Abstract
Cumulative evidence shows that fibrogenic stroma and stiff extracellular matrix (ECM) not only result from tumor growth but also play pivotal roles in cellular transformation and tumor initiation. This emerging concept may largely account for the increased cancer risk associated with environmental fibrogenic agents, such as asbestos and silica, and with chronic conditions that are fibrogenic, such as obesity and diabetes. It may also contribute to poor outcomes in patients treated with certain chemotherapeutics that can promote fibrosis, such as bleomycin and methotrexate. Although the mechanistic details of this phenomenon are still being unraveled, we provide an overview of the experimental evidence linking fibrogenic stroma and tumor initiation. In this Review, we will summarize the causes and consequences of fibrous stroma and how this stromal cue is transmitted to the nuclei of parenchymal cells through a physical continuum from the ECM to chromatin, as well as ECM-dependent biochemical signaling that contributes to cellular transformation.
INTRODUCTION
The presence of dense collagenous stroma is a strong indicator of tumor aggressiveness and therapeutic resistance. Desmoplasia, the formation of fibrous tumor stroma, accounts for the hard, lumpy appearance of many types of carcinomas (1) and is a hallmark of cancer presumed to result from the stromal response to malignant progression (2). However, cumulative evidence has shown that desmoplasia may actually precede the emergence of malignant cells. Desmoplasia is often detected in normal breast tissues by mammography and is a strong risk factor for breast cancer (3). Myofibroblasts (MyoFbs), the cells responsible for desmoplasia, are often found in premalignant lesions or tumor-adjacent normal tissues and are proposed as key players in tumor initiation (4-6). The presence of MyoFbs in noncancerous tissues is considered an indicator of cancer predisposition, and various types of tissue injuries and chronic wounds harboring abundant MyoFbs are considered premalignant (7).
MyoFbs, which are associated with tissues undergoing repair and with tumors, are highly contractile and secretory cells derived from local fibroblasts, epithelia, and other cell types (8). Although their major function is to promote wound repair, their excessive activation dysregulates the production of extracellular matrix (ECM) and growth factors, ultimately inducing tissue fibrosis and hypertrophic scars. The abundance of MyoFbs in tumor stroma led to the description of tumors as “wounds that never heal” (9). It is well established that the abundant MyoFbs, indicative of reactive stroma, greatly contribute to tumor progression and aggressiveness, and emerging evidence substantiates the direct involvement of MyoFbs in tumor initiation (6, 10).
The establishment of fibrous stroma by MyoFb accumulation stimulates parenchymal cells biochemically and mechanically. The stromal signals are eventually transmitted to the parenchymal nuclei through the physical continuum between the ECM and chromatin, which modulates the expression of specific genes to adapt to the changes or to remodel the stroma. Parenchymal and stromal cells thus coevolve through such dynamic and reciprocal regulation. Here, we present an overview of the advances in our understanding of the roles of fibrotic stroma in cellular transformation and tumor initiation and the mechanistic bases of these effects.
PHENOTYPIC INFLUENCE BY STROMAL FIBROBLASTS
Fibroblasts, one of the most abundant cell populations in the stroma, are a heterogeneous group of spindle-shaped cells of mesenchymal origin with phenotypes ranging from quiescent, noncontractile fibroblasts to activated, contractile MyoFbs (11). These subpopulations produce distinct sets of ECM proteins, including collagens (types I, III, and V), proteoglycans, fibronectin, elastin, and glycosaminoglycans, which altogether constitute the interstitial matrix (IM), as well as paracrine factors and ECM-modifying enzymes for establishing the IM scaffolds (12). The phenotype of fibroblasts strongly influences both the microenvironment and the phenotype of parenchymal cells. Fibroblasts manifest distinct tissue- and context-specific phenotypes and gene expression profiles through differential epigenetic mechanisms (13). Even within the same tissue, there are functionally distinct fibroblast subpopulations (Fig. 1) (14). There are three general categories of fibroblasts: normal fibroblasts (NFs), cancer-associated fibroblasts (CAFs), and MyoFbs.
Fig. 1. Heterogeneity of fibroblasts.
Normal tissue stroma mainly contains normal fibroblasts (NFs), which are quiescent and spindle-shaped. The cancer-associated fibroblasts (CAFs) in tumor stroma include NF-like cells and myofibroblasts (MyoFbs), which are proliferative and contractile. Several major markers distinguish NFs from CAFs and between different NF and CAF subtypes. NFs are involved in tumor suppression through neighbor suppression (the direct inhibition of the growth of abnormal cells through cell-cell contacts), paracrine signaling that inhibits cell growth and migration, and maintenance of normal ECM. CAFs promote tumor initiation and metastasis through the production of growth factors, chemokines, and fibrous ECM, as well as immunosuppression.
Subgroups of fibroblasts
NFs are quiescent mesenchymal cells, comprising several distinct subpopulations (Table 1). In contrast to the CAFs that assist tumor progression, NFs suppress tumor initiation and metastasis. Coculturing premalignant or malignant breast epithelial cells with NFs in three-dimensional (3D) conditions reverts the phenotype of the epithelia, restoring quiescence (15). In animals, NFs cotransplanted with breast cancer cells suppress tumor growth (16). Such tumor-suppressive functions of NFs are sustained through the production of a group of proteins [including bone morphogenetic protein receptor 2, transforming growth factor β (TGF-β) receptor 2, and forkhead box F1 (FOXF-1) and FOXF-2] that protect NFs from becoming MyoFbs under the influence of cancer cells (17).
Table 1.
NF subpopulations identified by single-cell RNA sequencing.
| Cell type | Marker expression | Morphology | Potential functions | Spatial distribution | Reference |
|---|---|---|---|---|---|
| – | lin− CD90+ CD39+ CD26− COL6A5+ | Spindle shaped | Anti-inflammatory | Upper dermis | (245) |
| Preadipocytes | lin− CD90+ CD36+ | Epithelioid | – | Lower dermis | |
| Pericytes | lin− CD90+ CD39− RGS5+ | – | – | Reticular dermis | |
| Uncharacterized | lin− CD90+ CD39+ CD26+ | Spindle shaped | – | Reticular dermis | |
| Uncharacterized | lin− CD90+ CD39− RGS5− | – | – | Reticular dermis | |
| Mesenchymal cells | SFRP2/DPP4 | Small and tubular with narrow elongated nuclei | Matrix deposition | Between collagen bundles | (246) |
| Bone marrow stromal cells | FMO1/LSP1 | Large with larger nuclei | Inflammatory cell retention | Interstitial and perivascular | |
| Progenitor or stem cells | CRABP1 | – | – | – | |
| COL11A1 | – | Connective tissue cell differentiation | – | ||
| FMO2 | – | – | – | ||
| PRG4 | – | – | – | ||
| C2ORF40 | – | – | – | ||
| Secretory reticular | WISP2, SLPI, CTHRC1, MFAP5, and TSPAN8 | – | Collagen/ECM production and organization | Reticular dermis | (247) |
| Secretory papillary | APCDD1, ID1, WIF1, COL18A1, and PTGDS | – | Collagen/ECM production and organization | Papillary dermis | |
| Proinflammatory | CCL19, APOE, CXCL2, CXCL3, and EFEMP1 | – | Inflammatory response | Whole dermis | |
| Mesenchymal | ASPN, POSTN, GPC3, TNN, and SFRP1 | – | ECM organization and skeletal system development | Reticular dermis |
The antitumor functions of NFs are mediated by at least four mechanisms. First, NFs suppress the growth of abnormal cells by direct cell-cell contact through a phenomenon termed “neighbor suppression” (18). Second, NFs secrete tumor-suppressive paracrine factors, such as tumor necrosis factor (TNF) and interleukin-6 (IL-6); protease inhibitors, such as whey acidic protein four-disulfide core domain 1 (WFDC1), which is an inhibitor of matrix metalloproteinase 9 (MMP-9), and tissue inhibitors of metalloproteases (TIMPs), which inhibit both MMPs and a disintegrin and metalloproteinases (ADAMs); and inhibitors of protumorigenic TGF-β–like proteoglycans and fibrillins (19-21). Third, NFs help maintain the IM integrity that elicits tumor suppression in a manner dependent on the cytoskeletal regulator Ras homolog family member A (RhoA). RhoA knockout in NFs reorganizes vimentin, increases the cells’ cytoplasmic rigidity, and induces the expression of proinflammatory genes that promote tumor growth (22). Last, NFs produce tissue-specific ECM that is substantially softer than CAF-derived ECM and suppresses tumor growth (23). Culturing cancer cells on NF-derived ECM inhibits the histone demethylase JMJD1a, an activator of Yes-associated protein 1 (YAP) and Tafazzin (TAZ), both of which encode mechanosensitive transcription factors that promote cancer cell growth. Conversely, culturing cancer cells on CAF-derived ECM induces YAP and TAZ expression and exacerbates cancer cell growth (24).
NFs of different tissue origins, host species, and ages show different degrees of tumor suppression. For example, NFs from naked mole rats, which have an extremely long life span and high resistance to developing cancers, produce high molecular weight hyaluronan (HMWHA) that is five times larger than human or mouse hyaluronic acid (HA). HMWHA protects tissues and organs from aging and oncogenic transformation through early contact inhibition (25). Conversely, NFs from aged humans produce decreased amounts of hyaluronic and proteoglycan link protein (HAPLN1), which enhances ECM alignment to stimulate tumor cell invasion and protumor immunity (26).
