhibitor of Apoptosis Proteins (IAPs) Mediate Collagen Type XI Alpha 1-Driven Cisplatin Resistance in Ovarian Cancer. Oncogene 2018, 37, 4809–4820. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, Y.-H.; Chang, T.-H.; Huang, Y.-F.; Chen, C.-C.; Chou, C.-Y. COL11A1 Confers Chemoresistance on Ovarian Cancer Cells through the Activation of Akt/c/EBPβ Pathway and PDK1 Stabilization. Oncotarget 2015, 6, 23748–23763. [Google Scholar] [CrossRef] [PubMed]
  175. La, T.; Liu, G.Z.; Farrelly, M.; Cole, N.; Feng, Y.C.; Zhang, Y.Y.; Sherwin, S.K.; Yari, H.; Tabatabaee, H.; Yan, X.G.; et al. A P53-Responsive MiRNA Network Promotes Cancer Cell Quiescence. Cancer Res. 2018, 78, 6666–6679. [Google Scholar] [CrossRef] [PubMed]
  176. Zvackova, I.; Matalova, E.; Lesot, H. Regulators of Collagen Fibrillogenesis during Molar Development in the Mouse. Front. Physiol. 2017, 8, 554. [Google Scholar] [CrossRef] [PubMed]
  177. Vizzini, A.; Pergolizzi, M.; Vazzana, M.; Salerno, G.; Di Sano, C.; Macaluso, P.; Arizza, V.; Parrinello, D.; Cammarata, M.; Parrinello, N. FACIT Collagen (1α-Chain) Is Expressed by Hemocytes and Epidermis during the Inflammatory Response of the Ascidian Ciona Intestinalis. Dev. Comp. Immunol. 2008, 32, 682–692. [Google Scholar] [CrossRef]
  178. Coulson-Thomas, V.J.; Coulson-Thomas, Y.M.; Gesteira, T.F.; de Paula, C.A.A.; Mader, A.M.; Waisberg, J.; Pinhal, M.A.; Friedl, A.; Toma, L.; Nader, H.B. Colorectal Cancer Desmoplastic Reaction Up-Regulates Collagen Synthesis and Restricts Cancer Cell Invasion. Cell Tissue Res. 2011, 346, 223–236. [Google Scholar] [CrossRef]
  179. Chivu-Economescu, M.; Necula, L.G.; Matei, L.; Dragu, D.; Bleotu, C.; Sorop, A.; Herlea, V.; Dima, S.; Popescu, I.; Diaconu, C.C. Collagen Family and Other Matrix Remodeling Proteins Identified by Bioinformatics Analysis as Hub Genes Involved in Gastric Cancer Progression and Prognosis. Int. J. Mol. Sci. 2022, 23, 3214. [Google Scholar] [CrossRef]
  180. Papanicolaou, M.; Parker, A.L.; Yam, M.; Filipe, E.C.; Wu, S.Z.; Chitty, J.L.; Wyllie, K.; Tran, E.; Mok, E.; Nadalini, A.; et al. Temporal Profiling of the Breast Tumour Microenvironment Reveals Collagen XII as a Driver of Metastasis. Nat. Commun. 2022, 13, 4587. [Google Scholar] [CrossRef]
  181. Yan, Y.; Liang, Q.; Liu, Y.; Zhou, S.; Xu, Z. COL12A1 as a Prognostic Biomarker Links Immunotherapy Response in Breast Cancer. Endocr. Relat. Cancer 2023, 30, e230012. [Google Scholar] [CrossRef]
  182. Wu, Y.; Xu, Y. Integrated Bioinformatics Analysis of Expression and Gene Regulation Network of COL12A1 in Colorectal Cancer. Cancer Med. 2020, 9, 4743–4755. [Google Scholar] [CrossRef]
  183. Yao, W.; Yao, Y.; He, W.; Zhao, C.; Liu, D.; Wang, G.; Wang, Z. PABPC1 Promotes Cell Proliferation and Metastasis in Pancreatic Adenocarcinoma by Regulating COL12A1 Expression. Immun. Inflamm. Dis. 2023, 11, e919. [Google Scholar] [CrossRef] [PubMed]
  184. Jensen, C.; Nielsen, S.H.; Mortensen, J.H.; Kjeldsen, J.; Klinge, L.G.; Krag, A.; Harling, H.; Jørgensen, L.N.; Karsdal, M.A.; Willumsen, N. Serum Type XVI Collagen Is Associated with Colorectal Cancer and Ulcerative Colitis Indicating a Pathological Role in Gastrointestinal Disorders. Cancer Med. 2018, 7, 4619–4626. [Google Scholar] [CrossRef] [PubMed]
  185. Ratzinger, S.; Grässel, S.; Dowejko, A.; Reichert, T.E.; Bauer, R.J. Induction of Type XVI Collagen Expression Facilitates Proliferation of Oral Cancer Cells. Matrix Biol. 2011, 30, 118–125. [Google Scholar] [CrossRef] [PubMed]
