Solid tumours are composed of cancer cells characterised by genetic mutations that underpin the disease, but also contain a suite of genetically normal cells and the extracellular matrix (ECM). These two latter components are constituents of the tumour microenvironment (TME), and are key determinants of tumour biology and thereby the outcomes for patients. The tumour ECM has been the subject of intense research over the past two decades, revealing key biochemical and mechanobiological principles that underpin its role in tumour cell proliferation and survival. However, the ECM also strongly influences the genetically normal immune cells within the microenvironment, regulating not only their proliferation and survival, but also their differentiation and access to tumour cells. Here we review recent advances in our knowledge of how the ECM regulates the tumour immune microenvironment and vice versa, comparing normal skin wound healing to the pathological condition of tumour progression.

Tumours consist not only of neoplastic cells but also an altered stroma or microenvironment that is distinct to that found in the corresponding normal tissues. The tumour microenvironment (TME) describes constituents of the tumour mass that are not tumour cells, and is a key focus of research because it contributes to tumorigenesis, invasion and metastasis, as well as to resistance to chemotherapy and radiotherapy. The TME comprises both cellular and non-cellular components. The cellular portion includes diverse stromal cells, including cancer-associated fibroblasts (CAFs), immune cells (tumour-associated macrophages [TAMs], lymphocytes, mast cells [MCs]), endothelial cells, pericytes, adipocytes, nerve cells and others [1,2]. A key feature of these cells is that they have not undergone oncogenic transformation. The non-cellular portion of the TME is the extracellular matrix (ECM), a protein scaffold that provides architectural and mechanical support, anchorage for cell adhesion, storage of water and various growth factors, and induces intracellular signaling pathways via cell–ECM interactions [3]. CAFs are among the most abundant cell types in the microenvironment of solid tumours, second only to tumour cells themselves. CAFs are activated or re-educated mesenchymal-like cells in the TME and exhibit considerable cellular heterogeneity. CAFs can exhibit tumour-promoting functions or tumour-suppressive capability depending on their particular context, and act by releasing various growth factors (such as epidermal growth factors [EGFs], transforming growth factor [TGF]-β and hepatocyte growth factor [HGF]) and an array of chemokines to induce different aspects of tumour cell behaviour [4]. CAFs also regulate tumour cell behaviour by regulating mechanical cues exerted by the ECM [5]. CAFs have an important role in regulating immune cell behaviour within the TME, including potentially their differentiation and identity [6], thereby modulating the ability of the immune system to find and destroy cancer cells [7].

Immune surveillance is compromised in many tumours [8–10]. Cancer cells evade immune destruction by polarising infiltrating immune cells such as macrophages and lymphocytes into tumour promoting or immunosuppressive states, depending on the context. Therefore, a tumour can be viewed not as a collection of relatively homogeneous cancer cells but instead as a novel organ in which cancer cells interact with different specialised stromal cells and ECM that operate together in a coordinated fashion [6]. By investigating and further understanding the acquired capabilities necessary for tumour growth and progression, measures may be taken, or interventions developed to target the specific mechanisms contributing to cancer progression. Here, we discuss key similarities between the microenvironments of epithelial cancers and healing wounds to leverage our existing knowledge of wound healing to elucidate the tumour microenvironment.

Tissue homoeostasis is a meticulously regulated and dynamic process that occurs throughout the lifetime of an organism including during development, and can be perturbed during pathological processes such as wound healing and tumorigenesis (Figure 1).

Biochemical, structural and mechanical interrelationships between the ECM and tumour-associated immune cells determine key disease outcomes

Figure 1
Biochemical, structural and mechanical interrelationships between the ECM and tumour-associated immune cells determine key disease outcomes

The ECM is largely produced, remodelled and degraded by a suite of genetically normal cell types within the microenvironment. This figure highlights key relationships between immune cells, CAFs and the ECM that regulate the biochemical, structural and mechanical characteristics of the ECM, which in turn regulate the cellular functions determining the course of the disease.

Figure 1
Biochemical, structural and mechanical interrelationships between the ECM and tumour-associated immune cells determine key disease outcomes

The ECM is largely produced, remodelled and degraded by a suite of genetically normal cell types within the microenvironment. This figure highlights key relationships between immune cells, CAFs and the ECM that regulate the biochemical, structural and mechanical characteristics of the ECM, which in turn regulate the cellular functions determining the course of the disease.

Close modal

Disruption of skin homoeostasis by wounding triggers a cascade of events requiring the interaction of local keratinocytes, fibroblasts of the connective tissue, and the immune system, aimed at restoring skin integrity. The first step of wound healing is haemostasis, during which the body responds to the injury and coagulation of blood stops leakage from damaged vessels. From this stage, the platelets begin restoration of tissue structure by stimulating angiogenesis and fibroblast activation, but also inflammation by recruiting neutrophils and monocytes. The inflammation and proliferation phases require the interaction of immune cells and connective tissue fibroblasts to clear debris and infection and restore the ECM. Wound healing is complete in the maturation phase where re-establishment of the connective tissue and re-epithelialisation are completed, and homoeostasis is restored [11].

In the early phase of wound healing, the provisional ECM deposited by macrophages and activated fibroblasts is a disorganised matrix consisting predominantly of fibrin and fibronectin (FN), which is subsequently replaced by the normal ECM, composed of collagen and glycoproteins, prior to resolution. The composition of the ECM that is established may influence the type and extent of inflammation that occurs, with scarless healing characterised by low levels of inflammation and scarring characterised by high levels of inflammation [12]. While it is still unclear which of these are cause and consequence, the clear association between these two phenomena is striking.

Similar molecular mechanisms and cellular behaviour are co-opted during tumorigenesis, except that wound healing is a finely tuned and self-limiting process; whereas tumorigenesis is, in contrast, characterised by dysregulated cellular behaviour and signalling pathways, which while being well-coordinated, are not self-limiting [13]. The similarities between the two processes have been appreciated for some time, since Dvorak postulated the idea that tumours are wounds that do not heal [14]. Like the wound healing microenvironment, the TME contains fibroblasts, immune cells (lymphocytes, macrophages, MCs), other stromal cells (endothelial cells, adipocytes, pericytes) as well as ECM.

In the acute inflammation response to wound healing, the first two immune cell types recruited into and surrounding the wound are neutrophils and monocytes, which differentiate into macrophages. Both neutrophils and macrophages boost inflammatory responses by secreting pro-inflammatory cytokines such as tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 [15]. Macrophages exist on a phenotypic continuum, ranging between the pro-inflammatory (M1) and anti-inflammatory (M2) polarised states. In the early phase of wound healing, expression both M1 and M2 markers are observed, while later in wound healing, macrophages exhibiting M2 markers predominate [16]. Intriguingly, the condition of chronic inflammation has long been associated with a microenvironment conducive for tumorigenesis and cancer progression [17,18]. TAMs are one of the main tumour-infiltrating immune cell types that participate in the formation of the TME. It has been demonstrated that M2 TAMs in TME promote tumour cell proliferation, invasion/metastasis and angiogenesis through their secretion of cytokines (IL-10, IL-1β), chemokines and growth factors (vascular endothelial growth factor [VEGF], TGF-β), as well as ECM remodelling proteases including matrix metalloproteinases (MMPs). In a manner similar to that proposed for wounds above, the properties of the tumour ECM strongly influence the type and extent of inflammation that occurs within the tumour.

The ECM is the acellular constituent of all tissues and organs of the body and is also a key component of tumours [19]. It consists not only of a wide range of macromolecules that form a three-dimensional fibrillar architecture that surrounds and supports cells but also serves as a reservoir of cytokines, chemokines, and growth factors that modulate cell fate. Furthermore, the ECM plays a significant role both in normal physiological homoeostasis and in pathological process such as inflammation, fibrosis and tumorigenesis. The tumour ECM forms a scaffold within which tumour cells and the cellular constituents of the TME exist. In cancers, it is well documented that the ECM has both anti-tumorigenic and pro-tumorigenic functions, including the potential to foster or indeed suppress cancer initiation, cancer cell migration/invasion, metastatic dissemination and distant colonisation. The ECM of tumours is commonly dysregulated and disorganised relative to the ECM of the corresponding normal tissue, and implicated in eliciting aberrant cellular behaviour in both cancer cells and stromal cells, thereby promoting cellular transformation and metastasis [20].

The ECM is synthesised by all cell types to varying amounts, but the main producers of ECM are cells of the connective tissue, including stromal cells such as fibroblasts, immune cells, and pericytes. Epithelial, neuronal, and muscle cells may also produce ECM depending on the state of the tissues they populate – for instance, whether the tissue is in the process of development, healing following injury, fibrotic or in a state of tumorigenesis [19,21]. Production of ECM by these cells is governed by a suite of signalling pathways generally termed mechanotransduction pathways, which are regulated by feedback from key properties of the ECM itself, a phenomenon termed mechanoreciprocity, but that may also be engaged by classical signalling molecules such as growth factors and cytokines [5,22].

Mechanotransduction pathways can be initiated by several means including by mechanically induced integrin clustering [23], engagement of discoidin domain receptor (DDR) tyrosine kinases or activation of mechanically regulated ion channels in a variety of cell types [24], and the leucocyte-associated immunoglobulin-like receptor 1 (LAIR1/CD305) in white blood cells [25]. Downstream of these mechanical inputs, signalling pathways including those mediated by mitogen-activated protein kinases (MAPKs), Rho-associated kinase (ROCK), Phosphoinositide 3-kinase (PI3K) and Yes-associated protein (YAP) and its homologues the TAZ proteins are key mechanotransduction pathways that convert mechanical signals into functional biochemical and mechanical outputs. For example, signalling through ROCK results in enhanced actomyosin contractility, thereby permitting cells to respond to external mechanical stresses, but also regulates the production and remodelling of ECM proteins within tissues in normal tissue homoeostasis, wound healing and tumorigenesis [26–29]. A similar mechanism is also regulated by YAP signalling [30,31], suggesting a principle of reciprocal regulation of intracellular and extracellular mechanical stresses [32] exerted by the cellular cytoskeleton and the ECM respectively.

The entire set of genes coding the matrix and matrix-associated proteins is termed the ‘matrisome’ [33]. In human and mouse genomes, there are approximately 1050 matrix genes, which can be categorised into those encoding core-matrisome proteins (∼280 genes) and matrisome-associated proteins (∼780 genes). The core-matrisome proteins are a collection of ECM glycoproteins, collagens and proteoglycans. The matrisome-associated proteins comprise ECM-affiliated proteins, ECM regulators and secreted factors. Using human tumour xenografts in mice and proteomic analysis, Naba et al. demonstrated that the tumour ECM is contributed by both tumour cells and stromal cells [33]. Tumour cells with varying metastatic potential secrete distinct suites of matrisome components. Furthermore, the stromal cell-secreted matrisome components differ according to the metastatic potential of the tumour cells, indicating cross-talk between the tumour cells and the stromal cells [33].