The efficiency of NF-mediated tumor suppression is also influenced by the nature of the tumor cells. NFs produce the TGF-β inhibitor asporin, a small leucine-rich proteoglycan, in response to TGF-β signaling from adjacent cancer cells. However, asporin production is influenced by the subtype of cancer cells. For example, luminal-type breast cancer induces asporin in NFs, whereas triple-negative breast cancer inhibits it through production of high amounts of IL-1β (27). Similarly, NFs secrete an antitumor protein, slit homolog 2 (SLIT2), which binds to the receptor roundabout homolog 1 (ROBO1) on cancer cells. Although the SLIT2-ROBO1 axis exerts antitumor functions in breast cancer by inhibiting nuclear translocation of β-catenin (28), it exerts protumor functions in osteosarcoma by increasing the glycolytic enzyme, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 (PFKFB2) (29).
CAFs are fibroblasts that have been activated by fibrotic signals from neoplasms and exhibit highly proliferative and migratory properties (30). Compared with NFs, CAFs are larger and contain more indented nuclei, conspicuous nucleoli, and cytosolic filament bundles (31). Similar to NFs, CAFs are a heterogeneous group of cells with different gene expression profiles and cellular origins (Table 2). They include myofibroblastic (myCAF) and inflammatory (iCAF) types as well as some intermediate types (32, 33). CAFs isolated from tumors promote tumor initiation, growth, and progression under experimental conditions in vitro and in vivo (16). Coculturing normal or premalignant breast epithelia with CAFs induces aberrant epithelial proliferation and morphogenesis (15), and cotransplantation of premalignant breast cells with either genetically engineered CAFs overexpressing hepatocyte growth factor (HGF) and TGF-β1 or fibroblasts from inflammatory lesions into mice induces mammary tumors (6, 16). Such protumor effects of CAFs are attributed to several major functions. Most notably, CAFs produce paracrine factors [such as HGF, TGF-β, and C-X-C motif chemokine ligand 12 (CXCL12)] that promote tumor growth and invasion (34). They also produce fibrous ECM proteins (such as collagen and elastin) that align and confine the epithelia, thus altering their morphology and polarity, ultimately inducing epithelial-mesenchymal transition (EMT) (16). These ECM fibers also exert mechanical force on the epithelia, activating oncogenic signals (34). CAFs also activate metabolic pathways to buffer toxic metabolites produced by cancer cells, such as lactate, thus sustaining tumor growth (35). By producing MMPs that remodel the ECM as they move, CAFs create a path that facilitates collective cancer cell invasion (36). They induce the angiogenesis that enables continued tumor growth through the production of proangiogenic factors, such as fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor 1 (VEGF1) (37, 38). Although some CAFs help establish an anti-inflammatory tumor microenvironment (TME) by recruiting immunosuppressive cells, such as M2-type macrophages and myeloid-derived suppressor cells (MDSCs; immature myeloid cells) (39), iCAFs produce proinflammatory factors like CXCL2, leukemia inhibitory factor (LIF), and IL-6, highlighting the heterogeneity among CAFs (32, 40, 41). Last, CAFs help induce drug resistance in tumors (see below for more details) (42).
Table 2. CAF subpopulations identified by single-cell RNA sequencing.
LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma.
| CAF subpopulations | Functions | Marker expression | Cell types | Cancer type | Reference |
|---|---|---|---|---|---|
| MyoFbs (myCAFs) | Actin cytoskeleton organization | α-SMAhi | Pancreas | Adenocarcinoma | (32) |
| Inflammatory CAFs (iCAFs) | Immunomodulation and monocyte recruitment | α-SMAlow and IL-6+ | |||
| Antigen-presenting CAFs (apCAFs) | Immunomodulation | MHC class II and CD74 | (248) | ||
| Cancer-promoting CAFs (pCAFs) | Promotion of tumor growth | Meflinneg α-SMApos | Pancreas | Adenocarcinoma | (57) |
| Cancer-restraining CAFs (rCAFs) | Suppression of tumor growth | Meflinpos α-SMAneg | |||
| PDPN CAFs (pCAFs) | Cell migration, immune regulation, wound healing, and extracellular fiber organization | PDPN, Cxcl12, Saa3, Cxcl1, IL16, Acta2, and Fbn1 | Breast | Adenocarcinoma | (249) |
| S100A4 CAFs (sCAFs) | Antigen presentation and ECM remodeling and protein folding and metabolic regulation | Spp1hiS100A4low | |||
| Spp1lowS100A4hi | |||||
| Matrix CAFs (mCAFs) | ECM structure | PDGFRα and FBLN1 | Breast | Adenocarcinoma | (250) |
| Vascular CAFs (vCAFs) | Vascular development and angiogenesis | NID2, CD31, αSMA, and PDGFRβ | |||
| Cycling CAFs (cCAFs) | Cell cycle, cell division, and angiogenesis | Ki-67 and PDGFRβ | |||
| Developmental CAFs (dCAFs) | Cell differentiation and development | SCRG1 and SOX9 | |||
| CAF-S1 | Immunosuppression | CD29Med FAPHi FSP1Low-Hi αSMAHi PDGFRβMed-Hi CAV1Low | Breast | Adenocarcinoma | (40, 230) |
| CAF-S2 | – | CD29Low FAPNeg FSP1Neg-Low αSMANeg PDGFRβNeg CAV1Neg | |||
| CAF-S3 | – | CD29Med FAPNeg FSP1Med-Hi αSMANeg-Low PDGFRβMed CAV1Neg-Low | |||
| CAF-S4 | Oxidative metabolism | CD29Hi FAPNeg FSP1Low-MedαSMAHi PDGFRβLow-Med CAV1Neg-Low | |||
| CAF-A | – | MMP-2, decorin, and collagen type 1 α2 | Colorectal | Adenocarcinoma | (251) |
| CAF-B | – | α-SMA, PDGFA, and transgelin | |||
| CAF-1 | Tissue remodeling | MMP-11, CAV1, and THY1 | Head and neck | Squamous | (252) |
| CAF-2 | Tissue remodeling | JUN, FOS, TGFBR3, and FGF7 | |||
| Cluster 1 | – | COL10A1 | Lung | Squamous/adenocarcinoma | (253) |
| Cluster 2 | – | COL4A1, α-SMAhi, MEF2C, MYH11, and ITGA7 | |||
| Cluster 4 | – | PLA2G2A | |||
| Cluster 5 | – | MMP-3 and mTOR | |||
| Cluster 6 | – | FIGF | |||
| Cluster 7 | – | CCL2 and mTOR | |||
| Proto-inflammatory fibroblasts (CAF-1) | Inflammation | SGK1, APOE, CXCL2, and PLA2G2A | Lung | Adenocarcinoma (LUAD)/squamous cell carcinoma (LUSC) | (33) |
| Proto-MyoFbs (CAF-2) | Matrix remodeling | CXCL12, IGF1, and COL1A1 | |||
| Catabolic proto-MyoFbs (CAF-3) | Catabolism and matrix remodeling | POSTN, FAP, and COL1A1 | |||
| MyoFbs (CAF-4) | Matrix remodeling | α-SMA, POSTN, and FAP |
Protumor activities of CAFs could also be attributed to mutations. Although CAFs were initially reported to be genetically normal (43, 44), next-generation sequencing technology has revoked this notion. For example, 10 to 50% of CAFs from colorectal tumors were found to harbor somatic copy number alterations, the most common of which is the gain of a whole chromosome 7, indicating clonal expansion (45). In addition, CAFs from skin squamous cell carcinoma often have NOTCH1 amplification and overexpression, which prevents the kinase ataxia-telangiectasia mutated (ATM) from initiating the DNA damage response (DDR) and growth arrest upon ultraviolet exposure, thus promoting their proliferation potential (46). Moreover, prostate cancer cells often promote Tumor protein p53 (TP53) expression in CAFs, which suppresses their growth and confers selective pressure on a subpopulation of CAFs that are TP53-null and therefore highly proliferative (47). Likewise, CAFs of more than 25% of sporadic breast tumors harbor TP53 mutations in association with lymph node metastases (48). Such stromal mutations could be, in part, due to high amounts of MMP-3, which increases reactive oxygen species (ROS) that elicit genotypic stress (49). Mutant stromal cells could also be derived from cancer cells undergoing EMT (see below).
In addition to mutations, the epigenetic profiles of stromal fibroblasts greatly change during cancer development (50). Such changes manifest as global genomic hypo- and hypermethylation of select genes (51). For example, TGF-β signaling induces NFs to increase their production of LIF, which then promotes expression of the DNA methyltransferase 3b (DNMT3b). DNMT3b–mediated repression of the gene encoding Src homology region 2 domain–containing phosphatase-1 (SHP-1) causes constitutive activation of Janus kinase 1 (JNK1)–signal transducer and activator of transcription 3 (STAT3) signaling, which promotes the conversion of NFs to CAFs (52). Epigenetic reprogramming also mediates the conversion of mesenchymal stem cells (MSCs) to CAFs in pancreatic ductal adenocarcinoma (PDAC). Lactate that accumulates in the PDAC TME induces α-ketoglutarate in MSCs, leading to the activation of the demethylase ten-eleven translocation (TET), which replaces cytosine methylation with hydroxymethylation, thus triggering MSC differentiation to CAFs (53). Furthermore, ~70% of human epidermal growth factor (EGF) receptor 2 (HER2)–positive breast tumors harbors CAFs with hypermethylation of the genes encoding the progesterone receptor (PGR), hydroxysteroid 17-β dehydrogenase 4 (HSD17B4), and cadherin 13 (CDH13), compared with 10% of HER2-negative tumors, highlighting the existence of cancer subtype–specific CAF profiles (54). See Polyak et al. (55) for a comprehensive review of CAF genetic and epigenetic profiles.