  186. Bauer, R.; Ratzinger, S.; Wales, L.; Bosserhoff, A.; Senner, V.; Grifka, J.; Grässel, S. Inhibition of Collagen XVI Expression Reduces Glioma Cell Invasiveness. Cell. Physiol. Biochem. 2011, 27, 217–226. [Google Scholar] [CrossRef] [PubMed]
  187. Pach, E.; Kümper, M.; Fromme, J.E.; Zamek, J.; Metzen, F.; Koch, M.; Mauch, C.; Zigrino, P. Extracellular Matrix Remodeling by Fibroblast-MMP14 Regulates Melanoma Growth. Int. J. Mol. Sci. 2021, 22, 12276. [Google Scholar] [CrossRef]
  188. Oudart, J.-B.; Doué, M.; Vautrin, A.; Brassart, B.; Sellier, C.; Dupont-Deshorgue, A.; Monboisse, J.-C.; Maquart, F.-X.; Brassart-Pasco, S.; Ramont, L. The Anti-Tumor NC1 Domain of Collagen XIX Inhibits the FAK/PI3K/Akt/MTOR Signaling Pathway through Avβ3 Integrin Interaction. Oncotarget 2016, 7, 1516–1528. [Google Scholar] [CrossRef]
  189. Thorlacius-Ussing, J.; Jensen, C.; Madsen, E.A.; Nissen, N.I.; Manon-Jensen, T.; Chen, I.M.; Johansen, J.S.; Diab, H.M.H.; Jørgensen, L.N.; Karsdal, M.A.; et al. Type XX Collagen Is Elevated in Circulation of Patients with Solid Tumors. Int. J. Mol. Sci. 2022, 23, 4144. [Google Scholar] [CrossRef]
  190. Qin, L.H.; Zhu, X.J.; Zhang, L.Y.; Chen, J.Q.; Jin, G.Y.; Xiang, L.J. Identification of Hub Genes and Pathways in the Development of Gastric Cancer by Gene Co-expression Network Analysis. J. Biol. Regul. Homeost. Agents. 2021, 35, 35–44. [Google Scholar] [CrossRef]
  191. Knupp, C.; Squire, J.M. Molecular Packing in Network-Forming Collagens; Elsevier: Amsterdam, the Netherlands, 2005; pp. 375–403. [Google Scholar]
  192. Egeblad, M.; Rasch, M.G.; Weaver, V.M. Dynamic Interplay between the Collagen Scaffold and Tumor Evolution. Curr. Opin. Cell Biol. 2010, 22, 697–706. [Google Scholar] [CrossRef]
  193. Liang, Y.; Lv, Z.; Huang, G.; Qin, J.; Li, H.; Nong, F.; Wen, B. Prognostic Significance of Abnormal Matrix Collagen Remodeling in Colorectal Cancer Based on Histologic and Bioinformatics Analysis. Oncol. Rep. 2020, 44, 1671–1685. [Google Scholar] [CrossRef]
  194. Friedl, P.; Alexander, S. Cancer Invasion and the Microenvironment: Plasticity and Reciprocity. Cell 2011, 147, 992–1009. [Google Scholar] [CrossRef] [PubMed]
  195. Torzilli, P.A.; Bourne, J.W.; Cigler, T.; Vincent, C.T. A New Paradigm for Mechanobiological Mechanisms in Tumor Metastasis. Semin. Cancer Biol. 2012, 22, 385–395. [Google Scholar] [CrossRef] [PubMed]
  196. Levental, K.R.; Yu, H.; Kass, L.; Lakins, J.N.; Egeblad, M.; Erler, J.T.; Fong, S.F.T.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell 2009, 139, 891–906. [Google Scholar] [CrossRef] [PubMed]
  197. Pöschl, E.; Schlötzer-Schrehardt, U.; Brachvogel, B.; Saito, K.; Ninomiya, Y.; Mayer, U. Collagen IV Is Essential for Basement Membrane Stability but Dispensable for Initiation of Its Assembly during Early Development. Development 2004, 131, 1619–1628. [Google Scholar] [CrossRef] [PubMed]
  198. Bignon, M.; Pichol-Thievend, C.; Hardouin, J.; Malbouyres, M.; Bréchot, N.; Nasciutti, L.; Barret, A.; Teillon, J.; Guillon, E.; Etienne, E.; et al. Lysyl Oxidase-like Protein-2 Regulates Sprouting Angiogenesis and Type IV Collagen Assembly in the Endothelial Basement Membrane. Blood 2011, 118, 3979–3989. [Google Scholar] [CrossRef]
  199. Večurkovská, I.; Stupák, M.; Kaťuchová, J.; Roškovičová, V.; Mašlanková, J. Comparative Analysis of Matrix Metalloproteinases by Zymography in Patients With Colorectal Carcinoma. Physiol. Res. 2023, 72, S593–S596. [Google Scholar] [CrossRef]
  200. Kalluri, R. Basement Membranes: Structure, Assembly and Role in Tumour Angiogenesis. Nat. Rev. Cancer 2003, 3, 422–433. [Google Scholar] [CrossRef]