Collagens, as the main structural element of the matrix, are generally the most abundant protein within the ECM, and indeed throughout the body. At least 28 different types of collagens have been identified in vertebrates. Collagens provide mechanical strength and bioactive sites for cell adhesion. Fibrillar collagens, such as collagen type I, are the major component of tumour desmoplasia and are critical players in tumour cell survival and metastasis in many tumour types, such as breast cancers [26,34], pancreatic ductal adenocarcinoma [28,35] and skin tumours [27]. Compared with the benign lesions in which type I and type III collagen bundles are regularly organised, malignant human breast tumours show high expression of type I and type III collagen and newly synthesised collagens are frequently arranged in abnormal bundles, particularly at the invasive front of malignant tumours. Fibroblast-like cells of the stroma rather than malignant epithelial cells contribute most to the high level of collagen within the tumour ECM. The aberrant and destructive pattern of collagen bundles suggests a more readily degradable TME and thus may facilitate breast tumour invasion [36]. Network-forming collagens are rich in basement membrane which define boundaries within tissues and separate epithelial or endothelial layers from the interstitial matrix and are essential for tissue polarity. It is well established that the basement membrane must be breached or traversed by cancer cells prior to invasion, metastasis and tumour angiogenesis [37–39]. MMPs, which are divided into secreted and membrane-bound MMPs are the major family of matrix-degrading proteolytic enzymes. MMPs cleave a wide range of substrates, such as collagen, FN, laminin and elastin (ELN) and involve in degradation of basement membranes and other ECM barriers, participating in both normal processes and pathogenesis noted in cancer progression [40]. By substrate specificity and cellular localisation, MMPs can be divided into collagenases (MMP-1, 8 and 13), gelatinases (MMP-2 and 9), stromelysins (MMP-3, 10 and 11), matrilysins (MMP-7 and 26) and membrane-type MT-MMPs (MMP-14, 15, 16 and 17) [41]. Of note, membrane type 1-MMP (MT1-MMP; also designated MMP-14) has important roles in matrix turnover and regulates key functional characteristics of breast cancer cells, such as migration, invasion and angiogenesis [41]. Up-regulation of active MT1-MMP has been noted in CAFs of the TME in mouse models of breast cancer [42]. MT1-MMP-mediated proteolysis by stromal cells is important in the metastatic process in mammary cancers of PyMT mice [43]. MMP14 gene expression is remarkably higher in tissues from invasive breast carcinoma compared with normal tissues. High MMP14 expression is related to poor survival in ERα-negative breast cancer and triple-negative breast cancer patients [41].

The other two groups of the core matrisome are glycoproteins and proteoglycans. Glycoproteins are a group of proteins with short, branched oligosaccharide chains covalently attached to a central polypeptide core. Proteoglycans are glycoproteins with attached glycosaminoglycans (GAGs) which confer on proteoglycans a high negative charge and the ability to store water and divalent cations such as calcium [44], and have been implicated in CAF education [45]. GAGs are long, linear, acidic carbohydrate polymers with repeating disaccharides comprising heparan sulphate/heparin (HS/HP), chondroitin sulphate/dermatan sulphate (CS/DS), keratan sulphate (KS) and hyaluronic acid (HA). Heparan sulphates on proteoglycans bind many secreted and growth factors into the ECM [46]. Dysregulation of glycoproteins including laminins, FNs, tenascins, and periostin (POSTN) has been observed in the TME. Laminins together with collagen IV are primary components of the basement membrane that provide a structural barrier to cancer cell invasion, intravasation, and extravasation [47]. The glycocalyx, mainly composed of proteoglycans and glycoproteins, is a surface layer that covers all cells, including cancer cells. The thickness of the glycocalyx on cancer cells is related to integrin-mediated cellular adhesion to ECM, cell growth, and cell survival [48]. A thick glycocalyx is frequently observed on cancer cells. By expression of large tumour-associated glycoproteins such as MUC1, a bulky glycocalyx on non-transformed mammary epithelial cells promoted focal adhesion assembly and facilitated integrin-dependent ERK and AKT signalling to support cell growth and survival. Large glycoproteins (such as MUC1 and CD44) are abundantly expressed on circulating tumour cells isolated from patients with metastatic breast cancer that indicated a bulky glycocalyx on tumour cells could favour tumour cell dissemination and metastasis [48].

FNs have the ability to bind simultaneously to cell surface receptors, collagen, proteoglycans, and other FN molecules providing essential ECM–cellular connections to cells through cell surface integrin receptors and other receptors to regulate cell adhesion, migration, and differentiation [49]. FN polymerisation into the ECM is required for the production of collagen-I and thrombospondin (TSP)-1 indicating that FN polymerisation is a critical regulator of ECM organisation and stability [50]. In cancer, FN is expressed by both CAFs and cancer cells. Alignment of the FN fibres is a prominent feature of both prostatic and pancreatic cancer stromata, in which FNs assembled by CAFs are deposited in an anisotropic manner, which guide cancer cells to migrate via αv integrin engagement on the surface of prostate cancer cells [51].

ELN is a fibrillar protein with highly elastic characteristics expressed in tissues that need the ability to resume their shape following stretching. As such it is a key constituent of the dermis and has the unusual characteristics of being a very long lived protein that is minimally turned over in health [52]. As such, adult wound healing does not regenerate the full ELN composition of unwounded skin, resulting in scars that lack elasticity. In contrast, one of the processes that establishes the tumour ECM is the production of ELN, termed tumour elastosis, which has been associated with both tumour-promotion and tumour-suppression [53]. These observations suggest context-dependent functions, possibly associated with the capacity peptides produced from ELN degradation to interact with the elastin receptor complex (ERC) [54] and other receptors such as integrins, as well as the ability of lysyl oxidase to cross-link ELN fibres with collagen fibres [55]. The ERC contains neuraminidase 1, which is activated upon ligand engagement with the ERC, causing cell type-dependent activation of effector pathways, including the tumour-promoting MAPK signalling pathway [56]. Inhibition of ELN degradation, lysyl oxidases and the ERC are therefore worth exploring as potential therapeutic approaches to both enhance scarless healing and suppress tumour progression. Tenascin-C (TNC), expressed by both transformed epithelial cells and stromal cells in the TME, is up-regulated in cancerous tissue and strongly associated with invasiveness and metastasis [57]. In breast cancer, the expression of TNC is correlated with the aggressiveness of pulmonary metastasis. TNC enhances the WNT pathway and thereby cancer cell proliferation, via its positive regulator Musashi homologue 1 (MSI1) and NOTCH signalling in breast cancer cells [58]. TNC produced by metastasis-initiating breast cancer cells is critical for metastasis outgrowth in the pulmonary parenchyma, whereupon infiltrating myofibroblasts or another stromal cells take over as a source of TNC to facilitate subsequent growth [58]. It is notable that TNC is also implicated in chemoresistance; in xenograft mouse models of breast cancer, exposure to multiple chemotherapy agents was shown to activate the JNK pathway which enhances the expression of secreted phosphoprotein 1 (SPP1) and TNC via c-Jun transcription factor, leading to reduced therapeutic efficacy and progression of lung metastasis. Inhibition of JNK signalling or down-regulation of SPP1 or TNC can sensitise mammary tumours to chemotherapy and suppress the progression of metastatic breast cancer [59].

POSTN is highly expressed, mainly by stromal cells such as CAFs and to a lesser extent by tumour cells, and this is frequently associated with poor prognosis and metastasis in solid tumours. POSTN interacts with integrins expressed on the surface of tumour cells and initiates numerous signalling pathways that regulate cell proliferation, cell survival, or cell migration [60]. In oral squamous cell carcinoma (OSCC), overexpression of POSTN is frequently observed and is correlated with the pattern of invasion and metastasis. POSTN-positive cases of OSCC had higher blood vessel density and recombinant POSTN enhanced capillary formation in vitro in a concentration-dependent manner which suggests that POSTN may promote angiogenesis [61]. In pancreatic ductal adenocarcinoma, POSTN interacts with α6β4 integrin in cancer cells leading to phosphorylation of FAK and AKT through activation of the PI3K pathway and promoting the invasiveness of tumour cells by increasing the motility of cells [62]. In head and neck squamous cell carcinoma, POSTN secreted by CAFs might promote cancer stemness through interacting with protein tyrosine kinase 7-Wnt/β-catenin signalling pathway in cancer cells [63]. In the MMTV-PyMT mouse breast cancer model, infiltrating cancer cells need to induce stromal POSTN expression to initiate lung metastatic colonisation. The production of POSTN then augments Wnt signalling to facilitate cancer stem cell maintenance and promote metastasis [64].

The foregoing highlights a strong pattern of ECM involvement with tumour progression by direct engagement of mechanotransduction pathways by ECM components. However, another way in which the ECM influences disease progression is via its effects on the tumour immune system, which will be discussed in the next several sections.

The immune microenvironment is strongly influenced by characteristics of the ECM, while also regulating its production and remodelling (Figure 1). Many immune cell types engage directly with the ECM, which defines their biology and function. Furthermore, different immune cell types have key roles in ECM homoeostasis, participating in the production of ECM components, structurally remodelling the ECM and degrading it as required.

Inflammation in skin wound healing and the TME

Inflammation during cutaneous wound healing serves key functions beyond host defence against invading pathogens. The primary immune cell players which include neutrophils, monocyte–macrophages, MCs, and T lymphocytes also produce cytokines, chemokines, growth factors, and proteases that support proliferation of the epithelial cells as well as production and remodelling of ECM by fibroblasts [65]. Fibroblasts of the connective tissue are critical for the restoration of the ECM during wound healing, but also have important interactions with the immune system.

Cross-talk between the immune system and fibroblasts of the connective tissue is critical for efficient wound healing. Disruption of this interplay can lead to a range of outcomes from relatively minor aberrations such as scar formation to severe problems such as fibrosis. The outcomes of the wound healing process therefore rely on the precisely regulated interactions between all cell types involved as well as with the ECM. However, the process that these immune cells play in wound healing can be hijacked to establish a pro-tumorigenic microenvironment, in which a cancer develops as the well-canvassed ‘wound that never heals’ [14]. In this section, we review the major players of the immune system during wound healing, and how they modulate the ECM, to draw lessons on how these cells may be co-opted to establish and maintain the TME.

Immune cell recruitment is initiated by signalling from damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and platelets, which mediate coagulation. Immune cells responding early include neutrophils and monocyte–macrophages, as well as tissue-resident MCs. These cell types all play critical roles in host protection, as well as activation of fibroblasts and keratinocytes. While T lymphocytes are also recruited, they are not critical for the wound healing processes. In homoeostasis, key immune functions are performed by tissue-resident immune cells, but during wound healing the primary active players are recruited via the vasculature. Mediators released by resident MCs can aid in permeability of the vasculature, but neutrophils in particular can transmigrate with very little remodelling of the vessels [66]. Recruitment of the immune cell players into the wound site is reliant on biochemical signaling, but also requires mechanical and structural alterations to allow passage of the cells. ECM collagen structure, organisation, and matrix pore size can also therefore influence immune cell infiltration [67,68]. Therefore, migration of immune cells through the connective tissue is dependent on MMP-mediated ECM degradation to enlarge pore diameter, and integrin- and actomyosin-dependent exertion of force for path generation.

What follows is a brief description of each immune cell type, how they are recruited and the functions they fulfil in wound healing and tumour progression. We also discuss factors expressed by the cell that can influence ECM structure, and mechanisms by which they interact with fibroblasts as the major ECM producers. In the final section, we discuss how these features of the immune cells act to either promote or suppress tumorigenesis.

The ECM in neutrophil recruitment and function

Neutrophils are recruited to the site of tissue injury by DAMPs also called Alarmins, which include molecules such as DNA and high mobility group box protein (HMGB)-1, which are referred to as nuclear DAMPs, ATP and uric acid, termed cytosolic DAMPs and components of the ECM [69]. These signals are produced by damaged keratinocytes and neutrophils are typically the first immune cells to respond to them [70]. Sustained neutrophil recruitment is mediated by chemokines and lipid mediators such as CXCL8/IL-8 family and leukotriene B4 (LTB4), which are also DAMP-activated and released from endothelial cells [71]. IL-8 (aka CXCL8) is a well-known chemoattractant for neutrophils and signals via the receptors CXCR1 and CXCR2. CXCR2-deficient mice have impaired cutaneous wound healing [72], highlighting the importance of this chemokine in the skin.