The heterogeneity among CAFs implies that some could promote tumor growth and others inhibit it, thereby contributing to the complexity of the TME. Whereas most studies report tumor-promoting effects of CAFs, one study reports that the depletion of α–smooth muscle actin (α-SMA)–positive CAFs worsens PDAC metastasis and prognosis (56). Other studies have identified cancer-promoting (pCAF; Meflin−/α-SMA+) versus cancer-restraining (rCAF; Meflin+/α-SMA−) CAF subpopulations in PDAC (57, 58) and linked Meflin+ CAFs to improved responsiveness of non–small cell lung carcinoma (NSCLC) to immune checkpoint inhibitors (57, 58).
MyoFbs comprise a subpopulation of CAFs (myCAFs) that are highly contractile and secretory. MyoFbs are also present at wound sites in the absence of tumors. MyoFbs arise from the activation of tissue-resident fibroblasts and other cell types, which are quiescent during tissue homeostasis, by tissue damage and wound healing signals. They produce paracrine factors and ECM proteins that promote wound healing (59). After wound healing is complete, as indicated by stress release through ECM softening or by the formation of granulation tissue, MyoFbs are cleared through apoptosis (60). Under the continuous wound healing signals present in fibrotic lesions and tumors, MyoFbs evade the termination signal and persist, exacerbating the disease conditions (9, 60). Activated fibroblasts in fibrotic tissues and tumor stroma are collectively referred to as MyoFbs because they are highly similar to one another both morphologically and functionally, with no definitive phenotypes or markers to distinguish between the two (61). Single-cell RNA sequencing has uncovered some differences between the transcriptomes of MyoFbs in fibrotic tissues and those in tumor stroma (Table 3), but the functional contributions of these differences are yet to be determined.
Table 3.
MyoFb subpopulations in fibrotic stroma and tumor stroma.
| MyoFbs in fibrotic stroma | MyoFbs in tumor stroma | |||||||
|---|---|---|---|---|---|---|---|---|
| Tissue | Disease | MyoFb precursors | Marker expression |
Reference | Disease | MyoFb precursors |
Marker expression |
Reference |
| Kidney | Kidney fibrosis* | Pericytes | NOTCH3+ RGS5+ PDGFRα− PDGFRβ+ | (254) | Kidney cancer–Wilms tumor* | – | α-SMA+ PLVAP+ PDGFRβ+ EMILIN1+ SLC14A1− | (255) |
| Fibroblast | MEG3+ PDGFRα+ PDGFRβ+ | |||||||
| Fibroblast | COLEC11+ CXCL12+ PDGFRβ+ | |||||||
| Lung | Pulmonary fibrosis* | Mesenchymal cells | α-SMA | (256) | Lung cancer (NSCLC)* | Mesenchymal cells | α-SMA, POSTN, and FAP | (33) |
| Idiopathic pulmonary fibrosis* | Unknown | PDGFRB, MYLK, NEBL, MYO10, MYO1D, RYR2, ITGA8, α-SMA, and COL | (257) | |||||
| Systemic sclerosis-associated interstitial lung disease (SSc-ILD)* | MFAP5hi fibroblasts | SFRP2, DPP4, COL10A1, DPEP1, TSPAN2, POSTN, and CTHRC1 | (258) | |||||
| Liver | Liver fibrosis* | Hepatic stellate cells | COL1A1, COL1A2, and COL3A1 | (259) | HCC† | Hepatic stellate cells | GFAP, LRAT, and PDGFRα | (260-263) |
| Liver cirrhosis* | Mesenchymal cells | PDGFRα+ and collagen | (264) | Fibrocytes | CD34 and collagen type 1 | (263, 265) | ||
| Pancreas | Chronic pancreatitis‡ | – | Tenascin Clo, α-SMA, desmin, and vimentin | (266) | PDAC‡ | – | Tenascin Chi, α-SMA, desmin, and vimentin | (266) |
| PDAC* | Fibroblasts | LRRC15 | (267) | |||||
| High-grade IPMN* | Fibroblasts | α-SMA+ CXCL12− DES− | (268) | |||||
| Skin | Cutaneous fibrosis§ | Adipocytes | COL5A1, COL5A2, and COL1A2 | (269) | Skin cancer§ | – | GREM1 | (270) |
| Bleomycin induced fibrosis† | Mesenchymal cells | CD29hi | (271) | |||||
Single-cell RNA sequencing.
Flow cytometry.
Transcriptome array.
Reverse transcription polymerase chain reaction.
MyoFbs are characterized by the production of α-SMA, which forms contractile stress fibers, and are distinct from other types of fibroblasts devoid of a contractile cytoskeleton (62). The high contractility of MyoFbs, along with increased ECM deposition and cross-linking, contributes to ECM stiffening (63). MyoFbs also manifest a stellate cell shape, unlike the spindle shape of NFs, as well as enhanced proliferation and migration, and the production of fibroblast activation protein (FAP), a marker of reactive stromal fibroblasts (61). MyoFbs are largely heterogeneous and originated from various types of locally residing cells; NFs are the main source in most tissues, but MyoFbs can also arise from epithelia, endothelia, pericytes, mesenchyme, smooth muscle, bone marrow, and immune cells (8, 64). In liver fibrosis, the major source of MyoFbs is the resident pericyte population, hepatic stellate cells (65). Circulating fibrocytes (bone marrow–derived CD34+ mesenchymal cells) can also be recruited and differentiate into α-SMA+ (CD34−) MyoFbs in the skin, liver, and lung (66).
MyoFb differentiation is primarily induced by paracrine factors, such as TGF-β and platelet-derived growth factor (PDGF), secreted from tumor cells and stromal cells (67). Differentiation of NFs to MyoFbs depends on the activation of the gene encoding α-SMA, ACTA2, which is otherwise inactivated by methylation of the promoter at three CpG islands. TGF-β signaling inhibits DNA methylation and derepresses ACTA2 transcription (68). Furthermore, TGF-β signaling promotes the activity of myocardin, a smooth muscle–specific transcriptional regulator involved in the expression of ACTA2 (69, 70). High expression of ACTA2 increases contractility and focal adhesions (FAs), both of which are hallmarks of MyoFbs (64). In contrast, transdifferentiation of epithelial cells into MyoFbs is driven by a form of TGF-β–induced EMT (type 2 EMT) that is associated with wound healing, wherein epithelial apical-basolateral polarity is converted into the mesenchymal front-rear polarity (71). This change is accomplished by a reduction in the expression of epithelial markers like E-cadherin, the acquisition of a spindle shape, and the increased expression of mesenchymal markers such as vimentin and ACTA2 (72). Type 2 EMT is implicated in various types of tissue and organ fibrosis associated with overactivation of TGF-β signals due to a reduction in SMAD specific E3 ubiquitin protein ligase 2 (SMURF2), a ubiquitin ligase that targets the TGF-β downstream effectors mothers against decapentaplegic homolog 2 (SMAD2) and SMAD3 for degradation (73). Other causative agents of type 2 EMT include hypoxia, collagenous ECM, cytokines (such as TGF-β and FGF2), inflammation, and ROS (72).
Protumor activities of MyoFbs
The abundance of α-SMA+ MyoFbs predicts poor prognosis and cancer recurrence (74). Coculturing cancer cells with MyoFbs or with MyoFb-conditioned medium promotes cancer cell invasiveness (75), and cotransplantation of MyoFbs with cancer cells into animals exacerbates tumor growth (76). This pathogenicity is attributed to the fibrogenic effects of MyoFbs, which are amplified by the TGF-β–induced infiltration of MyoFbs into the ECM and their recruitment α-SMA− stromal cells to become α-SMA+ MyoFbs (77). MyoFbs also produce large amounts of growth factors, ECM proteins, and ECM-modifying enzymes that together increase the rigidity and stability of the ECM, which both promotes tumor progression and invasion (see below) (8, 62) and compromises the termination of MyoFb activity (60). Moreover, as discussed above, MyoFbs and other CAFs activate specific metabolic pathways that buffer and recycle toxic metabolites produced by cancer cells, thus enabling continued cancer growth (35).