  201. Chamani, R.; Saberi, O.; Fathinejad, F. An Arresten-Derived Anti-Angiogenic Peptide Triggers Apoptotic Cell Death in Endothelial Cells. Mol. Biol. Rep. 2024, 51, 513. [Google Scholar] [CrossRef]
  202. Chamani, R.; Taleqani, M.H.; Imanpour, A.; Khatami, M. New Insights into Short Peptides Derived from the Collagen NC1 A1, A2, and A3 (IV) Domains: An Experimental and MD Simulations Study. Biochim. Biophys. Acta—Proteins Proteom. 2022, 1870, 140769. [Google Scholar] [CrossRef]
  203. Vautrin-Glabik, A.; Devy, J.; Bour, C.; Baud, S.; Choulier, L.; Hoarau, A.; Dupont-Deshorgue, A.; Sellier, C.; Brassart, B.; Oudart, J.-B.; et al. Angiogenesis Inhibition by a Short 13 Amino Acid Peptide Sequence of Tetrastatin, the A4(IV) NC1 Domain of Collagen IV. Front. Cell Dev. Biol. 2020, 8, 775. [Google Scholar] [CrossRef]
  204. Aikio, M.; Alahuhta, I.; Nurmenniemi, S.; Suojanen, J.; Palovuori, R.; Teppo, S.; Sorsa, T.; López-Otín, C.; Pihlajaniemi, T.; Salo, T.; et al. Arresten, a Collagen-Derived Angiogenesis Inhibitor, Suppresses Invasion of Squamous Cell Carcinoma. PLoS ONE 2012, 7, e51044. [Google Scholar] [CrossRef] [PubMed]
  205. Brassart-Pasco, S.; Brézillon, S.; Brassart, B.; Ramont, L.; Oudart, J.-B.; Monboisse, J.C. Tumor Microenvironment: Extracellular Matrix Alterations Influence Tumor Progression. Front. Oncol. 2020, 10, 397. [Google Scholar] [CrossRef] [PubMed]
  206. Assadian, S.; El-Assaad, W.; Wang, X.Q.D.; Gannon, P.O.; Barrès, V.; Latour, M.; Mes-Masson, A.-M.; Saad, F.; Sado, Y.; Dostie, J.; et al. P53 Inhibits Angiogenesis by Inducing the Production of Arresten. Cancer Res. 2012, 72, 1270–1279. [Google Scholar] [CrossRef] [PubMed]
  207. Kamphaus, G.D.; Colorado, P.C.; Panka, D.J.; Hopfer, H.; Ramchandran, R.; Torre, A.; Maeshima, Y.; Mier, J.W.; Sukhatme, V.P.; Kalluri, R. Canstatin, a Novel Matrix-Derived Inhibitor of Angiogenesis and Tumor Growth. J. Biol. Chem. 2000, 275, 1209–1215. [Google Scholar] [CrossRef]
  208. Panka, D.J.; Mier, J.W. Canstatin Inhibits Akt Activation and Induces Fas-Dependent Apoptosis in Endothelial Cells. J. Biol. Chem. 2003, 278, 37632–37636. [Google Scholar] [CrossRef] [PubMed]
  209. Pei, J.; Li, P.; Gao, Y.H.; Tian, B.G.; Wang, D.Y.; Zheng, Y.; Liu, L.Y.; Zhang, Z.Y.; Huang, S.S.; Wen, M.; et al. Type IV Collagen-Derived Angiogenesis Inhibitor: Canstatin Low Expressing in Brain-Invasive Meningiomas Using Liquid Chromatography–Mass Spectrometry (LC-MS/MS). J. Neurooncol. 2023, 161, 415–423. [Google Scholar] [CrossRef] [PubMed]
  210. Zhu, L.; Guo, Z.; Zhang, J.; Yang, Y.; Liu, C.; Zhang, L.; Gu, Z.; Li, Y.; Ding, Z.; Shi, G. Recombinant Human Arresten and Canstatin Inhibit Angiogenic Behaviors of HUVECs via Inhibiting the PI3K/Akt Signaling Pathway. Int. J. Mol. Sci. 2022, 23, 8995. [Google Scholar] [CrossRef]
  211. Guo, J.; Ma, X.; Liu, D.; Wang, F.; Xia, J.; Zhang, B.; Zhao, P.; Zhong, F.; Chen, L.; Long, Q.; et al. A Distinct Subset of Urothelial Cells with Enhanced EMT Features Promotes Chemotherapy Resistance and Cancer Recurrence by Increasing COL4A1-ITGB1 Mediated Angiogenesis. Drug Resist. Updates 2024, 76, 101116. [Google Scholar] [CrossRef]
  212. Wu, Y.; Liu, X.; Zhu, Y.; Qiao, Y.; Gao, Y.; Chen, J.; Ge, G. Type IV Collagen α 5 Chain Promotes Luminal Breast Cancer Progression through c-Myc-Driven Glycolysis. J. Mol. Cell Biol. 2023, 14, mjac068. [Google Scholar] [CrossRef]