In conjunction with their roles as innate defenders against pathogens, neutrophils can also produce pro-inflammatory cytokines and chemokines, promote angiogenesis, degrade the ECM, and influence the migration, proliferation and activation of keratinocytes and fibroblasts [73]. Once spent, neutrophils undergo apoptosis and are cleared by macrophages. This initiates a feed forward pro-resolution programme that is characterised by the release of the tissue repairing cytokines, TGF-β and IL-10.

These functions are facilitated by degranulation, which is the release of pre-formed and stored cytoplasmic granules, providing the materials for a quick response during tissue injury. There are four main types of granules – azurophilic (or primary) granules, specific (or secondary) granules, gelatinase (or tertiary) granules, and secretory vesicles (SVs) – each of which differs in its contents, signals required for granule secretion, and site of secretion [74]. In settings of infection and inflammation neutrophil extracellular traps (NETs) and extracellular vesicle exosomes [75] are also important sources for ECM-degrading proteases.

A significant component of primary granules are the serine proteases, of which elastase, proteinase 3, and cathepsin G are major components [76]. Neutrophil extravasation to site of injury can be minimally disruptive to vasculature. Migration through the vessel basement membrane, the part of the ECM first encountered by extravasating neutrophils, is facilitated by neutrophil-elastase and by passing through gaps in the pericytes where there are reduced levels of the ECM proteins laminin 10, collagen IV, and nidogen-2 [66]. In settings of disease and inflammation, the neutrophil serine proteases function to ingest pathogens, as well as to break down ECM to aid in migration [76]. Degradative targets within ECM include ELN, FN, laminin, and collagen IV [76]. Moreover, the serine proteases can promote MMP activity by cleavage of the pro-form of these enzymes to engage their catalytic activity [77].

The secondary and tertiary particles of neutrophils contain many matrix-degrading enzymes. Neutrophil-sourced MMP-8 can degrade collagen I and III, MMP-9 can cleave collagen IV [78], and MMP-25 can digest collagen IV and FN [79]. During wound healing, degradation of the ECM occurs to facilitate neo-angiogenesis within the restored tissue, which is key to normal regeneration. Neutrophils are a source of VEGF, a major growth factor that can stimulate angiogenesis [80] but can also cause epithelial cells to produce VEGF. Other pro-angiogenic factors such as MMPs, can degrade ECM and release ECM-bound VEGF which is then free to initiate signalling. Neutrophils are the only cells that can release MMP-9 free of its endogenous inhibitor, tissue inhibitor of metalloproteinases (TIMPs), and can directly deliver MMP-9 to sites of angiogenesis [81].

Neutrophils have also been known to activate fibroblasts during wound healing. During healing after myocardial infarction, neutrophils aid fibroblast in producing the provisional fibrin/fibronectin matrix, and stimulate fibroblast differentiation, collagen synthesis, and macrophage polarisation [82]. Formation of NETs induces activation of fibroblasts and differentiation into myofibroblast phenotype, which had increased connective tissue growth factor expression, collagen production, and proliferation/migration [83].

The very diverse functions of neutrophils in wound healing therefore requires a balance of factors to orchestrate migration, inflammation, angiogenesis, and fibroblast activation to produce effective healing. The opportunity for these same characteristics to be co-opted to detrimental effect in tumorigenesis are striking.

Tumour-associated neutrophils

Neutrophils are attracted to tumours by tumour-derived IL-1β, prostaglandin E2 (PGE2), and IL-8 [84]. In the early stages of tumorigenesis, as in wound healing, tumour-associated neutrophils (TANs) behave in a cytotoxic manner, producing high levels of TNF-α, NO, and H2O2 [85]. At later stages of tumorigenesis, neutrophils can exhibit a tumour-promoting phenotype. This change in function is associated with reduced prognostic value and a high incidence of metastasis [86]. Neutrophil polarisation is driven by TGF-β [87], and TANs are thought to exist within a continuum of states bounded by the pro-tumour N1 and anti-tumour N2 states [87].

The actions of tumour-promoting TANs include enhancement of angiogenesis, tumour cell proliferation, and immuno-suppression. TANs can suppress T-cell function in colorectal cancer (CRC) by secreting MMPs to activate TGF-β-mediated T-cell suppression [88]. TANs drive tumour angiogenesis by a range of mechanisms, including activation of pro-MMP-2 by elastase, cathepsin G, and proteinase-3 [77]. Neutrophil elastase and MMP-9 can each support angiogenesis by activation of VEGF [89,90]. Elastase from TANs can in fact activate latent forms of VEGF, platelet-derived growth factor (PDGF), and TGF-β to drive tumour cell proliferation and migration [90,91], but may also function via its role in regulating ECM homoeostasis [92].

ECM remodelling plays a role in the cellular mechanism underpinning NET-induced reversal of tumour cell dormancy and the resumption of tumour cell proliferation. NET-associated proteases neutrophil elastase and MMP-9 can induce re-awakening of dormant cancer cells through ECM remodelling cleaving of laminin-111, leading to activation of integrin a3β1 [93] and downstream signalling molecules including FAK, ROCK, YAP and MEK, resulting in tumour recurrence, disease relapse, and metastasis [93].

Macrophages in tissue and ECM homoeostasis

Macrophages are indispensable for cutaneous wound healing [94,95]. Monocytes are recruited to the wound site by a range of cytokine signals, and are terminally differentiated in situ into macrophages and dendritic cells. Chemoattractants for monocytes in wound healing include CCL-2 (MCP-1), CCL-3 (MIP1α), CCL-4 (MIP-1β), CCL-5, TSP-1, IL-1, IL-6, and TNF-α [96,97]. Neutrophils, which secrete many of these factors, can therefore recruit monocytes to the wound, and the mature macrophages then clear spent neutrophils from the wound site via phagocytosis.

Wound macrophages display heterogeneous phenotypes usually described as a continuum bound by the M1 and M2 phenotypic states [98]. At the initial stages of healing, most macrophages are polarised towards a pro-inflammatory M1 phenotypic state which aids in the clearance of pathogens, dead neutrophils, and dead tissue. After this initial phase, macrophages transition to a largely M2 anti-inflammatory phenotype and encourage re-epithelialisation, stimulate fibroblast migration and proliferation, and angiogenesis [95]. At the final phase of wound healing, macrophages release MMPs to digest the provisional ECM and then undergo apoptosis so that the skin can return to homoeostasis. In chronic wounds, macrophages retain their pro-inflammatory phenotype, resulting in persistent inflammation that impedes tissue repair.

Macrophage polarisation towards a more M1 state is mediated by interferon (IFN)-γ, TNF-α and GM-CSF, and can be identified by surface markers CD68, CD80 and CD86, and the secretion of pro-inflammatory cytokines such as IL-1β, IL-6, IL-12, IL-23, TNF-α, CXCL-9, and CXCL-10. Whereas IL-4, IL-13, IL-10, and TGF-β induce the M2 phenotype, which is characterised by the expression of CD204, CD206, and CD163 surface markers, and the secretion of anti-inflammatory cytokines such as IL-10, TGF-β, CCL-17, CCL-18, CCL-22, and CCL-24 [99]. Midway through healing, macrophages expressing CD301b produce high levels of IL-10, PDGF-β, and TGF-β [100] – these factors are necessary for efficient re-epithelisation, revascularisation, and fibroblast regeneration.

Macrophages are an important source of MMP-2 and MMP-9 to degrade ECM [101]. However, unlike neutrophil-released MMP-9, macrophage-produced MMP-9 is bound by TIMP1, which limits MMP-9 activity [81]. Therefore, macrophages appear to have an intrinsic modulatory mechanism for MMP function. MMPs and the role of macrophages in modulating their activity are important for the remodelling phase of wound healing to alter the composition of the nascent ECM and to re-establish normal ECM within the healed skin [98].

A key role of macrophages during wound healing is the mutual regulation of other cells of the microenvironment. Fibroblasts secrete CSF1, an essential growth factor for macrophages, and macrophages can conversely stimulate fibroblast proliferation by producing PDGFα and activated macrophages can enhance ECM hyaluronan and ELN synthesis by fibroblasts [102]. Neutrophil recruitment of monocytes induces their own phagocytosis and this CD18 (integrin β2)-dependent contact can stimulate the expression of TGF-β in macrophages [103]. TGF-β is a major cytokine that induces fibroblast differentiation into myofibroblasts, with concomitant α-SMA expression, and wound contraction resulting from the ability of these cells to physically contract the ECM [104,105].

Conversely, fibroblasts are also able to recruit macrophages. This can be by biochemical means, via fibroblast-secreted TSP-1 [97] or by physical means via fibroblasts-mediated construction of ECM tunnels that modulate macrophage migration [106] and the mechanosensation by macrophages of contractile forces exerted by myofibroblast upon the ECM [107], although this latter mechanism has only so far been demonstrated in vitro.

TAMs and the ECM

Polarisation of macrophages during cutaneous wound healing is critical for the switch from pro-inflammatory to anti-inflammatory and pro-re-epithelisation phases. However these characteristics of macrophages also contribute to their roles in tumorigenesis. As such, macrophages polarised towards the M1 phenotypic state are associated with tumour-suppressive functions such as supporting CD8+ cytotoxic T-cell activity. Macrophages polarised towards the M2 state are anti-inflammatory and associated with tumour-promoting functions such as ECM remodelling, angiogenesis, stimulating cancer cell proliferation and metastasis. Functions of macrophages that promote wound healing, particularly in the later phases of the process, are favourable for tumour progression and often co-opted into the process.

The role of TAMs is perhaps best studied in the context of their interaction with CAFs. TAMs and CAFs can induce activation and recruitment of each other, in a manner similar to that observed in normal macrophages and tissue fibroblasts during wound healing. CAFs release fibroblast growth factors that induce M2 polarity in macrophages. M2 cells release TGF-β causing fibroblast reprogramming to a tumour-promoting CAF state. It has been shown using an orthotopic CRC model, that recruitment of TAMs to the tumour site regulated a suite of ECM-associated genes which established key characteristics of the TME. Many of these ECM components were expressed by the TAMs directly, but others were produced by CAFs under the influence of TAMs [108].

One mechanism by which tumour-suppressing TAMs inhibit tumour growth is by remodelling tumour ECM to destroy the physical barrier that inhibits the recruitment of cytotoxic T cells, and thereby enhancing their access to the tumour. This mechanism has been harnessed for chimeric antigen receptor (CAR) macrophage cell therapy in a proof of principle study by engineering macrophages to express the CD147 antigen, a cell surface protein of the immunoglobulin superfamily with the ability to enhance the production of MMPs by an as yet imprecisely defined mechanism. CAR-147 macrophages expressing the CD147 antigen reduced ECM density and collagen content by increasing the production of MMP-3, MMP-14, and MMP-15, thereby facilitating the entry of cytotoxic T cells into 4T1 cell allografts in immune-competent mice [109].

MCs and their modulation of inflammation

MCs are key effector cells of the innate immune system whose principal function is to aid in host defence against pathogens. Present in almost all tissues in the body, they are largely located where the organism interfaces with the environment such in the skin, gastrointestinal tract, and respiratory tracts. Upon activation, MCs rapidly degranulate and release their protein-packed granules that contain pre-made chemokines, cytokines growth factors, histamine, and proteases. They are also capable of de novo synthesis of a suite of other proteins that are also released following activation [110].