In addition to these roles in promoting tumor growth and progression, MyoFbs play pivotal roles in tumor initiation. Premalignant cells cotransplanted with MyoFbs establish tumors in animals, whereas premalignant cells alone fail to do so (6, 16). This is primarily because MyoFbs help construct the “premalignant niche,” wherein premalignant cells accumulate mutations that favor their transition into malignancy (78-80). Indeed, various types of chronic wounds and damaged tissue populated with MyoFbs can eventually give rise to neoplasms (7, 81), and the presence of MyoFb-like, α-SMA–high cells in dense breast tissue increases breast cancer risk by four- to sixfold (3, 82). It has long been known that α-SMA–high cells that resemble those in the breast cancer stroma are present in the stroma of nonmalignant breast lesions but are virtually absent from normal breast tissues (4). It is thus proposed that the emergence of MyoFbs in nonmalignant tissues indicates a predisposition to tumor initiation (6, 10), concordant with the analogy of tumors to “nonhealing ulcers” (9, 81) and the association of up to 20% of cancers with the chronic inflammation and tissue damage that also favor fibrosis (7, 83). Many types of tissue injuries harbor excessively activated MyoFbs and could develop into malignant scars. These include nonhealing ulcers in diabetic and immunocompromised patients, Marjolin ulcers, surgical incisions, stomach ulcers, ulcerative colitis, bronchitis, and cirrhosis (83-85). In fact, cirrhosis, which is fibrosis of the liver caused by viral infection, metabolic disease, drug toxicity, or alcoholism, has a high chance (20%) of becoming malignant (86). Other causes for MyoFb activation linked to tumor initiation and promotion include chronic metabolic disorders (obesity and diabetes), environmental agents (asbestos and silica), and chemotherapeutic drugs (bleomycin, methotrexate, and cisplatin) (87-90).
The tumor-initiating capabilities of MyoFbs are largely attributed to their production of large amounts of paracrine factors, ECM, and ECM-modifying enzymes (34). These paracrine factors include TGF-β; FGFs; insulin-like growth factors (IGF1 and IGF2); HGF; PDGF; VEGF; stem cell factor; TNF; stromal cell–derived factor 1 (also called CXCL12); and IL-1, IL-6, IL-8, and IL-10 (91). In particular, fibroblasts overexpressing TGF-β and HGF induce tumor formation from premalignant cells cotransplanted into animals (6). This is partly because these factors induce global epigenetic changes within the receiving cells, particularly changes in the expression of genes involved in stemness and tumorigenesis that can drive epithelia to become tumor-initiating cells (92-94). The growth of tumor-initiating cells would also be assisted by MyoFbs metabolizing toxic metabolites produced by proliferating cells (35). Moreover, MyoFbs produce copious amounts of ECM proteins, including collagen I, fibronectin, fibrillin, elastin, proteoglycans, and tenascins (91), which serve not only as the building blocks for the stroma but also as ligands for cell surface receptors promoting cell proliferation and other tumor-initiating events (95). In addition, MyoFbs produce ECM–cross-linking enzymes, such as lysyl oxidase (LOX), LOX-like (LOXL), and transglutaminase (TG) as well as proteases that remodel the ECM, such as MMPs (8, 34, 62, 96, 97). These ECM components and ECM-modifying enzymes together can increase stromal rigidity, which mechanically induces tumor initiation and progression. Stiff ECM with high collagen density alone is sufficient to induce the malignant transformation of cultured normal breast cells (98, 99), demonstrating that the mechanical properties of the fibrotic stroma play a key role in tumor initiation. MMPs produced by MyoFbs could also increase ROS production in the tissue, promoting genomic instability that facilitates tumor initiation (49). Furthermore, the MyoFb-rich stroma compromises antitumor immunity, while also assisting in the development and survival of tumor-initiating cells (100, 101).
PHENOTYPIC INFLUENCE BY THE ECM
The ECM is a network of fibrous proteins embedded in a gel-like substrate made of proteoglycans. The core matrisome of the mammalian ECM includes ~300 proteins secreted from different resident cells that biochemically and mechanically regulate cell and tissue functions (102, 103). It not only supports tissues structurally but also provides or sequesters various signals to dynamically regulate diverse cellular functions, including growth, survival, morphogenesis, and differentiation (104). The ECM is constantly degraded and remodeled in response to growth, disease, and injury (105-107), diversifying its components and functionalities across tissues. Such variations are derived from differences in protein isoforms or homologs, post-translational modifications, ratios of individual components, and turnover rates, resulting in tissue-specific mechanical and biochemical properties (105, 108). As discussed above, MyoFbs have a profound effect on ECM structure and composition, although other cell types, such as adipocytes, macrophages, MSCs, and endothelial cells, also contribute to the ECM (109-113).
Properties of the ECM
The ECM is composed of several distinct families of evolutionarily conserved molecules. These include glycoproteins (such as collagen, laminin, and elastin), which form fibrous macromolecules and interact with cell surface receptors, and proteoglycans (such as heparin sulfate), which are attached to anionic glycosaminoglycans, exert hydrodynamic functions, and form linkages to macromolecules (114). The basement membrane (BM) component of the ECM surrounds the parenchyma and demarcates it from the stroma, whereas the IM constitutes the bulk of the ECM in the body (Fig. 2) (115).
Fig. 2. Properties of the ECM.
The ECM, here illustrated using breast tissue as an example, consists of the interstitial matrix (IM) and the basement membrane (BM). The IM, which constitutes the bulk of the ECM, is produced by resident stromal cells, serves as a structural scaffold, controls the availability of growth factors and other signaling molecules, and imparts mechanical stimuli to cells. The other component of the ECM, the BM, is produced by basal epithelial cells and is more compact and less porous than the IM. In many disease conditions, the IM becomes fibrous and stiff, and the BM is remodeled or compromised.
BM is more compact and less porous than the IM, containing type IV, XV, XVII, and XVIII collagens; laminins; entactin; and proteoglycans (105). BM is synthesized and organized by basal epithelial cells (116); in the mammary gland, it is primarily produced by myoepithelial cells, the outer cells of the bilayered epithelium (117). The deposition of BM proteins, especially laminins, is facilitated by Rho-associated kinase 1 (ROCK-1), which promotes the localization of the polarity protein partitioning-defective 1b (PAR-1b) to the basal surface of the epithelium (118). Deposited laminins bind to cell surface receptors (such as integrins), initiating the self-assembly of polymeric networks and cross-linking of other BM proteins (such as type IV collagen and nidogen) to establish the BM (119). The BM directly contacts the parenchymal cell surface, providing protection against mechanical and chemical stresses, mediating interactions between cells and the IM, and acting as a barrier between tissues. The BM also plays key roles in tissue differentiation, polarity, and homeostasis (120). Some BM components, such as laminins and type IV collagens, serve as reservoirs for endothelium-derived antiangiogenic factors (endostatin, arrestin, canstatin, and tumstatin), contributing to its tumor-suppressive roles (121). During embryogenesis, BMs are reprogrammed by spatiotemporally regulated MMPs to generate anisotropic patterns that mechanically influence tissue and organ morphogenesis (122). In tumors, vascular and epithelial BMs are actively remodeled during angiogenesis and cell invasion, respectively (121). BMs can also be mechanically ruptured by the protrusive force of dividing cells or the compressive force of an expanding epithelial mass, contributing to tumor cell invasion (123).
The IM constitutes a large part of the stroma, containing type I, III, V, VI, VII, and XII collagens as well as fibronectin, elastin, tenascin, glucosaminoglycans, mucopolysaccharides, and proteoglycans. IM proteins are synthesized by the resident stromal cells, in particular, fibroblasts. Fibrillar collagens (type I, II, III, V, and XI) are the most abundant proteins in the IM (124). Newly translated collagen forms triple-helical procollagen in the Golgi apparatus before it is secreted into the extracellular space, where the N and C termini are cleaved by MMPs to release the mature collagen that is cross-linked by LOX to stabilize the fibrillar structure (125). The IM plays roles in tissue hydration, growth factor binding, structurally supporting tissues, and linking neighboring tissues (115). IMs are bathed in interstitial fluid, which transports nutrients, oxygen, and metabolites between vessels and cells. Unlike the BM’s role as a selective tissue barrier, the IM is the site of a tissue’s first response to external cues. For example, TGF-β forms a latent complex that is sequestered in the IM until biochemical or mechanical stimuli trigger its release from the IM, thus initiating fibrogenic signaling (126). In addition, the IM itself mechanically stimulates cells. Increased collagen deposition and cross-linking increase the stiffness of the IM, which induces cell motility and proliferation (105). During tissue and organ morphogenesis, local increases in collagen deposition create mechanical anisotropies that drive cell migration in a particular direction (115).
Mechanical continuum from the ECM to the nucleus
Cells sense the biochemical and mechanical properties of the ECM and, in turn, regulate the expression of genes encoding ECM proteins and ECM-modifying enzymes (106, 107). Such dynamic and reciprocal ECM-nucleus interactions were initially proposed more than 40 years ago (127, 128) and have since been corroborated by the discovery that the ECM-nucleus linkage is not only biochemical but also mechanical through physical continuums that hardwire the ECM to the nucleus (Fig. 3) (129, 130). The viscous and elastic properties of the ECM, defined by Young’s elastic modulus, depend on the composition and organization of its constituents (131). Mechanotransduction transforms these physical properties into biochemical signals that influence cellular functions and tissue specificity. Increased ECM stiffness, in particular, is closely associated with pathological conditions (115).
Fig. 3. Physical continuum between the ECM and chromatin.
The signals received at cell-cell junctions (tight junction, adherens complexes, and desmosomes) and at cell-ECM junctions [focal adhesions (FAs) and hemidesmosomes] are transmitted to the nucleus through microfilaments, microtubules, and intermediate filaments. At the nucleus, these elements of the cytoskeleton are connected to the LINC complex (KASH and SUN) through nesprin. The LINC complex spans the outer (ONM) and inner (INM) nuclear membranes to connect the cytoskeleton to lamin in the nuclear lamina, which associates with chromosome in the nucleoplasm.