  213. Jansson, M.; Lindberg, J.; Rask, G.; Svensson, J.; Billing, O.; Nazemroaya, A.; Berglund, A.; Wärnberg, F.; Sund, M. Prognostic Value of Stromal Type IV Collagen Expression in Small Invasive Breast Cancers. Front. Mol. Biosci. 2022, 9, 904526. [Google Scholar] [CrossRef]
  214. Xia, Y.; Jiang, H.; Chen, J.; Xu, F.; Zhang, G.; Zhang, D. Low Dose Taxol Ameliorated Renal Fibrosis in Mice with Diabetic Kidney Disease by Downregulation of HIPK2. Life Sci. 2023, 320, 121540. [Google Scholar] [CrossRef] [PubMed]
  215. Sato, F.; Sagara, A.; Tajima, K.; Miura, S.; Inaba, K.; Ando, Y.; Oku, T.; Murakami, T.; Kato, Y.; Yumoto, T. COL8A1 Facilitates the Growth of Triple-Negative Breast Cancer via FAK/Src Activation. Breast Cancer Res. Treat. 2022, 194, 243–256. [Google Scholar] [CrossRef] [PubMed]
  216. Bruckner-Tuderman, L.; Höpfner, B.; Hammami-Hauasli, N. Biology of Anchoring Fibrils: Lessons from Dystrophic Epidermolysis Bullosa. Matrix Biol. 1999, 18, 43–54. [Google Scholar] [CrossRef] [PubMed]
  217. Chen, S.; Wei, Y.; Liu, H.; Gong, Y.; Zhou, Y.; Yang, H.; Tang, L. Analysis of Collagen Type X Alpha 1 (COL10A1) Expression and Prognostic Significance in Gastric Cancer Based on Bioinformatics. Bioengineered 2021, 12, 127–137. [Google Scholar] [CrossRef]
  218. Huang, H.; Li, T.; Ye, G.; Zhao, L.; Zhang, Z.; Mo, D.; Wang, Y.; Zhang, C.; Deng, H.; Li, G.; et al. High Expression of COL10A1 Is Associated with Poor Prognosis in Colorectal Cancer. Onco. Targets Ther. 2018, 11, 1571–1581. [Google Scholar] [CrossRef]
  219. Wang, C.; Wang, J.; Chen, S.; Li, K.; Wan, S.; Yang, L. COL10A1 as a Prognostic Biomarker in Association with Immune Infiltration in Prostate Cancer. Curr. Cancer Drug Targets 2024, 24, 340–353. [Google Scholar] [CrossRef]
  220. Xu, S.; Liu, D.; Qin, Z.; Liang, Z.; Xie, H.; Yi, B.; Wang, K.; Lin, G.; Liu, R.; Yang, K.; et al. Experimental Validation and Pan-Cancer Analysis Identified COL10A1 as a Novel Oncogene and Potential Therapeutic Target in Prostate Cancer. Aging 2023, 15, 15134–15160. [Google Scholar] [CrossRef]
  221. Liang, Y.; Xia, W.; Zhang, T.; Chen, B.; Wang, H.; Song, X.; Zhang, Z.; Xu, L.; Dong, G.; Jiang, F. Upregulated Collagen COL10A1 Remodels the Extracellular Matrix and Promotes Malignant Progression in Lung Adenocarcinoma. Front. Oncol. 2020, 10, 573534. [Google Scholar] [CrossRef]
  222. Bruns, R.R. Beaded Filaments and Long-Spacing Fibrils: Relation to Type VI Collagen. J. Ultrastruct. Res. 1984, 89, 136–145. [Google Scholar] [CrossRef]
  223. Gara, S.K.; Grumati, P.; Urciuolo, A.; Bonaldo, P.; Kobbe, B.; Koch, M.; Paulsson, M.; Wagener, R. Three Novel Collagen VI Chains with High Homology to the A3 Chain. J. Biol. Chem. 2008, 283, 10658–10670. [Google Scholar] [CrossRef]
  224. Li, X.; Li, Z.; Gu, S.; Zhao, X. A Pan-Cancer Analysis of Collagen VI Family on Prognosis, Tumor Microenvironment, and Its Potential Therapeutic Effect. BMC Bioinform. 2022, 23, 390. [Google Scholar] [CrossRef] [PubMed]
  225. Wishart, A.L.; Conner, S.J.; Guarin, J.R.; Fatherree, J.P.; Peng, Y.; McGinn, R.A.; Crews, R.; Naber, S.P.; Hunter, M.; Greenberg, A.S.; et al. Decellularized Extracellular Matrix Scaffolds Identify Full-Length Collagen VI as a Driver of Breast Cancer Cell Invasion in Obesity and Metastasis. Sci. Adv. 2020, 6, eabc3175. [Google Scholar] [CrossRef] [PubMed]
  226. Kim, K.; Byun, Y.J.; Zheng, C.-M.; Moon, S.; Jo, S.J.; Kang, H.W.; Kim, W.T.; Choi, Y.H.; Moon, S.-K.; Kim, W.-J.; et al. COL6A1 Expression as a Potential Prognostic Biomarker for Risk Stratification of T1 High Grade Bladder Cancer: Unveiling the Aggressive Nature of a Distinct Non-Muscle Invasive Subtype. Investig. Clin. Urol. 2024, 65, 94. [Google Scholar] [CrossRef] [PubMed]