MC activity is required for proper wound healing [111], as well as to restrain scar ECM formation via the deployment of proteases [112,113]. Functionally, MCs exert a protective effect by directly targeting pathogens, but also recruit and activate neutrophils, macrophages, and T lymphocytes [114,115]. The release of histamine can facilitate vasoactivation [116], and pro-inflammatory cytokines IL-6 and IL-8, and VEGF can recruit other immune cells [117]. Additionally, MCs can contribute to the polarisation of macrophages in wounds [118]. The MC-specific proteases chymase and tryptase are also released during early inflammation and serve to break down the ECM [119]. But conversely, MCs can also modulate fibroblast activation [120] and ECM production.

MCs are recruited to the wound site by chemokines such as CCL-2 and CCL-5, as well as growth factors such as stem cell factor (SCF). Membrane-bound SCF on dermal fibroblasts can induce MC progenitor maturation by binding to surface c-kit [121]. The SCF stimulation can also aid in the adherence of MCs to FN [122]. Thrombin has also been found to stabilise MC adhesion to FN and laminin, in addition to activation to release cytokines and proteases [123]. Indeed, the attachment to FN has been shown to stabilise MCs and modulate pro-inflammatory cytokine release [124,125].

MCs have roles in ECM production, processing and degradation. MC proteases can degrade collagen [126], FN [127], laminin [112,113] and activate the latent forms of MMP-2 and MMP-9 [128]. MC produced prostaglandin D2 and leukotriene D4, carbopeptidase A, and tryptase can stimulate dermal fibroblast collagen production and proliferation [129]. MC chymase can activate endothelin-1, which in turn can induce myofibroblast differentiation and collagen synthesis in dermal fibroblasts through the RhoA/Rho kinase pathways [130]

In the settings of infection, MCs can also recruit neutrophils by producing TNF [131], tryptase [132], and leukotrienes, LTB4 and LTC4 [133].

MC-mediated regulation of the tumour ECM

MCs accumulate in tumour margins in proximity to vessels. In early tumours, it is thought that MCs are pro-inflammatory and recruit cells to support anti-tumour functions. However during later stages MCs mediate angiogenesis and metastasis. The observed heterogeneity in the functions of MCs in tumour settings, and the specific consequences of MC accumulation likely depends on tumour type and grade [134].

As in normal tissue, MCs in tumours can produce both pro- and anti-inflammatory cytokines and chemokines, growth factors to stimulate angiogenesis, and proteinases to remodel the ECM. MC proteases chymase and tryptase can stimulate angiogenesis in cutaneous carcinoma, and tryptase has mitogenic effect on dermal fibroblasts. Additionally, these proteases can activate the pro-forms of MMP-2 and MMP-9 to facilitate ECM remodelling [135]. However, the precise conditions under which MCs exhibit pro- vs. anti-tumour effects upon the ECM and the suite of immune cells with which they cooperate in order to exert these effects still remain somewhat ill-defined. Nevertheless, the accumulation of MC at tumour margins suggests a role for these enigmatic cells in tumorigenesis.

T cells in tissue homoeostasis and cancer

T lymphocytes are not crucial for effective wound healing; however they can contribute to each relevant process by secreting cytokines and growth factors to regulate immune response and fibroblast activation.

In an murine excisional wounding model, it was observed that CD4+ T cells are the predominant subtype recruited into the wound [136]. This time-course study by Chen et al. demonstrated that CD4+ and CD8+ T cells migrated into cutaneous wounds, with CD4+ T cells present from day 1 (peak numbers were observed at day 5 and had dropped by day 10), and CD8+ cells recruited from day 3 (peak numbers observed at day 7 and had dropped by day 21). On day 7, CD4+ T cells expressed IFN-γ, TGF-β, IL-10, and IL-17 at high levels (relative to their naïve counterparts). CD8+ T cells had high IFN-γ and TGF-β. These data show that the recruitment of T cells into the healing wound is tightly regulated and that the recruited cells produce a suite of inflammatory cytokines with established functions in wound healing. Surprisingly however, skin deficient in CD4+ or CD8+ T lymphocytes did not exhibit impaired wound closure, collagen content, or angiogenesis, suggesting that T cells are not required for a normal wound healing response in this model, even though depletion of CD8+ T cells in wounds reduced but did not abolish the recruitment of macrophages and neutrophils [136].

T lymphocytes are also observed to be recruited early in the healing wound process in a severe combined immunodeficient (SCID) murine model and were predominantly of the CD4+ subtype [137]. However, in this case, T- and B-cell deficiency during cutaneous wound healing in SCID mice caused fibrotic scarring, elevated leucocyte and macrophage infiltration, and little neo-angiogenesis, but nevertheless accelerated healing compared with wildtype controls. Restoration of CD4+ (but not CD4 and Treg) cells could restore wound healing [137]. These contrasting observations suggest a complex role for subsets of T cells in wound healing via modulation of other immune cell types and it is still unclear whether this involves modulation of the ECM.

T cells in immune regulation

Regulatory T lymphocytes

The majority of CD4+ T cells that accumulate after wounding are regulatory T lymphocytes (Treg) [138]. These cells are recruited from secondary lymphoid organs and are highly activated phenotype. The major function of Tregs in skin wounding is to limit the accumulation of IFN-γ producing T cells and pro-inflammatory macrophages in the wounded skin [138]. FoxP3-expressing regulatory T cells (Treg) are essential for tissue immune homoeostasis. The EGFR, well known to be expressed on epidermal cells, is also expressed on Tregs as is the EGFR ligand, amphiregulin (AREG). In full-thickness wounds, Tregs accumulate in skin, and limit IFN-γ production and M1 macrophage accumulation [138]. Depletion of Tregs ‘early’ in wound healing caused greater attenuation of wound healing than when depletion was carried out later in the process, suggesting a role for Tregs in regulating the early immune response of wound healing. Direct roles for Tregs in ECM-mediated regulation of the immune response remains to be uncovered.

T cells and fibroblast activation and consequences for ECM homoeostasis

T cells can produce heparin-binding epidermal growth factor-like growth factor (HB-EGF) and basic FGF (bFGF), factors that stimulate fibroblast proliferation and activation [139]. bFGF is synthesised by CD4+ and CD8+ T lymphocytes, whereas HB-EGF is primarily produced by CD4+ T lymphocytes. CD4+ TH2 cells produce IL-13 and TAMPS to stimulate collagen synthesis [140], whereas TH1 cells release IFN-γ to degrade collagen [141] indirectly via the action of fibroblasts.

TH2 cytokines include IL-4, IL-5, IL-9, and IL-13, and many have established roles in ECM regulation. In skin, IL-13 can drive myofibroblast activation and increase collagen production [142]. Additionally, fibroblasts express receptors for IL-4 and it has also been shown to stimulate collagen production [143], suggesting that TH1 cells may also be able to stimulate the production of ECM in this way. IL-13 has been shown to induce POSTN production in asthmatic airway [144], further suggesting that T lymphocytes may regulate ECM production under certain pathologic conditions by activating fibroblasts within the local microenvironment.

T cells and ECM regulation in pathology

Although T cells do not play a critical role in wound healing, they are utilised in settings of infection and inflammation during healing. In these cases, T cells have the ability to actively remodel the ECM to aid in interstitial migration.

Activated CD4+ T cells can express mRNA for MMP-9, MT1-MMP, MT4-MMP (also designated MMP-17), ADAM-9, ADAM-15, ADAM-17, cathepsin L, urokinase-type plasminogen activator (uPA), TIMP-2, PAI-1, but local collagen lysis was not a driver of migration [145]. T-cell migration does not require the activity of proteases, but rather T cells use an amoeboid movement mechanism, driven by adaptive morphology permitting crawling along and between collagen [145] and fibronectin fibrils [146], via an αv-integrin-dependent mechanism [147] and involves the physical deformation of the ECM structure by the exertion of mechanical force.

Tumour-infiltrating lymphocytes

Treg cells have the most defined role in cutaneous wound healing, in which they facilitate wound re-epithelialisation by suppression of inflammation responses. In a similar manner, tumour Tregs accumulate in the tumour stroma and hinder anti-tumour immunity [148]. Although the CD4+FoxP3+ Treg subset of CD4+ cells are a significant portion, CD4+FoxP3 T helper subset of T lymphocytes also have significant pro-tumour activity. Additionally, CD8+ cytotoxic cells are also significant anti-tumour actors. The literature on T-cell immunity in cancer is vast, and a full discussion is outside the scope of this review. Only those studies relevant to ECM interactions will be discussed in this section.

In the context of ECM and immune cells, we highlight that ECM has a significant impact on T-cell accessibility to the tumour cells for their function. Overall, while the infiltration of T cells into solid tumours can correlate with positive prognosis, there are many factors that determine T-cell function within the tumour. The TME can impact the ability of T cells to enter the tumour, but also can have an effect on the cytotoxic activity of the lymphocytes within the tumour [149].

The distribution of T cells in the tumour environment could be guided by the presence of chemoattractants (such as CCL5) in the stroma, rather than the tumour cell regions. The composition of the stroma ECM can have a significant impact on the ability of the lymphocytes to enter the tumour mass. The density of the collagen in ECM can inhibit T-cell locomotion and their stimulation by cytokines cannot overcome this barrier [150]. Examination of human lung tumours determined that T lymphocytes prefer migration in tumour stroma regions containing low levels of FN and collagen [151], consistent with the observed ability of dense ECM to trap these cells.

The effect of collagen on T cells may not only influence migratory potential, but also tumour-infiltrating lymphocyte (TIL) activity. Collagens are the functional ligands for the immune inhibitory receptor LAIR1/CD305 [152]. T cells express LAIR-1 and cross-linking to the collagen ligand can inhibit cellular TCR-mediated function [153]. This effect of LAIR-1 immuno-suppression on CD8+ T cells potentiates PD-1/PD-L1 blockade resistance in lung tumours in mice [154], demonstrating a key mechanism by which ECM biology impacts upon the ability of the immune system to modulate tumour growth, with implications for immune therapy.

We have described so far how the regulation of the immune system during cutaneous wound healing begins with pro-inflammatory signals that potentiate the influx of immune cells that fight microbial invasion, but also commence the process of activating fibroblasts, stimulate proliferation of keratinocytes, and promote angiogenesis. Anti-inflammatory signals are then engaged to re-establish homoeostasis, with the restoration and remodelling of the ECM and completion of re-epithelialisation.

During cancer progression, the cellular interactions can exhibit these same pathways may become tumour suppressive or tumour promoting. TANs, TAMs, and TILs all participate in the establishment of the TME by engaging cellular process aligned to their functions in wound healing. An emerging concept is that immune cells, CAFs, and the ECM are regulated in a co-ordinated fashion downstream of signals from the tumour, with CAFs taking a lead role in the in situ differentiation of key components of the microenvironment [6].

In this review, we have discussed key aspects of the ECM and the tumour immune system that determine the course of the disease. What is clear is that cancers are able to co-opt the normal functions of immune cells such as those important for wound healing, as well as the ECM into the process of tumorigenesis and tumour progression, although these are modified by the pathological characteristics of the disease. Nevertheless, such divergences in the behaviour of genetically normal cells associated with tumours are key to uncovering the next generation of cancer targets and biomarkers to assist in managing this disease.

No datasets have been used in producing this work.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the Australian Government via a Research Training Program Ph.D. Scholarship (to C.-H.C.); the Australian Research Council Future Fellowship [grant number FT120100132 (to M.S.S.)]; the National Health and Medical Research Council [grant numbers GNT1103712, GNT1145319 and GNT1184809 (to M.S.S.)]; the Hospital Research Foundation and Vonbri Foundation grant (to M.S.S.); the Royal Adelaide Hospital Research Fund (to M.S.S.); and the Beat Cancer Project of the Cancer Council of South Australia (to M.S.S.).