Integrins, heterodimeric transmembrane receptors composed of α and β subunits, are the most well-studied cell surface receptors transmitting mechanical ECM signals into cells. The extracellular domain of the α subunit determines ligand specificity, and the cytosolic tail of the β subunit affects a multitude of downstream signaling pathways through its connection to the cytoskeleton (132). Whereas integrins αvβ3 and α3β1 support the formation of FA complexes at the cell-IM interface, integrin α6β4 supports the formation of hemidesmosomes (HDs) at the cell-BM interface (133-135).
FAs are multiprotein complexes involved in cellular contractility and invasiveness. They form when cells contact a stiff substrate and are the primary sites to receive and transmit mechanical cues from the ECM. During cell migration, FAs activate Rho, Rac, and Cdc42 guanosine triphosphatases (GTPases) to trigger cytoskeletal reorganization, generating tension within a cell to form filopodia (134, 136). FA formation is triggered by clustering integrin heterodimers that connect their ECM ligands to the actin cytoskeleton, enabling the bidirectional signaling and force transmission (134, 137). Formation and maturation of FAs involve “inside-out” integrin signaling. At the nascent adhesions, integrin heterodimers are in an inactive state with low binding affinity. Integrin-ECM interaction then leads to the generation of phosphatidylinositol-4,5-bisphosphate [PtdIns (4, 5) P2] that anchors FA kinase (FAK) to the nascent adhesion. FAK, in turn, recruits talin, an adaptor protein linked to actin cytoskeletons. Talin then binds to the cytosolic domain of the integrin β subunit, converting the receptor dimers to an active, high-affinity state. Activated integrins form clusters, further promoting ligand affinity and accumulation to establish mature FAs. Integrin dimers at this stage can receive and transmit extracellular stimuli through “outside-in” signaling (137-140). In stiff ECM, the mechanical tension reinforces FAs by promoting their size and maturation rate to produce respective cytoskeletal activities for force transmission (141-143).
HDs, the other type of integrin-mediated adhesions between epithelial cells and the ECM, link the ECM to the cytokeratin network and are essential for tissue differentiation and polarity. Type I HDs are found in basal stratified epithelia, such as skin, and are composed of integrin α6β4, CD151 (tetraspanin), plectin, BP180, and BP230. The integrin ligand in this case is laminin-332, and plectin and BP230 bind cytokeratin 5 (CK-5) and CK-14. Type II HDs are found in simple epithelia, such as the intestine, and are composed of integrin α6β4 and plectin only. In this case, the integrin ligand is also laminin-332, but plectin binds CK-8 and CK-18 (144). HDs provide epithelial tissues with structural stability and integrity against mechanical stress while also playing a role as mechanosensors (145). Mechanical tension generated during tissue morphogenesis activates the kinase PAK-1, which phosphorylates cytokeratins to induce maturation of HDs to resist mechanical stress (135). The formation of stable HDs also depends on the proteolytic processing of laminin-332 (146). Whereas laminin-332 is processed at the α3 chain and binds integrin α6β4, leading to the formation of stable HDs in normal cells, laminin-332 is often unprocessed at the α3 chain in tumors and instead binds integrin α3β1, which is normally involved in FA formation (147). HDs are rapidly disassembled and reassembled during migration and invasion (148).
FAs and HDs link the ECM to the cytoskeletal network of structural and regulatory proteins that can both generate and transmit forces to control cellular movement and shape, as well as nuclear morphology and integrity (149, 150). The three structural components of the cytoskeleton—microfilaments (MFs), microtubules (MTs), and intermediate filaments (IFs)—regulate one other (151). For example, at lamellipodia, the growth of MTs activates the GTPase Rac to promote polymerization of the actin molecules that constitute MFs (152). In response to external cues, these polymers are rapidly disassembled and reassembled into different cytoskeletal structures (149, 151). Such pliability makes them highly dynamic, enabling changes in cell shape, mobility, and rigidity (151).
The assembly and disassembly of the actin filaments that constitute MFs depend on the Rho family GTPases that regulate the balance between monomeric actin (G-actin) and filamentous actin (F-actin) (153). These dynamic filaments drive cell movements and form cross-linked networks that establish cell shape and rigidity (151). During cell migration, the MF network extends toward the leading edge of the cell, forming membrane protrusions that attach to the substrate through FAs. This triggers myosin-mediated contraction of the actin filaments, generating traction forces that push the cell forward (154). MFs are also critical for mechanosensing and mechanotransduction because tensional stress causes MFs to change their conformations and interactions with regulatory proteins. In response to stiff ECM, Rho-dependent actin polymerization increases, which, in turn, stiffens the cell interior (155). MFs contribute to the membrane skeleton, the cortical actin rim, and stress fibers. MFs of the membrane skeleton, which lies beneath the plasma membrane and controls membrane architecture and cell shape, are cross-linked by spectrin and associate with several proteins (ankyrin, protein 4.1, adducin, and α-catenin) that link them to transmembrane proteins (156). In the cortical actin rim, which is associated with cell-cell and cell-ECM adhesion complexes that strengthen cell adhesions and generate the outward tension that controls cell shape, MFs constitute the cortical bundles that establish adhesion belts linked to adherens junctions and tight junctions. The cortical actin rim is organized by various actin-binding proteins (spectrin, α-actinin, α-catenin, vinculin, filamin, cortactin, Wiskott–Aldrich syndrome protein (WASP), vasodilator-stimulated phosphoprotein (VASP), gelsolin, cofilin, ezrin-radixin-moesin, and heat shock protein 27) (156). Stress fibers, which are linked to FAs, adherens junctions, and the nuclear membrane, are actomyosin bundles in which short F-actins with alternating polarity are cross-linked by α-actinin and bound by vinculin, talin, and filamin. Stress fibers generate inward tension to induce cell contraction and to counteract outward tension generated by the cortical actin rim (156, 157).
MTs are rigid, polymeric hollow tubes that are composed of dimers of α- and β-tubulin and serve as tracks for moving cargoes and as scaffolds for intracellular trafficking, mitosis, and organizing organelles (151). The slow-growing minus ends of MTs are anchored to MT organizing centers (MTOCs, the centrosomes), whereas the fast-growing plus ends extend toward the cell surface (158). The layout of MTs in a cell depends on the context. For example, in differentiating epithelia, centrosomes are localized subapically, orienting the MT minus ends toward the apical surface with the plus ends toward the basal surface, generating apicobasal polarity (159). In migrating fibroblasts, centrosomes are localized between the nucleus and the leading edge, with the plus ends extending toward the leading edge (160). MTs also play roles in mechanosensing and mechanotransduction. Tension-induced deformation of MTs activates Src signaling to restrict MT growth at the plus ends, which helps control organ size and shape (161).
IFs are the major structural components of both the cytosol and nucleus, comprising at least 65 distinct proteins. IFs help establish structural integrity and absorb mechanical stress. Unlike MFs and MTs, which are dynamic polymers of globular proteins, IFs are stable, insoluble filaments composed of long continuous segments of α helices. The assembly of IFs is regulated developmentally and in cell- and organelle-specific manners, as seen for cytokeratins (epithelia), desmin (muscle), vimentin (mesenchymal cells), glial fibrillary acidic protein (GFRAP, glia cells), neurofilaments (neurons), and lamins (nuclear envelope) (162). Cytosolic IFs help stabilize cell shape by contributing to various adhesion complexes (163). In cell-cell adhesions (desmosomes), IFs are anchored to adhesion plaques (desmoplakin) linked to adhesion proteins (desmoglein and desmocollin), connecting the plasma membranes of two adjacent cells through linker proteins (plakoglobin and plakophilin) (164). In cell-ECM adhesions (HDs), IFs are linked to the ECM laminin receptor integrin α6β4 through plectin and BP230 (144).
Nuclear IFs are solely assembled by lamins and constitute the nuclear lamina, a fibrillar network associated with the inner nuclear membrane. The nuclear lamina provides a platform for chromatin organization and gene transcription (165) and connects the nucleus to the cytoskeleton, which provides both structural support for the nucleus and a mechanical continuum transmitting external force from ECM to nucleus (Fig. 3) (165, 166). The linker of nucleoskeleton and cytoskeleton (LINC) complex mechanically connects the nuclear lamina to the cytoskeleton and is composed of the Sad1 and UNC-84 domain proteins SUN1 and SUN2, which are linked to lamins and span the inner nuclear membrane, and the Klarsicht, nuclear anchorage protein 1 (ANC-1), and Syne homology domain (KASH) proteins, which are linked to SUN and span the outer nuclear membrane. Nesprins embedded in the outer nuclear membrane connect LINC to the cytoskeleton by interacting with both KASH and cytoskeletal proteins. Nesprin-1 and nesprin-2 link to MFs, nesprin-3 links to IFs, and nesprin-4 links to MTs (166, 167). In addition to being important for DNA damage repair and chromosome anchoring during meiosis, LINC is also required for maintaining nuclear shape, cell polarization, and migration under mechanical stimuli (167). SUN mediates the connection between LINC and the nuclear lamina by interacting with lamin A. Whereas the amounts of B-type lamins, which assemble into randomly organized filaments and confer elasticity and deformability to the nucleus (167-169), are fairly constant, the amounts of A-type lamins (lamin A and lamin C) vary and positively correlate with tissue stiffness because they increase the rigidity of the nuclear envelope (169). Because of its interaction with LINC, lamin A serves as a molecular shock absorber that protects nuclear architecture from mechanical stress (167). In response to tension, lamin A amounts increase and LINC is stabilized through additional recruitment of the complex proteins and actin filaments, imparting stiffness to the nucleus to resist the force (167, 168). Because lamin A binds to chromosomes, it also links mechanical tension in the cytoskeleton to gene regulation (167).