  227. Cescon, M.; Rampazzo, E.; Bresolin, S.; Da Ros, F.; Manfreda, L.; Cani, A.; Della Puppa, A.; Braghetta, P.; Bonaldo, P.; Persano, L. Collagen VI Sustains Cell Stemness and Chemotherapy Resistance in Glioblastoma. Cell. Mol. Life Sci. 2023, 80, 233. [Google Scholar] [CrossRef]
  228. Papalazarou, V.; Drew, J.; Juin, A.; Spence, H.J.; Whitelaw, J.; Nixon, C.; Salmeron-Sanchez, M.; Machesky, L.M. Collagen VI Expression Is Negatively Mechanosensitive in Pancreatic Cancer Cells and Supports the Metastatic Niche. J. Cell Sci. 2022, 135, jcs259978. [Google Scholar] [CrossRef]
  229. Liu, Y.; Murazzi, I.; Fuller, A.M.; Pan, H.; Irizarry-Negron, V.M.; Devine, A.; Katti, R.; Skuli, N.; Ciotti, G.E.; Pak, K.; et al. Sarcoma Cells Secrete Hypoxia-Modified Collagen VI to Weaken the Lung Endothelial Barrier and Promote Metastasis. Cancer Res. 2024, 84, 977–993. [Google Scholar] [CrossRef]
  230. Luo, Y.; Ye, Y.; Zhang, Y.; Chen, L.; Qu, X.; Yi, N.; Ran, J.; Chen, Y. New Insights into COL26A1 in Thyroid Carcinoma: Prognostic Prediction, Functional Characterization, Immunological Drug Target and CeRNA Network. Transl. Cancer Res. 2023, 12, 3384–3408. [Google Scholar] [CrossRef]
  231. Reese-Petersen, A.L.; Willumsen, N.; Palau, P.; Nunez, J.; Sun, S.; Jensen, T.M.; Kamall, B.; Karsdal, M.A.; Genovese, F. Evaluation of a Novel Biomarker of Type XXVIII Collagen Formation, PRO-C28, in Samples from Cancer and Heart Failure with Preserved Ejection Fraction Patients. J. Pharm. Biomed. Anal. 2021, 204, 114272. [Google Scholar] [CrossRef]
  232. Sakai, L.Y.; Keene, D.R.; Morris, N.P.; Burgeson, R.E. Type VII Collagen Is a Major Structural Component of Anchoring Fibrils. J. Cell Biol. 1986, 103, 1577–1586. [Google Scholar] [CrossRef]
  233. Gelse, K. Collagens—Structure, Function, and Biosynthesis. Adv. Drug Deliv. Rev. 2003, 55, 1531–1546. [Google Scholar] [CrossRef]
  234. Oh, S.E.; Oh, M.Y.; An, J.Y.; Lee, J.H.; Sohn, T.S.; Bae, J.M.; Choi, M.-G.; Kim, K.-M. Prognostic Value of Highly Expressed Type VII Collagen (COL7A1) in Patients With Gastric Cancer. Pathol. Oncol. Res. 2021, 27, 1609860. [Google Scholar] [CrossRef] [PubMed]
  235. You, W.-K.; Bonaldo, P.; Stallcup, W.B. Collagen VI Ablation Retards Brain Tumor Progression Due to Deficits in Assembly of the Vascular Basal Lamina. Am. J. Pathol. 2012, 180, 1145–1158. [Google Scholar] [CrossRef] [PubMed]
  236. Poomsawat, S.; Kariya, A.; Nimmanon, T.; Kosanwat, T.; Juengsomjit, R.; Sirima, S. Diagnostic Potential of Type VII Collagen during Oral Carcinogenesis. J. Appl. Oral. Sci. 2023, 31, e20220486. [Google Scholar] [CrossRef] [PubMed]
  237. Franzke, C.-W.; Bruckner, P.; Bruckner-Tuderman, L. Collagenous Transmembrane Proteins: Recent Insights into Biology and Pathology. J. Biol. Chem. 2005, 280, 4005–4008. [Google Scholar] [CrossRef]
  238. Jones, V.A.; Patel, P.M.; Gibson, F.T.; Cordova, A.; Amber, K.T. The Role of Collagen XVII in Cancer: Squamous Cell Carcinoma and Beyond. Front. Oncol. 2020, 10, 352. [Google Scholar] [CrossRef]
  239. Krenacs, T.; Kiszner, G.; Stelkovics, E.; Balla, P.; Teleki, I.; Nemeth, I.; Varga, E.; Korom, I.; Barbai, T.; Plotar, V.; et al. Collagen XVII Is Expressed in Malignant but Not in Benign Melanocytic Tumors and It Can Mediate Antibody Induced Melanoma Apoptosis. Histochem. Cell Biol. 2012, 138, 653–667. [Google Scholar] [CrossRef]