αSMA

alpha smooth muscle actin

bFGF

basic FGF

CAF

cancer-associated fibroblast

CAR

chimeric antigen receptor

CRC

colorectal cancer

DAMP

damage-associated molecular pattern

ECM

extracellular matrix

ELN

elastin

ERα

estrogen receptor alpha

ERC

elastin receptor complex

FN

fibronectin

GAG

glycosaminoglycan

HB-EGF

heparin-binding epidermal growth factor-like growth factor

HGF

hepatocyte growth factor

IFN

interferon

IL

interleukin

LAIR1/CD305

leucocyte-associated immunoglobulin-like receptor

LTB4

leukotriene B4

MAPK

mitogen-activated protein kinase

MC

mast cell

MMP

matrix metalloproteinase

MT1-MMP

membrane type 1-matrix metalloproteinase

NET

neutrophil extracellular trap

OSCC

oral squamous cell carcinoma

PD-1

programmed cell death protein 1

PD-L1

programmed cell death ligand 1

PDGFRα

platelet-derived growth factor receptor alpha

PI3K

phosphoinositide 3-kinase

POSTN

periostin

ROCK

Rho-associated kinase

SCF

stem cell factor

SCID

severe combined immunodeficiency

SPP1

secreted phosphoprotein 1

TAM

tumour-associated macrophage

TAMP

tumour-associated molecular patterns

TAN

tumour-associated neutrophil

TGF

transforming growth factor

TME

tumour microenvironment

TIL

tumour-infiltrating lymphocyte

TIMP

tissue inhibitor of metalloproteinase

TNC

tenascin-C

TNF

tumour necrosis factor

Treg

regulatory T lymphocyte/cell

TSP

thrombospondin

VEGF

vascular endothelial growth factor

YAP

Yes-associated protein

1.
Soysal
S.D.
,
Tzankov
A.
and
Muenst
S.E.
(
2015
)
Role of the tumor microenvironment in breast cancer
.
Pathobiology
82
,
142
152
[PubMed]
2.
Allen
M.
and
Louise Jones
J.
(
2011
)
Jekyll and Hyde: the role of the microenvironment on the progression of cancer
.
J. Pathol.
223
,
163
177
3.
Cox
T.R.
(
2021
)
The matrix in cancer
.
Nat. Rev. Cancer
21
,
217
238
4.
Ping
Q.
,
Yan
R.
,
Cheng
X.
,
Wang
W.
,
Zhong
Y.
,
Hou
Z.
et al.
(
2021
)
Cancer-associated fibroblasts: overview, progress, challenges, and directions
.
Cancer Gene Ther.
28
,
984
999
5.
Poltavets
V.
,
Kochetkova
M.
,
Pitson
S.M.
and
Samuel
M.S.
(
2018
)
The role of the extracellular matrix and its molecular and cellular regulators in cancer cell plasticity
.
Front. Oncol.
8
,
431
[PubMed]
6.
Kochetkova
M.
and
Samuel
M.S.
(
2021
)
Differentiation of the tumor microenvironment: are CAFs the organizer?
Trends Cell Biol.
,
[PubMed]
7.
Sahai
E.
,
Astsaturov
I.
,
Cukierman
E.
,
DeNardo
D.G.
,
Egeblad
M.
,
Evans
R.M.
et al.
(
2020
)
A framework for advancing our understanding of cancer-associated fibroblasts
.
Nat. Rev. Cancer
20
,
174
186
[PubMed]
8.
Balta
E.
,
Wabnitz
G.H.
and
Samstag
Y.
(
2021
)
Hijacked immune cells in the tumor microenvironment: molecular mechanisms of immunosuppression and cues to improve T cell-based immunotherapy of solid tumors
.
Int. J. Mol. Sci.
22
,
5736
[PubMed]
9.
Grivennikov
S.I.
,
Greten
F.R.
and
Karin
M.
(
2010
)
Immunity, inflammation, and cancer
.
Cell
140
,
883
899
[PubMed]
10.
Deligne
C.
and
Midwood
K.S.
(
2021
)
Macrophages and extracellular matrix in breast cancer: partners in crime or protective allies?
Front. Oncol.
11
,
620773
,
[PubMed]
11.
Shaw
T.J.
and
Martin
P.
(
2009
)
Wound repair at a glance
.
J. Cell Sci.
122
,
3209
3213
[PubMed]
12.
Szpaderska
A.M.
and
DiPietro
L.A.
(
2005
)
Inflammation in surgical wound healing: friend or foe?
Surgery
137
,
571
573
[PubMed]
13.
Arwert
E.N.
,
Hoste
E.
and
Watt
F.M.
(
2012
)
Epithelial stem cells, wound healing and cancer
.
Nat. Rev. Cancer
12
,
170
180
[PubMed]
14.
Dvorak
H.F.
(
1986
)
Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing
.
N. Engl. J. Med.
315
,
1650
1659
[PubMed]
15.
Eming
S.A.
,
Krieg
T.
and
Davidson
J.M.
(
2007
)
Inflammation in wound repair: molecular and cellular mechanisms
.
J. Invest. Dermatol.
127
,
514
525
[PubMed]
16.
Deonarine
K.
,
Panelli
M.C.
,
Stashower
M.E.
,
Jin
P.
,
Smith
K.
,
Slade
H.B.
et al.
(
2007
)
Gene expression profiling of cutaneous wound healing
.
J. Transl. Med.
5
,
11
[PubMed]
17.
Coussens
L.M.
and
Werb
Z.
(
2002
)
Inflammation and cancer
.
Nature
420
,
860
867
[PubMed]
18.
O’Byrne
K.J.
and
Dalgleish
A.G.
(
2001
)
Chronic immune activation and inflammation as the cause of malignancy
.
Br. J. Cancer
85
,
473
483
[PubMed]
19.
Frantz
C.
,
Stewart
K.M.
and
Weaver
V.M.
(
2010
)
The extracellular matrix at a glance
.
J. Cell Sci.
123
,
4195
4200
[PubMed]
20.
Lu
P.
,
Weaver
V.M.
and
Werb
Z.
(
2012
)
The extracellular matrix: a dynamic niche in cancer progression
.
J. Cell Biol.
196
,
395
406
[PubMed]
21.
Gregorio
I.
,
Braghetta
P.
,
Bonaldo
P.
and
Cescon
M.
(
2018
)
Collagen VI in healthy and diseased nervous system
.
Dis. Model. Mech.
11
,
dmm032946
[PubMed]
22.
Johan
M.Z.
and
Samuel
M.S.
(
2019
)
Rho-ROCK signaling regulates tumor-microenvironment interactions
.
Biochem. Soc. Trans.
47
,
101
108
[PubMed]
23.
Sun
Z.
,
Costell
M.
and
Fassler
R.
(
2019
)
Integrin activation by talin, kindlin and mechanical forces
.
Nat. Cell Biol.
21
,
25
31
[PubMed]
24.
Ranade
S.S.
,
Syeda
R.
and
Patapoutian
A.
(
2015
)
Mechanically activated ion channels
.
Neuron
87
,
1162
1179
[PubMed]
25.
Meyaard
L.
,
Adema
G.J.
,
Chang
C.
,
Woollatt
E.
,
Sutherland
G.R.
,
Lanier
L.L.
et al.
(
1997
)
LAIR-1, a novel inhibitory receptor expressed on human mononuclear leukocytes
.
Immunity
7
,
283
290
[PubMed]
26.
Boyle
S.T.
,
Poltavets
V.
,
Kular
J.
,
Pyne
N.T.
,
Sandow
J.J.
,
Lewis
A.C.
et al.
(
2020
)
ROCK-mediated selective activation of PERK signalling causes fibroblast reprogramming and tumour progression through a CRELD2-dependent mechanism
.
Nat. Cell Biol.
22
,
882
895
[PubMed]
27.
Samuel
M.S.
,
Lopez
J.I.
,
McGhee
E.J.
,
Croft
D.R.
,
Strachan
D.
,
Timpson
P.
et al.
(
2011
)
Actomyosin-mediated cellular tension drives increased tissue stiffness and β-catenin activation to induce epidermal hyperplasia and tumor growth
.
Cancer Cell
19
,
776
791
[PubMed]
28.
Rath
N.
,
Morton
J.P.
,
Julian
L.
,
Helbig
L.
,
Kadir
S.
,
Mcghee
E.J.
et al.
(
2017
)
ROCK signaling promotes collagen remodeling to facilitate invasive pancreatic ductal adenocarcinoma tumor cell growth
.
EMBO Mol. Med.
9
,
198
218
[PubMed]
29.
Kular
J.
,
Scheer
K.G.
,
Pyne
N.T.
,
Allam
A.H.
,
Pollard
A.N.
,
Magenau
A.
et al.
(
2015
)
A negative regulatory mechanism involving 14-3-3zeta limits signaling downstream of ROCK to regulate tissue stiffness in epidermal homeostasis
.
Dev. Cell
35
,
759
774
[PubMed]
30.
Nukuda
A.
,
Sasaki
C.
,
Ishihara
S.
,
Mizutani
T.
,
Nakamura
K.
,
Ayabe
T.
et al.
(
2015
)
Stiff substrates increase YAP-signaling-mediated matrix metalloproteinase-7 expression
.
Oncogenesis
4
,
e165
[PubMed]
31.
Liu
F.
,
Lagares
D.
,
Choi
K.M.
,
Stopfer
L.
,
Marinkovic
A.
,
Vrbanac
V.
et al.
(
2015
)
Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis
.
Am. J. Physiol. Lung Cell. Mol. Physiol.
308
,
L344
L357
[PubMed]
32.
Boyle
S.T.
and
Samuel
M.S.
(
2016
)
Mechano-reciprocity is maintained between physiological boundaries by tuning signal flux through the Rho-associated protein kinase
.
Small GTPases
7
,
139
146
[PubMed]
33.
Naba
A.
,
Clauser
K.R.
,
Hoersch
S.
,
Liu
H.
,
Carr
S.A.
and
Hynes
R.O.
(
2012
)
The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices
.
Mol. Cell. Proteomics
11
,
M111.014647
[PubMed]
34.
Boyle
S.T.
,
Mittal
P.
,
Kaur
G.
,
Hoffmann
P.
,
Samuel
M.S.
and
Klingler-Hoffmann
M.
(
2020
)
Uncovering tumor–stroma inter-relationships using MALDI mass spectrometry imaging
.
J. Proteome Res.
19
,
4093
4103
[PubMed]
35.
Vennin
C.
,
Chin
V.T.
,
Warren
S.C.
,
Lucas
M.C.
,
Herrmann
D.
,
Magenau
A.
et al.
(
2017
)
Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis
.
Sci. Transl. Med.
9
,
384
eaai8504
[PubMed]
36.
Kauppila
S.
,
Stenback
F.
,
Risteli
J.
,
Jukkola
A.
and
Risteli
L.
(
1998
)
Aberrant type I and type III collagen gene expression in human breast cancer in vivo
.
J. Pathol.
186
,
262
268
[PubMed]
37.
Kalluri
R.
(
2003
)
Basement membranes: structure, assembly and role in tumour angiogenesis
.
Nat. Rev. Cancer
3
,
422
433
[PubMed]
38.
Amenta
P.S.
,
Hadad
S.
,
Lee
M.T.
,
Barnard
N.
,
Li
D.
and
Myers
J.C.
(
2003
)
Loss of types XV and XIX collagen precedes basement membrane invasion in ductal carcinoma of the female breast
.
J. Pathol.
199
,
298
308
[PubMed]
39.
Amenta
P.
,