Nuclear responses to mechanical stress
Although the nucleus is generally 2 to 10 times stiffer than the cytosol, it is elastic and deformable (170). As discussed above, A-type lamins impart stiffness to the nuclear envelope, whereas B-type lamins confer the elasticity and deformability that allow for cell survival, migration, and differentiation (167-169). Chromosomes in the nucleoplasm also contribute to the viscoelasticity of the nucleus: Chromosome condensation during cellular differentiation stiffens the nucleus, whereas chromosome decondensation during mitosis softens the nucleus (170). Thus, the mechanical properties of the nucleus are determined by the elastic, deformable nuclear lamina and the viscoelastic nucleoplasm (170).
Cellular and nuclear deformation by external stresses can induce either local chromatin stretching to absorb the stress or global chromatin decondensation to dissipate the stress. This is mediated by a shift in A-type lamin interactions from highly compact (H3K9me3-marked) to less-compact (H3K27me3-marked) heterochromatin, which activates the mechanosensitive transcriptional regulators megakaryoblastic leukemia 1 (MAL)/serum response factor (SRF) complex, YAP, and TAZ (171). For long-term effects, however, the nucleus eventually increases its deformability, for example, by increasing the lamin B–to–lamin A ratio (171). When a cell is extremely deformed, such as during migration through densely packed ECM fibers, the nucleus becomes extremely compressed, leading to nuclear membrane rupture that causes DNA damage (170, 172).
Pathogenic effects of stiff ECM
There are three major causes of the ECM becoming fibrous and stiff in diseases such as fibrosis and cancer (Fig. 4, A to C). The first is dysregulated matrix biosynthesis and remodeling by MyoFbs. As discussed above, MyoFbs produce copious amounts of fibrogenic factors, ECM proteins, and ECM-modifying enzymes that are required for tissue repair, but excessive amounts of these products increase the rigidity and stability of the ECM (Fig. 4A) (8, 96). The second major cause of ECM stiffening is the increased contractile forces generated by MyoFb α-SMA stress fibers. Stress fibers within cells transmit contractile forces to the ECM through their connections to the integrins in FAs (αvβ5 and αvβ3). This induces the remodeling and stabilization of the local ECM to increase the stiffness (Fig. 4B) (63). MyoFb contraction further stimulates fibrogenic signaling through positive feedback mechanisms, such as liberation of TGF-β from the ECM-bound latent complex (173). Moreover, the increased ECM stiffness increases the traction forces of MyoFbs against the ECM, also triggering the release of TGF-β from the ECM (174). The third major cause of ECM stiffening is advanced glycation end products (AGEs), strong oxidizing agents that induce nonenzymatic collagen cross-linking (175). AGEs are produced by the conjugation of sugars to various biomolecules, such as proteins and nucleic acids, and their amounts increase during aging and in chronic diseases such as diabetes and cardiovascular disease (176). AGEs also induce MyoFb formation from NFs (differentiation) and epithelia (type 2 EMT) (Fig. 4C) (177). In fact, increased MyoFb formation by AGEs partly accounts for higher cancer risks in older people and individuals with chronic disorders (178).
Fig. 4. Three major mechanisms of ECM stiffening.
(A) Cancer cells and stromal MyoFbs secrete large amounts of fibrogenic factors (such as FGF and TGF-β), fibrillar collagen, and ECM cross-linking (LOX) and remodeling (MMP) enzymes. (B) Lockstep model of ECM stiffening by MyoFb contraction. ECM fibers are connected to MyoFb stress fibers through FAs. The contraction of MyoFbs loosens adjacent ECM fibers, which activates local ECM remodeling and stabilizing factors to increase local ECM rigidity. As MyoFbs retract, they stretch the ECM, increasing ECM tension. Repetition of the cycle further increases ECM rigidity. (C) ECM stiffening is triggered by advanced glycation end products (AGEs). AGEs nonenzymatically cross-link collagen, which stiffens the ECM, and stimulate the receptor RAGE, which increases the MyoFb population by both activating NFs and promoting type 2 EMT in epithelia.
Excessive synthesis or cross-linking of collagen I stiffens the ECM and underlies the pathogenesis of many diseases, including cancer, fibrosis, and chronic disorders. Stiff ECM exerts tensional stress that disrupts tissue structures and promotes transformation, metastasis, and cell contractility through various signaling pathways, including integrin β1, FAK, extracellular signal–regulated kinases (ERKs), and RhoA (105). Enzymatic cross-linking of collagen I is catalyzed by enzymes, like LOX and TG, that are increased in metastatic tumors (108, 179). Conversely, nonenzymatic cross-linking of ECM proteins occurs through glycation (180). AGEs not only stiffen the ECM but also activate their cognate receptor, RAGE, which promotes cancer progression by triggering proliferative signaling and inhibiting cell death pathways (181, 182). Furthermore, the mechanical tension of stiff ECM itself plays critical roles in cancer initiation by inducing the transformation of normal cells (183, 184). Stiff ECM also promotes the Warburg effect, a shift to aerobic glycolytic metabolism (98, 185, 186), and nuclear translocation of YAP and TAZ, which stimulate the expression of growth-associated genes (168, 187). The tensional force of stiff ECM that is ultimately transmitted to the nucleus through LINC (129, 130, 168) also leads to nuclear stiffening and genomic instability (150, 168).
In addition to highly cross-linked ECM fibers, tumor stroma also contain abnormally high amounts of interstitial fluid due to leaky vasculature (188) and high amounts of the glycosaminoglycan HA (189), both of which contribute to increased interstitial pressure. The resulting pressure gradient between tumors and normal tissues creates fluid flow, activating integrin signals that promote tumor cell invasion (188).
Stiff ECM drives hallmarks of cancer
Experimental evidence attests to the notion that normal or nonmalignant cells can acquire a malignant phenotype after encountering stiff ECM. Culturing normal mammary epithelia in high-density collagen matrix alone helps them acquire a proliferative and invasive phenotype (98, 99), and crossing a mouse strain that develops spontaneous mammary tumors with a strain overexpressing collagen I greatly exacerbates tumor development (10). These effects are largely due to the increased mechanical—not biochemical—stimuli of stiff ECM, as demonstrated by in vitro studies using inert synthetic matrices (such as polyacrylamide hydrogel) instead of bioactive collagen (98, 184). To understand how the mechanical stimulus of stiff ECM contributes to tumor initiation, several studies describe its roles in driving different hallmarks of cancer (95, 190), the processes necessary for the transformation of normal human cells to malignant tumors (95, 190). Here, we summarize some of the insights into how cancer hallmarks are ascribed to stiff ECM (Fig. 5, A and B).
Fig. 5. Mechanisms of stiff ECM–induced tumor initiation.
(A) Fibrogenic signals increase the numbers of MyoFb-like cells and cause ECM stiffening, which eventually leads to malignant transformation of the parenchyma characterized by sustained proliferation, evasion of growth suppression, resistance to cell death, and replicative immortality. (B) The tension of the stiff ECM is transmitted to the nuclear membrane of parenchymal cells. This may deform or even rupture the nuclear membrane, leading to genome instability through chromosome segregation errors, DNA damage, defective damage repair, and/or reactivation of transposons.
Sustained proliferative signaling and the evasion of growth suppression are the major consequences of stiff ECM. Normal cells embedded in compliant matrices differentiate and maintain homeostatic tissue architecture and functions, with their proliferation controlled by mitogenic signals from ligand-activated growth factor receptors that promote cell cycle progression and contact inhibition mediated by the cell-cell junction proteins E-cadherin and zonula occludens-1 (ZO-1), which induces G1 arrest at high cell density. In cancer cells or normal cells surrounded by stiff ECM, various mechanisms can block negative feedback of growth factor signaling, cause mitogenic signaling to become autonomous, and/or override or bypass contact inhibition. The internalization of cell surface receptors through clathrin-mediated endocytosis is a negative feedback mechanism that limits growth factor signaling (191). However, stiff ECM increases the affinity of integrin αvβ5 for its ECM ligand, forming long-lived plaques on the cell surface that are resistant to endocytosis (192). Stiff ECM also activates FAK-Rac signaling, which promotes mitogen-independent cell cycle progression by transcriptionally inducing cyclin D1 (193), and induces translocation of the mechanosensitive transcription regulator YAP into the nucleus, where it activates the expression of genes encoding cell cycle promoters, such as E2F1 (194). Furthermore, stiff ECM is also associated with the mislocalization of E-cadherin and ZO-1 and greatly lowers the threshold of EGF necessary to counteract contact inhibition (195). Induction of the DDR upon genotoxic stress is another major growth suppressor that is inhibited by stiff ECM. Under normal conditions, DNA damage induces cell cycle arrest through tumor suppressors such as Rb, p53, breast cancer 1 (BRCA1), and phosphatase and tensin homolog (PTEN), which enables to DNA repair; however, the expression and function of these proteins are reduced by stiff ECM, thus impairing cell cycle arrest upon DNA damage (193, 196-198).