  240. Kashiwagi, R.; Funayama, R.; Aoki, S.; Matsui, A.; Klein, S.; Sato, Y.; Suzuki, T.; Murakami, K.; Inoue, K.; Iseki, M.; et al. Collagen XVII Regulates Tumor Growth in Pancreatic Cancer through Interaction with the Tumor Microenvironment. Cancer Sci. 2023, 114, 4286–4298. [Google Scholar] [CrossRef]
  241. Tong, D.; Tanaka, M.; Eguchi, H.; Okazaki, Y.; Muramatsu, M.; Arai, T. COL17A1 Germline Variant p.Ser1029Ala and Mucosal Malignant Melanoma: An Autopsy Study. Mol. Clin. Oncol. 2021, 16, 32. [Google Scholar] [CrossRef]
  242. Lothong, M.; Sakares, W.; Rojsitthisak, P.; Tanikawa, C.; Matsuda, K.; Yodsurang, V. Collagen XVII Inhibits Breast Cancer Cell Proliferation and Growth through Deactivation of the AKT/MTOR Signaling Pathway. PLoS ONE 2021, 16, e0255179. [Google Scholar] [CrossRef]
  243. Ohta, Y.; Fujii, M.; Takahashi, S.; Takano, A.; Nanki, K.; Matano, M.; Hanyu, H.; Saito, M.; Shimokawa, M.; Nishikori, S.; et al. Cell–Matrix Interface Regulates Dormancy in Human Colon Cancer Stem Cells. Nature 2022, 608, 784–794. [Google Scholar] [CrossRef]
  244. Crespo-Bravo, M.; Thorlacius-Ussing, J.; Nissen, N.I.; Pedersen, R.S.; Boisen, M.K.; Liljefors, M.; Johansen, A.Z.; Johansen, J.S.; Karsdal, M.A.; Willumsen, N. Levels of Type XVII Collagen (BP180) Ectodomain Are Elevated in Circulation from Patients with Multiple Cancer Types and Is Prognostic for Patients with Metastatic Colorectal Cancer. BMC Cancer 2023, 23, 949. [Google Scholar] [CrossRef] [PubMed]
  245. Zhang, H.; Fredericks, T.; Xiong, G.; Qi, Y.; Rychahou, P.G.; Li, J.-D.; Pihlajaniemi, T.; Xu, W.; Xu, R. Membrane Associated Collagen XIII Promotes Cancer Metastasis and Enhances Anoikis Resistance. Breast Cancer Res. 2018, 20, 116. [Google Scholar] [CrossRef] [PubMed]
  246. Dai, Y.; Zhang, X.; Ou, Y.; Zou, L.; Zhang, D.; Yang, Q.; Qin, Y.; Du, X.; Li, W.; Yuan, Z.; et al. Anoikis Resistance–Protagonists of Breast Cancer Cells Survive and Metastasize after ECM Detachment. Cell Commun. Signal. 2023, 21, 190. [Google Scholar] [CrossRef] [PubMed]
  247. Chen, Y.; Huang, W.; Ouyang, J.; Wang, J.; Xie, Z. Identification of Anoikis-Related Subgroups and Prognosis Model in Liver Hepatocellular Carcinoma. Int. J. Mol. Sci. 2023, 24, 2862. [Google Scholar] [CrossRef]
  248. Xu, F.; Chang, K.; Ma, J.; Qu, Y.; Xie, H.; Dai, B.; Gan, H.; Zhang, H.; Shi, G.; Zhu, Y.; et al. The Oncogenic Role of COL23A1 in Clear Cell Renal Cell Carcinoma. Sci. Rep. 2017, 7, 9846. [Google Scholar] [CrossRef]
  249. Keller, C.R.; Ruud, K.F.; Martinez, S.R.; Li, W. Identification of the Collagen Types Essential for Mammalian Breast Acinar Structures. Gels 2022, 8, 837. [Google Scholar] [CrossRef]
  250. Oh, S.P.; Kamagata, Y.; Muragaki, Y.; Timmons, S.; Ooshima, A.; Olsen, B.R. Isolation and Sequencing of CDNAs for Proteins with Multiple Domains of Gly-Xaa-Yaa Repeats Identify a Distinct Family of Collagenous Proteins. Proc. Natl. Acad. Sci. USA 1994, 91, 4229–4233. [Google Scholar] [CrossRef]
  251. Lakshmanachetty, S.; Koster, M.I. Emerging Roles for Collagen XV and XVIII in Cancer Progression. Exp. Dermatol. 2016, 25, 346–347. [Google Scholar] [CrossRef]
  252. Mutolo, M.J.; Morris, K.J.; Leir, S.-H.; Caffrey, T.C.; Lewandowska, M.A.; Hollingsworth, M.A.; Harris, A. Tumor Suppression by Collagen XV Is Independent of the Restin Domain. Matrix Biol. 2012, 31, 285–289. [Google Scholar] [CrossRef]
  253. Wang, S.; Zhou, M.; Xia, Y. COL15A1 Interacts with P4HB to Regulate the Growth and Malignancy of HepG2.2.15 Cells. Biochem. Biophys. Res. Commun. 2023, 681, 20–28. [Google Scholar] [CrossRef]