Briggs
K.
,
Xu
K.
,
Gamboa
E.
,
Jukkola
A.
,
Li
D.
et al.
(
2000
)
Type XV collagen in human colonic adenocarcinomas has a different distribution than other basement membrane zone proteins
.
Hum. Pathol.
31
,
359
366
[PubMed]
40.
Murphy
G.
and
Nagase
H.
(
2008
)
Progress in matrix metalloproteinase research
.
Mol. Aspects Med.
29
,
290
308
[PubMed]
41.
Piperigkou
Z.
,
Kyriakopoulou
K.
,
Koutsakis
C.
,
Mastronikolis
S.
and
Karamanos
N.K.
(
2021
)
Key matrix remodeling enzymes: functions and targeting in cancer
.
Cancers
13
,
1441
[PubMed]
42.
Amara
N.
,
Tholen
M.
and
Bogyo
M.
(
2018
)
Chemical tools for selective activity profiling of endogenously expressed MMP-14 in multicellular models
.
ACS Chem. Biol.
13
,
2645
2654
[PubMed]
43.
Szabova
L.
,
Chrysovergis
K.
,
Yamada
S.S.
and
Holmbeck
K.
(
2008
)
MT1-MMP is required for efficient tumor dissemination in experimental metastatic disease
.
Oncogene
27
,
3274
3281
[PubMed]
44.
Hynes
R.O.
and
Naba
A.
(
2012
)
Overview of the matrisome–an inventory of extracellular matrix constituents and functions
.
Cold Spring Harb. Perspect. Biol.
4
,
a004903
45.
Vennin
C.
,
Melenec
P.
,
Rouet
R.
,
Nobis
M.
,
Cazet
A.S.
,
Murphy
K.J.
et al.
(
2019
)
CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan
.
Nat. Commun.
10
,
3637
[PubMed]
46.
Sarrazin
S.
,
Lamanna
W.C.
and
Esko
J.D.
(
2011
)
Heparan sulfate proteoglycans
.
Cold Spring Harb. Perspect. Biol.
3
,
a004952
a
47.
Chang
J.
and
Chaudhuri
O.
(
2019
)
Beyond proteases: basement membrane mechanics and cancer invasion
.
J. Cell Biol.
218
,
2456
2469
[PubMed]
48.
Paszek
M.J.
,
Dufort
C.C.
,
Rossier
O.
,
Bainer
R.
,
Mouw
J.K.
,
Godula
K.
et al.
(
2014
)
The cancer glycocalyx mechanically primes integrin-mediated growth and survival
.
Nature
511
,
319
325
[PubMed]
49.
Schwarzbauer
J.E.
and
Desimone
D.W.
(
2011
)
Fibronectins, their fibrillogenesis, and in vivo functions
.
Cold Spring Harb. Perspect. Biol.
3
,
a005041
50.
Sottile
J.
and
Hocking
D.C.
(
2002
)
Fibronectin polymerization regulates the composition and stability of extracellular matrix fibrils and cell-matrix adhesions
.
Mol. Biol. Cell
13
,
3546
3559
[PubMed]
51.
Erdogan
B.
,
Ao
M.
,
White
L.M.
,
Means
A.L.
,
Brewer
B.M.
,
Yang
L.
et al.
(
2017
)
Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin
.
J. Cell Biol.
216
,
3799
3816
[PubMed]
52.
Petersen
E.
,
Wagberg
F.
and
Angquist
K.A.
(
2002
)
Serum concentrations of elastin-derived peptides in patients with specific manifestations of atherosclerotic disease
.
Eur. J. Vasc. Endovasc. Surg.
24
,
440
444
[PubMed]
53.
Chen
Y.
,
Klingen
T.A.
,
Wik
E.
,
Aas
H.
,
Vigeland
E.
,
Liestol
K.
et al.
(
2014
)
Breast cancer stromal elastosis is associated with mammography screening detection, low Ki67 expression and favourable prognosis in a population-based study
.
Diagn. Pathol.
9
,
230
[PubMed]
54.
Bennasroune
A.
,
Romier-Crouzet
B.
,
Blaise
S.
,
Laffargue
M.
,
Efremov
R.G.
,
Martiny
L.
et al.
(
2019
)
Elastic fibers and elastin receptor complex: Neuraminidase-1 takes the center stage
.
Matrix Biol.
84
,
57
67
[PubMed]
55.
Tenti
P.
and
Vannucci
L.
(
2020
)
Lysyl oxidases: linking structures and immunity in the tumor microenvironment
.
Cancer Immunol. Immunother.
69
,
223
235
[PubMed]
56.
Scandolera
A.
,
Odoul
L.
,
Salesse
S.
,
Guillot
A.
,
Blaise
S.
,
Kawecki
C.
et al.
(
2016
)
The Elastin receptor complex: a unique matricellular receptor with high anti-tumoral potential
.
Front. Pharmacol.
7
,
32
[PubMed]
57.
Orend
G.
and
Chiquet-Ehrismann
R.
(
2006
)
Tenascin-C induced signaling in cancer
.
Cancer Lett.
244
,
143
163
[PubMed]
58.
Oskarsson
T.
,
Acharyya
S.
,
Zhang
X.H.-F.
,
Vanharanta
S.
,
Tavazoie
S.F.
,
Morris
P.G.
et al.
(
2011
)
Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs
.
Nat. Med.
17
,
867
874
[PubMed]
59.
Insua‐Rodríguez
J.
,
Pein
M.
,
Hongu
T.
,
Meier
J.
,
Descot
A.
,
Lowy
C.M.
et al.
(
2018
)
Stress signaling in breast cancer cells induces matrix components that promote chemoresistant metastasis
.
EMBO Mol. Med.
10
,
e9003
[PubMed]
60.
González-González
L.
and
Alonso
J.
(
2018
)
Periostin: a matricellular protein with multiple functions in cancer development and progression
.
Front. Oncol.
8
,
225
[PubMed]
61.
Siriwardena
B.S.M.S.
,
Kudo
Y.
,
Ogawa
I.
,
Kitagawa
M.
,
Kitajima
S.
,
Hatano
H.
et al.
(
2006
)
Periostin is frequently overexpressed and enhances invasion and angiogenesis in oral cancer
.
Br. J. Cancer
95
,
1396
1403
[PubMed]
62.
Baril
P.
,
Gangeswaran
R.
,
Mahon
P.C.
,
Caulee
K.
,
Kocher
H.M.
,
Harada
T.
et al.
(
2007
)
Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: role of the β4 integrin and the PI3k pathway
.
Oncogene
26
,
2082
2094
[PubMed]
63.
Yu
B.
,
Wu
K.
,
Wang
X.
,
Zhang
J.
,
Wang
L.
,
Jiang
Y.
et al.
(
2018
)
Periostin secreted by cancer-associated fibroblasts promotes cancer stemness in head and neck cancer by activating protein tyrosine kinase 7
.
Cell Death Dis.
9
,
1082
64.
Malanchi
I.
,
Santamaria-Martínez
A.
,
Susanto
E.
,
Peng
H.
,
Lehr
H.-A.
,
Delaloye
J.-F.
et al.
(
2012
)
Interactions between cancer stem cells and their niche govern metastatic colonization
.
Nature
481
,
85
89
65.
Eming
S.A.
,
Krieg
T.
and
Davidson
J.M.
(
2007
)
Inflammation in wound repair: molecular and cellular mechanisms
.
J. Invest. Dermatol.
127
,
514
525
[PubMed]
66.
Wang
S.
,
Voisin
M.B.
,
Larbi
K.Y.
,
Dangerfield
J.
,
Scheiermann
C.
,
Tran
M.
et al.
(
2006
)
Venular basement membranes contain specific matrix protein low expression regions that act as exit points for emigrating neutrophils
.
J. Exp. Med.
203
,
1519
1532
[PubMed]
67.
Fischer
R.S.
,
Sun
X.
,
Baird
M.A.
,
Hourwitz
M.J.
,
Seo
B.R.
,
Pasapera
A.M.
et al.
(
2021
)
Contractility, focal adhesion orientation, and stress fiber orientation drive cancer cell polarity and migration along wavy ECM substrates
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
22
,
e2021135118
68.
Wolf
K.
,
Te Lindert
M.
,
Krause
M.
,
Alexander
S.
,
Te Riet
J.
,
Willis
A.L.
et al.
(
2013
)
Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force
.
J. Cell Biol.
201
,
1069
1084
[PubMed]
69.
Pittman
K.
and
Kubes
P.
(
2013
)
Damage-associated molecular patterns control neutrophil recruitment
.
J. Innate Immun.
5
,
315
323
70.
Kim
M.H.
,
Liu
W.
,
Borjesson
D.L.
,
Curry
F.R.
,
Miller
L.S.
,
Cheung
A.L.
et al.
(
2008
)
Dynamics of neutrophil infiltration during cutaneous wound healing and infection using fluorescence imaging
.
J. Invest. Dermatol.
128
,
1812
1820
[PubMed]
71.
De Oliveira
S.
,
Rosowski
E.E.
and
Huttenlocher
A.
(
2016
)
Neutrophil migration in infection and wound repair: going forward in reverse
.
Nat. Rev. Immunol.
16
,
378
391
[PubMed]
72.
Devalaraja
R.M.
,
Nanney
L.B.
,
Du
J.
,
Qian
Q.
,
Yu
Y.
,
Devalaraja
M.N.
et al.
(
2000
)
Delayed wound healing in CXCR2 knockout mice
.
J. Invest. Dermatol.
115
,
234
244
[PubMed]
73.
Theilgaard-Monch
K.
,
Knudsen
S.
,
Follin
P.
and
Borregaard
N.
(
2004
)
The transcriptional activation program of human neutrophils in skin lesions supports their important role in wound healing
.
J. Immunol.
172
,
7684
7693
[PubMed]
74.
Yin
C.
and
Heit
B.
(
2018
)
Armed for destruction: formation, function and trafficking of neutrophil granules
.
Cell Tissue Res.
371
,
455
471
[PubMed]
75.
Genschmer
K.R.
,
Russell
D.W.
,
Lal
C.
,
Szul
T.
,
Bratcher
P.E.
,
Noerager
B.D.
et al.
(
2019
)
Activated PMN exosomes: pathogenic entities causing matrix destruction and disease in the lung
.
Cell
176
,
113
126
[PubMed]
76.
Korkmaz
B.
,
Moreau
T.
and
Gauthier
F.
(
2008
)
Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physiopathological functions
.
Biochimie
90
,
227
242
[PubMed]
77.
Shamamian
P.
,
Schwartz
J.D.
,
Pocock
B.J.
,
Monea
S.
,
Whiting
D.
,
Marcus
S.G.
et al.
(
2001
)
Activation of progelatinase A (MMP-2) by neutrophil elastase, cathepsin G, and proteinase-3: a role for inflammatory cells in tumor invasion and angiogenesis
.
J. Cell. Physiol.
189
,
197
206
[PubMed]
78.
Delclaux
C.
,
Delacourt
C.
,
D'Ortho
M.P.
,
Boyer
V.
,
Lafuma
C.
and
Harf
A.
(
1996
)
Role of gelatinase B and elastase in human polymorphonuclear neutrophil migration across basement membrane
.
Am. J. Respir. Cell Mol. Biol.
14
,
288
295
[PubMed]
79.
Wilgus
T.A.
,
Roy
S.
and
McDaniel
J.C.
(
2013
)
Neutrophils and wound repair: positive actions and negative reactions