Resistance to apoptosis is another effect of stiff ECM that is closely associated with tumor initiation. The survival of normal adherent cells requires their attachment to the ECM through integrins, and detachment induces a form of caspase-mediated apoptosis called anoikis. Anoikis is essential for the formation and maintenance of normal tissues, such as lumen formation in alveoli, and protects against dysplasia by preventing anchorage-dependent cells from growing outside their normal setting (199). Cancer cells acquire resistance to anoikis through various mechanisms, allowing them to metastasize to different environments. One way is to induce an “integrin switch” that modulates integrin profiles and therefore sensitivity to ECM detachment (200). The second way is to constitutively activate downstream mediators of integrin signaling, such as phosphoinositide 3-kinase (PI3K) and AKT (201). The third way is to induce EMT, which, in turn, increases the production of antiapoptotic (Bcl-2 family) and prosurvival proteins (PI3K and Akt) (201). A fourth way is to directly increase the production of antiapoptotic proteins, such as survivin (202). All these mechanisms are induced in normal cells encountering stiff ECM (95). Furthermore, resistance to apoptosis owing to stiff ECM is a major cause for reduced efficacy of antitumor therapies. For example, stiff ECM inhibits doxorubicin-induced, p53-mediated apoptosis in breast tumors (203).
Replicative immortality is another consequence of stiff ECM that may contribute to tumorigenesis. Immortality is largely attributed to the activation of telomere maintenance mechanisms (204). Telomeres, repetitive DNA sequences at the ends of chromosomes, progressively shorten as normal cells divide because of incomplete DNA synthesis or damage, leading to replicative senescence and aging. However, a rare cell that constitutively produces telomerase, the enzyme that repairs the shortened telomere, can divide indefinitely (205). Telomerase promotes EMT, is essential for stem cell self-renewal and pluripotency (206), and is closely associated with stiff ECM (190). For example, telomerase is increased in idiopathic pulmonary fibrosis, which is characterized by stiff ECM, abundant MyoFbs, and hypertrophy and is directly associated with increased risk of developing lung cancer (207).
Last, genome instability and mutations, early events in tumorigenesis, can also result from stiff ECM. In fact, several critical mutations found in transformed cells are already present in nonmalignant cells from benign fibrotic breast tissue biopsied from women who later developed breast cancer (208). Genome instability arises from many different causes, including chromosome segregation errors, DNA damage, defective DNA damage repair, and increased activity of transposable elements. Faithful chromosome segregation is essential for genome stability and survival of the organism. However, the fidelity of chromosome segregation depends on proper tissue architecture, which can be disturbed by stiff ECM (184, 209). Cells confined between the rigid ECM fibers can undergo asymmetric multidaughter mitosis that generates aneuploid cells (210). Migration of cells through densely packed, stiff ECM also compresses the nuclei, which can result in nuclear deformation or nuclear membrane rupture that increases replication stress and DNA damage, thereby compromising genomic integrity (170, 172). Compression of the nucleus by stiff ECM can also inhibit major DNA repair proteins, such as BRCA1 and p53BP1, from entering the nucleus (211). Thus, the rate of incurred mutations is predicted to be directly proportional to the frequency and magnitude of mechanical stress exerted on the nuclei (212). Cumulative stresses caused by the chronic disruption of nuclear architecture by mechanical tension from stiff ECM could alter chromatin structure and function, potentially leading to the dysregulation of oncogenes or tumor suppressors or to the reactivation of transposable elements that are otherwise repressed, which would promote mutagenesis and genomic instability (170).
FIBROBLASTS IN RESISTANCE TO CANCER THERAPIES
Mechanisms of CAF-induced therapeutic resistance
In addition to promoting tumor initiation and progression, MyoFbs and other CAFs help induce therapeutic resistance (42). Chemotherapy itself can also stimulate CAF differentiation (213). There are several major mechanisms through which CAFs promote drug resistance. First, CAF-derived paracrine factors can not only induce epigenetic changes in stemness genes but also trigger the expression of stem factors (such as Wnt and HGF) within epithelia, driving them to become tumor-initiating cells or cancer stem cells (92-94, 214), which are inherently resistant to chemotherapeutic agents due to having an EMT phenotype and the ability to inactivate drugs (215). Second, increases in mechanical stress within a tumor, driven by CAF-derived ECM proteins and ECM-modifying enzymes, could collapse blood vessels, which not only impairs drug delivery but also causes hypoxia and subsequent acidosis that reduces the efficacy of anticancer drugs (216, 217).
A third mechanism is through the activation of survival pathways by nuclear factor κB (NF-κB) signaling. Most anticancer drugs activate NF-κB signaling in CAFs, which, in turn, increases the release of paracrine factors (such as WNT16B, secreted frizzled related protein 2 (SFRP2), IL-6, IL-17A, and HGF) that induce survival signaling in cancer cells through ERK1/2 and PI3K–ATK–NF-κB pathways (218, 219). This drives the expression of antiapoptotic proteins, such as B-cell lymphoma-2 (BCL-2), and the efflux pump multidrug-resistant protein 1 (220). In aggressive cancers, NF-κB is constitutively active in both CAFs and tumor cells (221). Fourth, CAF-secreted MMPs promote a proinflammatory signature (COX-2, NF-κB, and HIF-1 expression) in cancer cells, leading to the production of ROS, which induces EMT that, in turn, promotes invasiveness and stemness and lowers the efficacy of certain anticancer drugs (222-224). Fifth, under drug treatment, both CAFs and tumor cells undergo a metabolic switch to glycolysis. This induces the expression of antiapoptotic proteins and drug efflux pumps, EMT, autophagy, and acidification, leading to multidrug resistance (225, 226). Furthermore, a glycolytic metabolite, lactate, serves as a signaling molecule instructing CAFs to secrete HGF, which also induces drug resistance (225, 227).
Last, CAFs suppress antitumor immunity and compromise immunotherapy through multiple mechanisms. A subset of CAFs (FAP+ CAFs) facilitate the formation of protumor myeloid cells, including M2-type macrophages and MDSCs that inhibit CD8+ cytotoxic T cells (228, 229). Another subpopulation of CAFs (CAF-S1 cells; see Table 2) promotes the formation of regulatory T cells that inhibit both CD4+ helper and CD8+ cytotoxic T cells (230). CAFs also produce paracrine factors that inhibit natural killer cells [prostaglandin E2 (PGE2) and indoleamine 2,3-dioxygenase (IDO)] and dendritic cells (IL-6), as well as immune checkpoint proteins [programmed death-ligand 1 (PDL-1) and PDL-2] that inhibit T cell activation (229).
Epithelial-mesenchymal plasticity of cancer cells
In addition to assisting cancer cells in acquiring resistance to chemo- and immunotherapies, CAFs may also play direct roles in the resistance. Although CAFs have long been considered as mere assistants of cancer cells, CAFs could in fact be cancer cells that have camouflaged to evade anticancer immunity and treatments. Cancer cells often undergo EMT to acquire therapy resistance (222-224, 231) but may revert to epithelia through MET (mesenchymal-epithelial transition) after the drug is withdrawn (232, 233). These transitions can cause the cancer cells to adopt mixed epithelial-mesenchymal features, a phenomenon termed “epithelial-mesenchymal plasticity,” which is associated with enhanced aggressiveness and therapeutic resistance (234). One such clinical manifestation is metaplastic breast cancer (MpBC), a rare, aggressive cancer accounting for less than 1% of the total breast cancer cases. MpBC carcinomas are composed of multiple cell populations that have undergone metaplastic differentiation into nonglandular cells, such as spindle-shaped cells. These cells produce high amounts of EMT markers and exhibit stem cell (tumor-initiating cell)–like features, including drug resistance (235). MpBC, being positive for the epithelial marker cytokeratin, is thought to arise from the epithelial compartment through metaplasia or EMT as cells adapt to tissue-damaging stimuli (235-237). Thus, such transformation could be triggered by drug treatment for a less aggressive epithelial cancer. It is plausible that this type of aggravated cancer progression and the resulting drug resistance would be blocked by a combinatory therapy of an anticancer drug and an EMT inhibitor or a drug targeted to tumorigenic stroma (238-240).
CONCLUSIONS
Dynamic reciprocity between the ECM and nuclear matrix was initially proposed by M. Bissell and P. Bornstein 40 years ago. In this model, the ECM exerts mechanical and biochemical influences on the nucleus through cell surface receptors, leading to changes in gene expression that, in turn, affect the ECM, resulting in a cycle of dynamic cell-ECM interactions (127, 128). Their provocative hypothesis was finally validated 15 years later by the pioneering studies of D. Ingber, substantiating the physical continuum that hardwires the ECM to the nucleus (129, 130). These decades of ground-breaking studies provoked a paradigm shift in the fields of life sciences and medicine, eventually giving forth the previously unknown concepts of tissue microenvironment and mechanobiology playing pivotal roles in regulating tissue and organ homeostasis and disease pathogenesis (241).