  254. Yao, T.; Hu, W.; Chen, J.; Shen, L.; Yu, Y.; Tang, Z.; Zang, G.; Zhang, Y.; Chen, X. Collagen XV Mediated the Epithelial-Mesenchymal Transition to Inhibit Hepatocellular Carcinoma Metastasis. J. Gastrointest. Oncol. 2022, 13, 2472–2484. [Google Scholar] [CrossRef] [PubMed]
  255. Liu, Y.; Wang, J.; Shou, Y.; Xu, W.; Huang, Z.; Xu, J.; Chen, K.; Liu, J.; Liu, D.; Liang, H.; et al. Restoring the Epigenetically Silenced LncRNA COL18A1-AS1 Represses CcRCC Progression by Lipid Browning via MiR-1286/KLF12 Axis. Cell Death Dis. 2022, 13, 578. [Google Scholar] [CrossRef] [PubMed]
  256. Devarajan, R.; Izzi, V.; Peltoketo, H.; Rask, G.; Kauppila, S.; Väisänen, M.-R.; Ruotsalainen, H.; Martínez-Nieto, G.; Karppinen, S.-M.; Väisänen, T.; et al. Targeting Collagen XVIII Improves the Efficiency of ErbB Inhibitors in Breast Cancer Models. J. Clin. Investig. 2023, 133, e159181. [Google Scholar] [CrossRef] [PubMed]
  257. Yan, B.; Liu, L.; Zhao, L.; Hinz, U.; Luo, Y.; An, X.; Gladkich, J.; de la Torre, C.; Huang, Z.; Schrapel, D.; et al. Tumor and Stroma COL8A1 Secretion Induces Autocrine and Paracrine Progression Signaling in Pancreatic Ductal Adenocarcinoma. Matrix Biol. 2022, 114, 84–107. [Google Scholar] [CrossRef] [PubMed]
  258. Vaday, G.G.; Lider, O. Extracellular Matrix Moieties, Cytokines, and Enzymes: Dynamic Effects on Immune Cell Behavior and Inflammation. J. Leukoc. Biol. 2000, 67, 149–159. [Google Scholar] [CrossRef]
  259. Shi, R.; Zhang, Z.; Zhu, A.; Xiong, X.; Zhang, J.; Xu, J.; Sy, M.; Li, C. Targeting Type I Collagen for Cancer Treatment. Int. J. Cancer 2022, 151, 665–683. [Google Scholar] [CrossRef]
  260. Huang, H.-Y.; Chen, L.-Q.; Sun, W.; Du, H.-H.; Dong, S.; Ahmed, A.M.Q.; Cao, D.; Cui, J.-H.; Zhang, Y.; Cao, Q.-R. Collagenase IV and Clusterin-Modified Polycaprolactone-Polyethylene Glycol Nanoparticles for Penetrating Dense Tumor Tissues. Theranostics 2021, 11, 906–924. [Google Scholar] [CrossRef]
  261. Wang, J.; Wu, Q.; Wang, Y.; Xiang, L.; Feng, J.; Zhou, Z.; Fu, Q.; Zhang, L. Collagenase-Loaded PH-Sensitive Nanocarriers Efficiently Remodeled Tumor Stroma Matrixes and Improved the Enrichment of Nanomedicines. Nanoscale 2021, 13, 9402–9414. [Google Scholar] [CrossRef]
  262. García-Olmo, D.; Villarejo Campos, P.; Barambio, J.; Gomez-Heras, S.G.; Vega-Clemente, L.; Olmedillas-Lopez, S.; Guadalajara, H.; Garcia-Arranz, M. Intraperitoneal Collagenase as a Novel Therapeutic Approach in an Experimental Model of Colorectal Peritoneal Carcinomatosis. Sci. Rep. 2021, 11, 503. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the major components of tumor microenvironment in solid tumors. The tumor microenvironment mainly consists of tumor cells, immune cells, stromal cells, and extracellular matrix. The primary factor contributing to matrix stiffness is the excessive presence of collagen, which significantly impacts various crucial biological processes throughout tumor development.
Figure 1. Schematic representation of the major components of tumor microenvironment in solid tumors. The tumor microenvironment mainly consists of tumor cells, immune cells, stromal cells, and extracellular matrix. The primary factor contributing to matrix stiffness is the excessive presence of collagen, which significantly impacts various crucial biological processes throughout tumor development.
Ijms 25 13523 g001
Figure 2. Classification of collagens based on supramolecular assembly and a schematic overview of the major roles of each collagen type in tumorigenesis.