.
Adv. Wound Care
2
,
379
388
[PubMed]
80.
Gaudry
M.
,
Bregerie
O.
,
Andrieu
V.
,
El Benna
J.
,
Pocidalo
M.A.
and
Hakim
J.
(
1997
)
Intracellular pool of vascular endothelial growth factor in human neutrophils
.
Blood
90
,
4153
4161
[PubMed]
81.
Ardi
V.C.
,
Kupriyanova
T.A.
,
Deryugina
E.I.
and
Quigley
J.P.
(
2007
)
Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis
.
Proc. Natl. Acad. Sci. U.S.A.
104
,
20262
20267
[PubMed]
82.
Curaj
A.
,
Schumacher
D.
,
Rusu
M.
,
Staudt
M.
,
Li
X.
,
Simsekyilmaz
S.
et al.
(
2020
)
Neutrophils modulate fibroblast function and promote healing and scar formation after murine myocardial infarction
.
Int. J. Mol. Sci.
21
,
3685
[PubMed]
83.
Chrysanthopoulou
A.
,
Mitroulis
I.
,
Apostolidou
E.
,
Arelaki
S.
,
Mikroulis
D.
,
Konstantinidis
T.
et al.
(
2014
)
Neutrophil extracellular traps promote differentiation and function of fibroblasts
.
J. Pathol.
233
,
294
307
[PubMed]
84.
Jamieson
T.
,
Clarke
M.
,
Steele
C.W.
,
Samuel
M.S.
,
Neumann
J.
,
Jung
A.
et al.
(
2012
)
Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis
.
J. Clin. Invest.
122
,
3127
3144
[PubMed]
85.
Mishalian
I.
,
Bayuh
R.
,
Levy
L.
,
Zolotarov
L.
,
Michaeli
J.
and
Fridlender
Z.G.
(
2013
)
Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression
.
Cancer Immunol. Immunother.
62
,
1745
1756
[PubMed]
86.
Foekens
J.A.
,
Ries
C.
,
Look
M.P.
,
Gippner-Steppert
C.
,
Klijn
J.G.
and
Jochum
M.
(
2003
)
The prognostic value of polymorphonuclear leukocyte elastase in patients with primary breast cancer
.
Cancer Res.
63
,
337
341
[PubMed]
87.
Fridlender
Z.G.
,
Sun
J.
,
Kim
S.
,
Kapoor
V.
,
Cheng
G.
,
Ling
L.
et al.
(
2009
)
Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN
.
Cancer Cell
16
,
183
194
[PubMed]
88.
Germann
M.
,
Zangger
N.
,
Sauvain
M.O.
,
Sempoux
C.
,
Bowler
A.D.
,
Wirapati
P.
et al.
(
2020
)
Neutrophils suppress tumor-infiltrating T cells in colon cancer via matrix metalloproteinase-mediated activation of TGFbeta
.
EMBO Mol. Med.
12
,
e10681
[PubMed]
89.
Nozawa
H.
,
Chiu
C.
and
Hanahan
D.
(
2006
)
Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis
.
Proc. Natl. Acad. Sci. U.S.A.
103
,
12493
12498
[PubMed]
90.
Wada
Y.
,
Yoshida
K.
,
Tsutani
Y.
,
Shigematsu
H.
,
Oeda
M.
,
Sanada
Y.
et al.
(
2007
)
Neutrophil elastase induces cell proliferation and migration by the release of TGF-alpha, PDGF and VEGF in esophageal cell lines
.
Oncol. Rep.
17
,
161
167
[PubMed]
91.
Houghton
A.M.
,
Rzymkiewicz
D.M.
,
Ji
H.
,
Gregory
A.D.
,
Egea
E.E.
,
Metz
H.E.
et al.
(
2010
)
Neutrophil elastase-mediated degradation of IRS-1 accelerates lung tumor growth
.
Nat. Med.
16
,
219
223
[PubMed]
92.
Chua
F.
and
Laurent
G.J.
(
2006
)
Neutrophil elastase: mediator of extracellular matrix destruction and accumulation
.
Proc. Am. Thorac. Soc.
3
,
424
427
[PubMed]
93.
Albrengues
J.
,
Shields
M.A.
,
Ng
D.
,
Park
C.G.
,
Ambrico
A.
,
Poindexter
M.E.
et al.
(
2018
)
Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice
.
Science
361
,
eaao4227
[PubMed]
94.
Goren
I.
,
Allmann
N.
,
Yogev
N.
,
Schurmann
C.
,
Linke
A.
,
Holdener
M.
et al.
(
2009
)
A transgenic mouse model of inducible macrophage depletion: effects of diphtheria toxin-driven lysozyme M-specific cell lineage ablation on wound inflammatory, angiogenic, and contractive processes
.
Am. J. Pathol.
175
,
132
147
[PubMed]
95.
Mirza
R.
,
DiPietro
L.A.
and
Koh
T.J.
(
2009
)
Selective and specific macrophage ablation is detrimental to wound healing in mice
.
Am. J. Pathol.
175
,
2454
2462
[PubMed]
96.
Low
Q.E.
,
Drugea
I.A.
,
Duffner
L.A.
,
Quinn
D.G.
,
Cook
D.N.
,
Rollins
B.J.
et al.
(
2001
)
Wound healing in MIP-1alpha(-/-) and MCP-1(-/-) mice
.
Am. J. Pathol.
159
,
457
463
[PubMed]
97.
Moodley
Y.
,
Rigby
P.
,
Bundell
C.
,
Bunt
S.
,
Hayashi
H.
,
Misso
N.
et al.
(
2003
)
Macrophage recognition and phagocytosis of apoptotic fibroblasts is critically dependent on fibroblast-derived thrombospondin 1 and CD36
.
Am. J. Pathol.
162
,
771
779
[PubMed]
98.
Hesketh
M.
,
Sahin
K.B.
,
West
Z.E.
and
Murray
R.Z.
(
2017
)
Macrophage phenotypes regulate scar formation and chronic wound healing
.
Int. J. Mol. Sci.
18
,
1545
[PubMed]
99.
Wu
K.
,
Lin
K.
,
Li
X.
,
Yuan
X.
,
Xu
P.
,
Ni
P.
et al.
(
2020
)
Redefining tumor-associated macrophage subpopulations and functions in the tumor microenvironment
.
Front. Immunol.
11
,
1731
[PubMed]
100.
Shook
B.
,
Xiao
E.
,
Kumamoto
Y.
,
Iwasaki
A.
and
Horsley
V.
(
2016
)
CD301b+ macrophages are essential for effective skin wound healing
.
J. Invest. Dermatol.
136
,
1885
1891
[PubMed]
101.
Spiller
K.L.
,
Anfang
R.R.
,
Spiller
K.J.
,
Ng
J.
,
Nakazawa
K.R.
,
Daulton
J.W.
et al.
(
2014
)
The role of macrophage phenotype in vascularization of tissue engineering scaffolds
.
Biomaterials
35
,
4477
4488
[PubMed]
102.
Honda
T.
,
Tsutsui
S.
,
Kohchi
C.
and
Inagawa
H.
(
2020
)
Enhanced effect of hyaluronan and elastin synthesis in fibroblasts through lipopolysaccharide-activated macrophages
.
Anticancer Res.
40
,
4681
4685
[PubMed]
103.
Peters
T.
,
Sindrilaru
A.
,
Hinz
B.
,
Hinrichs
R.
,
Menke
A.
,
Al-Azzeh
E.A.
et al.
(
2005
)
Wound-healing defect of CD18(-/-) mice due to a decrease in TGF-beta1 and myofibroblast differentiation
.
EMBO J.
24
,
3400
3410
[PubMed]
104.
Desmouliere
A.
,
Geinoz
A.
,
Gabbiani
F.
and
Gabbiani
G.
(
1993
)
Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts
.
J. Cell Biol.
122
,
103
111
[PubMed]
105.
Montesano
R.
and
Orci
L.
(
1988
)
Transforming growth factor beta stimulates collagen-matrix contraction by fibroblasts: implications for wound healing
.
Proc. Natl. Acad. Sci. U.S.A.
85
,
4894
4897
[PubMed]
106.
Ford
A.J.
,
Orbach
S.M.
and
Rajagopalan
P.
(
2019
)
Fibroblasts stimulate macrophage migration in interconnected extracellular matrices through tunnel formation and fiber alignment
.
Biomaterials
209
,
88
102
[PubMed]
107.
Pakshir
P.
,
Alizadehgiashi
M.
,
Wong
B.
,
Coelho
N.M.
,
Chen
X.
,
Gong
Z.
et al.
(
2019
)
Dynamic fibroblast contractions attract remote macrophages in fibrillar collagen matrix
.
Nat. Commun.
10
,
1850
[PubMed]
108.
Afik
R.
,
Zigmond
E.
,
Vugman
M.
,
Klepfish
M.
,
Shimshoni
E.
,
Pasmanik-Chor
M.
et al.
(
2016
)
Tumor macrophages are pivotal constructors of tumor collagenous matrix
.
J. Exp. Med.
213
,
2315
2331
[PubMed]
109.
Zhang
W.
,
Liu
L.
,
Su
H.
,
Liu
Q.
,
Shen
J.
,
Dai
H.
et al.
(
2019
)
Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix
.
Br. J. Cancer
121
,
837
845
[PubMed]
110.
Galli
S.J.
,
Gaudenzio
N.
and
Tsai
M.
(
2020
)
Mast cells in inflammation and disease: recent progress and ongoing concerns
.
Annu. Rev. Immunol.
38
,
49
77
[PubMed]
111.
Zimmermann
C.
,
Troeltzsch
D.
,
Gimenez-Rivera
V.A.
,
Galli
S.J.
,
Metz
M.
,
Maurer
M.
et al.
(
2019
)
Mast cells are critical for controlling the bacterial burden and the healing of infected wounds
.
Proc. Natl. Acad. Sci. U.S.A.
116
,
20500
20504
[PubMed]
112.
Vangansewinkel
T.
,
Geurts
N.
,
Quanten
K.
,
Nelissen
S.
,
Lemmens
S.
,
Geboes
L.
et al.
(
2016
)
Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6
.
FASEB J.
30
,
2040
2057
[PubMed]
113.
Vangansewinkel
T.
,
Lemmens
S.
,
Geurts
N.
,
Quanten
K.
,
Dooley
D.
,
Pejler
G.
et al.
(
2019
)
Mouse mast cell protease 4 suppresses scar formation after traumatic spinal cord injury
.
Sci. Rep.
9
,
3715
[PubMed]
114.
Dudeck
A.
,
Koberle
M.
,
Goldmann
O.
,
Meyer
N.
,
Dudeck
J.
,
Lemmens
S.
et al.
(
2019
)
Mast cells as protectors of health
.
J. Allergy Clin. Immunol.
144
,
S4
S18
[PubMed]
115.
Succar
J.
,
Giatsidis
G.
,
Yu
N.
,
Hassan
K.
,
Khouri
R.
Jr
,
Gurish
M.F.
et al.
(
2019
)
Mouse mast cell protease-4 recruits leukocytes in the inflammatory phase of surgically wounded skin
.
Adv. Wound Care
8
,
469
475
[PubMed]
116.
Jin
H.
,
Koyama
T.
,
Hatanaka
Y.
,
Akiyama
S.
,
Takayama
F.
and
Kawasaki
H.
(
2006
)
Histamine-induced vasodilation and vasoconstriction in the mesenteric resistance artery of the rat
.
Eur. J. Pharmacol.
529
,
136
144
[PubMed]
117.
Metz
M.
,
Grimbaldeston
M.A.
,
Nakae
S.
,
Piliponsky
A.M.
,
Tsai
M.
and
Galli
S.J.
(
2007
)
Mast cells in the promotion and limitation of chronic inflammation
.
Immunol. Rev.
217
,
304
328
[PubMed]
118.
Tellechea
A.
,
Bai
S.
,
Dangwal
S.
,
Theocharidis
G.
,
Nagai
M.
,
Koerner
S.
et al.
(
2020
)