Figure 2. Classification of collagens based on supramolecular assembly and a schematic overview of the major roles of each collagen type in tumorigenesis.
Ijms 25 13523 g002
Table 1. Collagen families, types, and their role in tumor progression.
Table 1. Collagen families, types, and their role in tumor progression.
Collagen FamilyCollagen TypeTumorRolesReferences
Fibrillar CollagenType ITriple-negative breast carcinoma, pancreatic ductal adenocarcinoma, and gastric carcinomaIncreased metastasis, proliferation, and invasion.[139,146,147,148,149,150,151,152,153,154,155]
Type II *Not reportedNot reportedNot reported
Type IIIBreast cancerReduction in invasiveness, tumor growth suppression; increased cell quiescence; inhibition of autophagy[48,146,155,156,157,158,159]
Type VGastric cancerIncreased proliferation and epithelial–mesenchymal transition[160,161,162]
Type XIPapillary thyroid carcinoma, breast cancer, colorectal carcinoma, esophageal cancer, gastric cancer, pancreatic cancer, lung cancer, and ovarian cancerPromotion of proliferation, drug resistance, and tumor invasiveness[63,163,164,165,166,167,168,169,170,171,172,173,174]
Type XXVIIMelanoma cellsPromotion of tumor cells quiescence[175]
Type XXIV *Not reportedNot reportedNot reported
FACITsType XIIBreast cancer, colorectal cancer, pancreatic adenocarcinoma, and ovarian cancerPromotion of tumor progression and metastasis[179,180,181,182,183]
Type XVIColorectal cancer (CRC), glioblastoma, and oral squamous cell carcinomaIncreased invasiveness[184,185,186]
Type XIVMelanomaReduction in malignancy[187]
Type XIXMelanomaInhibition of tumor cell migration and invasion[188]
Type XXPancreatic ductal adenocarcinomaNot reported[189]
Type XXIIPancreatic ductal adenocarcinomaNot reported[189]
Type XXIGastric carcinomaIncreased tumor progression[190]
Type IX *Not reportedNot reportedNot reported
Network-forming collagenType IVBreast carcinoma, colorectal tumor, hepatocellular carcinomaPromotion of tumor proliferation, durotaxis, angiogenesis, and metastasis. Reduction in angiogenesis[192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214]
Type VIIIBreast and pancreatic ductal adenocarcinomaPromotion of tumor cell invasion and migration, as well as drug resistance[149,215,257]
Type XBreast cancer, colorectal cancer, gastric cancer, and lung adenocarcinomaPromotion of tumor cell proliferation, migration, and invasion[218,219,220,221]
Beaded filament-forming collagenType VIGlioblastoma, ovarian cancer, and pancreatic ductal adenocarcinomaPromotion of metastasis, angiogenesis, and tumor progression[224,225,226,227,228,229]
Type XXVIThyroid carcinomaNot reported[230]
Type XXVIIILung cancerNot reported[231]
Anchoring
fibril
Type VIIOral squamous cell carcinoma and gastric carcinomaPromotion of tumor progression; block invasiveness[232,233,234,235,236]
Transmembrane collagenType XVIISquamous cell carcinoma; breast cancer, colorectal cancerPromotion of cell migration and invasion. Block tumor progression[238,239,240,241,242,243,244,245,246]
Type XIIIBreast, clear cell renal cell carcinoma, and prostate cancerTumor progression, invasion, resistance to anoikis, and tumor metastasis[245,246,247]
Type XXIIIClear cell renal cell carcinomaPromotion of tumor progression, adhesion, and migration[248]
Type XXVBreast cancerNot reported[249]
MultiplexinType XVCervical cancer; hepato-carcinomaReduction in growth, invasion, and migration[251,252,253,254]
Type XVIIIClear cell renal carcinoma, breast cancerIncreased proliferation and migration[252,256,257]
* Roles of collagens II, XXIV, and IX in tumor progression uncleared.
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

Lo Buglio, G.; Lo Cicero, A.; Campora, S.; Ghersi, G. The Multifaced Role of Collagen in Cancer Development and Progression. Int. J. Mol. Sci. 2024, 25, 13523. https://doi.org/10.3390/ijms252413523

AMA Style

Lo Buglio G, Lo Cicero A, Campora S, Ghersi G. The Multifaced Role of Collagen in Cancer Development and Progression. International Journal of Molecular Sciences. 2024; 25(24):13523. https://doi.org/10.3390/ijms252413523

Chicago/Turabian Style

Lo Buglio, Gabriele, Alessandra Lo Cicero, Simona Campora, and Giulio Ghersi. 2024. "The Multifaced Role of Collagen in Cancer Development and Progression" International Journal of Molecular Sciences 25, no. 24: 13523. https://doi.org/10.3390/ijms252413523

APA Style

Lo Buglio, G., Lo Cicero, A., Campora, S., & Ghersi, G. (2024). The Multifaced Role of Collagen in Cancer Development and Progression. International Journal of Molecular Sciences, 25(24), 13523. https://doi.org/10.3390/ijms252413523

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