Topical application of a mast cell stabilizer improves impaired diabetic wound healing
.
J. Invest. Dermatol.
140
,
901
911
[PubMed]
119.
Schwartz
L.B.
(
1990
)
Neutral Proteases of Mast Cells
,
Karger
120.
Yamamoto
T.
,
Hartmann
K.
,
Eckes
B.
and
Krieg
T.
(
2000
)
Mast cells enhance contraction of three-dimensional collagen lattices by fibroblasts by cell-cell interaction: role of stem cell factor/c-kit
.
Immunology
99
,
435
439
[PubMed]
121.
Leist
M.
,
Sunder
C.A.
,
Drube
S.
,
Zimmermann
C.
,
Geldmacher
A.
,
Metz
M.
et al.
(
2017
)
Membrane-bound stem cell factor is the major but not only driver of fibroblast-induced murine skin mast cell differentiation
.
Exp. Dermatol.
26
,
255
262
[PubMed]
122.
Dastych
J.
and
Metcalfe
D.D.
(
1994
)
Stem cell factor induces mast cell adhesion to fibronectin
.
J. Immunol.
152
,
213
219
[PubMed]
123.
Vliagoftis
H.
(
2002
)
Thrombin induces mast cell adhesion to fibronectin: evidence for involvement of protease-activated receptor-1
.
J. Immunol.
169
,
4551
4558
[PubMed]
124.
Hamawy
M.M.
,
Oliver
C.
,
Mergenhagen
S.E.
and
Siraganian
R.P.
(
1992
)
Adherence of rat basophilic leukemia (RBL-2H3) cells to fibronectin-coated surfaces enhances secretion
.
J. Immunol.
149
,
615
621
[PubMed]
125.
Ra
C.
,
Yasuda
M.
,
Yagita
H.
and
Okumura
K.
(
1994
)
Fibronectin receptor integrins are involved in mast cell activation
.
J. Allergy Clin. Immunol.
94
,
625
628
[PubMed]
126.
Zhao
X.O.
,
Lampinen
M.
,
Rollman
O.
,
Sommerhoff
C.P.
,
Paivandy
A.
and
Pejler
G.
(
2021
)
Mast cell chymase affects the functional properties of primary human airway fibroblasts: implications for asthma
.
J. Allergy Clin. Immunol
149
,
2
718
727
,
[PubMed]
127.
Tchougounova
E.
,
Pejler
G.
and
Abrink
M.
(
2003
)
The chymase, mouse mast cell protease 4, constitutes the major chymotrypsin-like activity in peritoneum and ear tissue. A role for mouse mast cell protease 4 in thrombin regulation and fibronectin turnover
.
J. Exp. Med.
198
,
423
431
[PubMed]
128.
Tchougounova
E.
,
Lundequist
A.
,
Fajardo
I.
,
Winberg
J.O.
,
Abrink
M.
and
Pejler
G.
(
2005
)
A key role for mast cell chymase in the activation of pro-matrix metalloprotease-9 and pro-matrix metalloprotease-2
.
J. Biol. Chem.
280
,
9291
9296
[PubMed]
129.
Abe
M.
,
Kurosawa
M.
,
Ishikawa
O.
and
Miyachi
Y.
(
2000
)
Effect of mast cell-derived mediators and mast cell-related neutral proteases on human dermal fibroblast proliferation and type I collagen production
.
J. Allergy Clin. Immunol.
106
,
S78
S84
[PubMed]
130.
Kiya
K.
,
Kubo
T.
,
Kawai
K.
,
Matsuzaki
S.
,
Maeda
D.
,
Fujiwara
T.
et al.
(
2017
)
Endothelial cell-derived endothelin-1 is involved in abnormal scar formation by dermal fibroblasts through RhoA/Rho-kinase pathway
.
Exp. Dermatol.
26
,
705
712
[PubMed]
131.
Malaviya
R.
,
Ikeda
T.
,
Ross
E.
and
Abraham
S.N.
(
1996
)
Mast cell modulation of neutrophil influx and bacterial clearance at sites of infection through TNF-alpha
.
Nature
381
,
77
80
[PubMed]
132.
Huang
C.
,
Friend
D.S.
,
Qiu
W.T.
,
Wong
G.W.
,
Morales
G.
,
Hunt
J.
et al.
(
1998
)
Induction of a selective and persistent extravasation of neutrophils into the peritoneal cavity by tryptase mouse mast cell protease 6
.
J. Immunol.
160
,
1910
1919
[PubMed]
133.
Malaviya
R.
and
Abraham
S.N.
(
2000
)
Role of mast cell leukotrienes in neutrophil recruitment and bacterial clearance in infectious peritonitis
.
J. Leukoc. Biol.
67
,
841
846
[PubMed]
134.
Komi
D.E.A.
and
Redegeld
F.A.
(
2020
)
Role of mast cells in shaping the tumor microenvironment
.
Clin. Rev. Allergy Immunol.
58
,
313
325
[PubMed]
135.
Coussens
L.M.
,
Raymond
W.W.
,
Bergers
G.
,
Laig-Webster
M.
,
Behrendtsen
O.
,
Werb
Z.
et al.
(
1999
)
Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis
.
Genes Dev.
13
,
1382
1397
[PubMed]
136.
Chen
L.
,
Mehta
N.D.
,
Zhao
Y.
and
DiPietro
L.A.
(
2014
)
Absence of CD4 or CD8 lymphocytes changes infiltration of inflammatory cells and profiles of cytokine expression in skin wounds, but does not impair healing
.
Exp. Dermatol.
23
,
189
194
[PubMed]
137.
Wang
X.
,
Balaji
S.
,
Steen
E.H.
,
Li
H.
,
Rae
M.M.
,
Blum
A.J.
et al.
(
2019
)
T lymphocytes attenuate dermal scarring by regulating inflammation, neovascularization, and extracellular matrix remodeling
.
Adv. Wound Care
8
,
527
537
[PubMed]
138.
Nosbaum
A.
,
Prevel
N.
,
Truong
H.A.
,
Mehta
P.
,
Ettinger
M.
,
Scharschmidt
T.C.
et al.
(
2016
)
Cutting edge: regulatory T cells facilitate cutaneous wound healing
.
J. Immunol.
196
,
2010
2014
[PubMed]
139.
Blotnick
S.
,
Peoples
G.E.
,
Freeman
M.R.
,
Eberlein
T.J.
and
Klagsbrun
M.
(
1994
)
T lymphocytes synthesize and export heparin-binding epidermal growth factor-like growth factor and basic fibroblast growth factor, mitogens for vascular cells and fibroblasts: differential production and release by CD4+ and CD8+ T cells
.
Proc. Natl. Acad. Sci. U.S.A.
91
,
2890
2894
[PubMed]
140.
Firszt
R.
,
Francisco
D.
,
Church
T.D.
,
Thomas
J.M.
,
Ingram
J.L.
and
Kraft
M.
(
2014
)
Interleukin-13 induces collagen type-1 expression through matrix metalloproteinase-2 and transforming growth factor-beta1 in airway fibroblasts in asthma
.
Eur. Respir. J.
43
,
464
473
[PubMed]
141.
Mikko
M.
,
Fredriksson
K.
,
Wahlstrom
J.
,
Eriksson
P.
,
Grunewald
J.
and
Skold
C.M.
(
2008
)
Human T cells stimulate fibroblast-mediated degradation of extracellular matrix in vitro
.
Clin. Exp. Immunol.
151
,
317
325
[PubMed]
142.
O'Reilly
S.
,
Ciechomska
M.
,
Fullard
N.
,
Przyborski
S.
and
van Laar
J.M.
(
2016
)
IL-13 mediates collagen deposition via STAT6 and microRNA-135b: a role for epigenetics
.
Sci. Rep.
6
,
25066
[PubMed]
143.
Nguyen
J.K.
,
Austin
E.
,
Huang
A.
,
Mamalis
A.
and
Jagdeo
J.
(
2020
)
The IL-4/IL-13 axis in skin fibrosis and scarring: mechanistic concepts and therapeutic targets
.
Arch. Dermatol. Res.
312
,
81
92
[PubMed]
144.
Suzaki
I.
,
Kawano
S.
,
Komiya
K.
,
Tanabe
T.
,
Akaba
T.
,
Asano
K.
et al.
(
2017
)
Inhibition of IL-13-induced periostin in airway epithelium attenuates cellular protein expression of MUC5AC
.
Respirology
22
,
93
100
[PubMed]
145.
Wolf
K.
,
Muller
R.
,
Borgmann
S.
,
Brocker
E.B.
and
Friedl
P.
(
2003
)
Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases
.
Blood
102
,
3262
3269
[PubMed]
146.
Fernandes
N.R.J.
,
Reilly
N.S.
,
Schrock
D.C.
,
Hocking
D.C.
,
Oakes
P.W.
and
Fowell
D.J.
(
2020
)
CD4(+) T cell interstitial migration controlled by fibronectin in the inflamed skin
.
Front. Immunol.
11
,
1501
[PubMed]
147.
Overstreet
M.G.
,
Gaylo
A.
,
Angermann
B.R.
,
Hughson
A.
,
Hyun
Y.M.
,
Lambert
K.
et al.
(
2013
)
Inflammation-induced interstitial migration of effector CD4(+) T cells is dependent on integrin alphaV
.
Nat. Immunol.
14
,
949
958
[PubMed]
148.
Togashi
Y.
,
Shitara
K.
and
Nishikawa
H.
(
2019
)
Regulatory T cells in cancer immunosuppression - implications for anticancer therapy
.
Nat. Rev. Clin. Oncol.
16
,
356
371
[PubMed]
149.
Chen
Q.
,
Wang
W.C.
and
Evans
S.S.
(
2003
)
Tumor microvasculature as a barrier to antitumor immunity
.
Cancer Immunol. Immunother.
52
,
670
679
[PubMed]
150.
Applegate
K.G.
,
Balch
C.M.
and
Pellis
N.R.
(
1990
)
In vitro migration of lymphocytes through collagen matrix: arrested locomotion in tumor-infiltrating lymphocytes
.
Cancer Res.
50
,
7153
7158
[PubMed]
151.
Salmon
H.
,
Franciszkiewicz
K.
,
Damotte
D.
,
Dieu-Nosjean
M.C.
,
Validire
P.
,
Trautmann
A.
et al.
(
2012
)
Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors
.
J. Clin. Invest.
122
,
899
910
[PubMed]
152.
Lebbink
R.J.
,
de Ruiter
T.
,
Adelmeijer
J.
,
Brenkman
A.B.
,
van Helvoort
J.M.
,
Koch
M.
et al.
(
2006
)
Collagens are functional, high affinity ligands for the inhibitory immune receptor LAIR-1
.
J. Exp. Med.
203
,
1419
1425
[PubMed]
153.
Maasho
K.
,
Masilamani
M.
,
Valas
R.
,
Basu
S.
,
Coligan
J.E.
and
Borrego
F.
(
2005
)
The inhibitory leukocyte-associated Ig-like receptor-1 (LAIR-1) is expressed at high levels by human naive T cells and inhibits TCR mediated activation
.
Mol. Immunol.
42
,
1521
1530
[PubMed]
154.
Peng
D.H.
,
Rodriguez
B.L.
,
Diao
L.
,
Chen
L.
,
Wang
J.
,
Byers
L.A.
et al.
(
2020
)
Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8(+) T cell exhaustion
.
Nat. Commun.
11
,
4520
[PubMed]

Author notes

*

These authors contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).