These concepts have reshaped cancer research and treatment based on the well-established fact that most tumors harbor stiff, desmoplastic stroma. The fibrous tumor stroma was initially thought to be the mere consequence of malignant progression; however, cumulative evidence has shown that stiff ECM also drives tumor initiation. This knowledge has pushed forward the development of new cancer therapeutics targeting the stiff stroma. Most of these approaches aim to ameliorate the stiffness of tumor stroma with inhibitors of fibrogenic factors (such as TGF-β), ECM cross-linking enzymes (such as LOX and TG), ECM remodeling enzymes (MMPs), or collagen biosynthesis enzymes (such as prolyl hydroxylase) (242). Another method uses an N-terminal α-SMA peptide that inhibits MyoFb contraction or inhibitors of molecules involved in signal transduction induced by stiff ECM (such as integrins, FAK, RhoA-ROCK, and YAP/TAZ) (242). There also have been new technologies that deliver or activate chemotherapeutics specifically in stiff ECM (243, 244). These new therapeutics, however, have, so far, only been used for cancer treatment not for prevention. We speculate that combining a mechanoresponsive delivery or activation system with a mitigator of stromal stiffness could possibly be applied to preventing cancers associated with tissue fibrosis or chronic wounds. Such an exciting possibility warrants further investigation.
Acknowledgments:
We thank all the members in Furuta laboratory and the Department of Cancer Biology at University of Toledo College Medicine and Life Sciences.
Funding:
This work was supported by the start-up fund from University of Toledo Health Science Campus, College of Medicine and Life Sciences, Department of Cancer Biology to S.F.; Ohio Cancer Research Grant (project no. 5017) to S.F.; Medical Research Society (Toledo Foundation) Award to S.F.; American Cancer Society Research Scholar Grant (RSG-18-238-01-CSM) to S.F.; and National Cancer Institute Research Grant (R01CA248304) to S.F.
Footnotes
Competing interests: The authors declare that they have no competing interests.
REFERENCES AND NOTES
- 1.Shao Z-M, Nguyen M, Barsky SH, Human breast carcinoma desmoplasia is PDGF initiated. Oncogene 19, 4337–4345 (2000). [DOI] [PubMed] [Google Scholar]
- 2.DeClerck YA, Desmoplasia: A response or a niche? Cancer Discov. 2, 772–774 (2012). [DOI] [PubMed] [Google Scholar]
- 3.Boyd NF, Martin LJ, Yaffe MJ, Minkin S, Mammographic density and breast cancer risk: Current understanding and future prospects. Breast Cancer Res. 13, 223 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sappino AP, Skalli O, Jackson B, Schürch W, Gabbiani G, Smooth-muscle differentiation in stromal cells of malignant and non-malignant breast tissues. Int. J. Cancer 41, 707–712 (1988). [DOI] [PubMed] [Google Scholar]
- 5.Huliák I, Bodai L, Czepán M, Kovács D, Szabó A, Tiszlavicz L, Lázár G, Rakonczay Z Jr., Hegyi P, Boros IM, Kirics Mi, Genetic, epigenetic and transcriptional comparison of esophagus tumor-associated and adjacent normal myofibroblasts. Oncol. Rep 41, 839–852 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA, Reconstruction of functionally normal and malignant human breast tissues in mice. Proc. Natl. Acad. Sci. U.S.A 101, 4966–4971 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA, The evolving relationship of wound healing and tumor stroma. JCI insight 3, e99911 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Otranto M, Sarrazy V, Bonté F, Hinz B, Gabbiani G, Desmouliere A, The role of the myofibroblast in tumor stroma remodeling. Cell Adh. Migr 6, 203–219 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dvorak HF, Tumors: Wounds that do not heal. New England Journal of Medicine 315, 1650–1659 (1986). [DOI] [PubMed] [Google Scholar]
- 10.Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, White JG, Keely PJ, Collagen density promotes mammary tumor initiation and progression. BMC Med. 6, 11 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.McAnulty RJ, Fibroblasts and myofibroblasts: Their source, function and role in disease. Int. J. Biochem. Cell Biol 39, 666–671 (2007). [DOI] [PubMed] [Google Scholar]
- 12.Tracy LE, Minasian RA, Caterson EJ, Extracellular matrix and dermal fibroblast function in the healing wound. Adv Wound Care (New Rochelle) 5, 119–136 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Foote AG, Wang Z, Kendziorski C, Thibeault SL, Tissue specific human fibroblast differential expression based on RNAsequencing analysis. BMC Genomics 20, 308 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fries KM, Blieden T, Looney RJ, Sempowski GD, Silvera MR, Willis RA, Phipps RP, Evidence of fibroblast heterogeneity and the role of fibroblast subpopulations in fibrosis. Clin. Immunol. Immunopathol 72, 283–292 (1994). [DOI] [PubMed] [Google Scholar]
- 15.Sadlonova A, Novak Z, Johnson MR, Bowe DB, Gault SR, Page GP, Thottassery JV, Welch DR, Frost AR, Breast fibroblasts modulate epithelial cell proliferation in three-dimensional in vitro co-culture. Breast Cancer Res. 7, R46–R59 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dumont N, Liu B, Defilippis RA, Chang H, Rabban JT, Karnezis AN, Tjoe JA, Marx J, Parvin B, Tlsty TD, Breast fibroblasts modulate early dissemination, tumorigenesis, and metastasis through alteration of extracellular matrix characteristics. Neoplasia (New York, N.Y.) 15, 249–262 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, Washington MK, Neilson EG, Moses HL, TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 303, 848–851 (2004). [DOI] [PubMed] [Google Scholar]
- 18.Stoker MGP, Shearer M, O'Neil C, Growth inhibition of polyoma-transformed cells by contact with static normal fibroblasts. J. Cell Sci 1, 297–310 (1966). [DOI] [PubMed] [Google Scholar]
- 19.Costanza B, Umelo IA, Bellier J, Castronovo V, Turtoi A, Stromal modulators of TGF-β in cancer. J. Clin. Med 6, 7 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Degeorges A, Tatoud R, Fauvel-Lafeve F, Podgorniak MP, Millot G, de Cremoux P, Calvo F, Stromal cells from human benign prostate hyperplasia produce a growth-inhibitory factor for LNCaP prostate cancer cells, identified as interleukin-6. Int. J. Cancer 68, 207–214 (1996). [DOI] [PubMed] [Google Scholar]
- 21.Shimoda M, Principe S, Jackson HW, Luga V, Fang H, Molyneux SD, Shao YW, Aiken A, Waterhouse PD, Karamboulas C, Hess FM, Ohtsuka T, Okada Y, Ailles L, Ludwig A, Wrana JL, Kislinger T, Khokha R, Loss of the Timp gene family is sufficient for the acquisition of the CAF-like cell state. Nat. Cell Biol 16, 889–901 (2014). [DOI] [PubMed] [Google Scholar]
- 22.Alkasalias T, Alexeyenko A, Hennig K, Danielsson F, Lebbink RJ, Fielden M, Turunen SP, Lehti K, Kashuba V, Madapura HS, Bozoky B, Lundberg E, Balland M, Guvén H, Klein G, Gad AKB, Pavlova T, RhoA knockout fibroblasts lose tumor-inhibitory capacity in vitro and promote tumor growth in vivo. Proc. Natl. Acad. Sci. U.S.A 114, E1413–E1421 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Buckley CD, Filer A, in Kelley and Firestein’s Textbook of Rheumatology, S T, Ed. (Elsevier, 2017), vol. 44, pp. 231–249. [Google Scholar]
- 24.Kaukonen R, Mai A, Georgiadou M, Saari M, de Franceschi N, Betz T, Sihto H, Ventelä S, Elo L, Jokitalo E, Westermarck J, Kellokumpu-Lehtinen PL, Joensuu H, Grenman R, Ivaska J, Normal stroma suppresses cancer cell proliferation via mechanosensitive regulation of JMJD1a-mediated transcription. Nat. Commun 7, 12237–12237 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tian X, Azpurua J, Hine C, Vaidya A, Myakishev-Rempel M, Ablaeva J, Mao Z, Nevo E, Gorbunova V, Seluanov A, High-molecular-mass hyaluronan mediates the cancer resistance of the naked mole rat. Nature 499, 346–349 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kaur A, Ecker BL, Douglass SM, Kugel III CH, Webster MR, Almeida FV, Somasundaram R, Hayden J, Ban E, Ahmadzadeh H, Franco-Barraza J, Shah N,Mellis IA, Keeney F, Kossenkov A, Tang HY, Yin X, Liu Q, Xu X, Fane M, Brafford P, Herlyn M, Speicher DW, Wargo JA, Tetzlaff MT, Haydu LE, Raj A, Shenoy V, Cukierman E, Weeraratna AT, Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility. Cancer Discov. 9, 64–81 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Maris P, Blomme A, Palacios AP, Costanza B, Bellahcène A, Bianchi E, Gofflot S, Drion P, Trombino GE, di Valentin E, Cusumano PG, Maweja S, Jerusalem G, Delvenne P, Lifrange E, Castronovo V, Turtoi A, Asporin is a fibroblast-derived TGF-β1 inhibitor and a tumor suppressor associated with good prognosis in breast cancer. PLoS Med. 12, e1001871 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chang PH, Hwang-Verslues WW, Chang YC, Chen CC, Hsiao M, Jeng YM, Chang KJ, Lee EYHP, Shew JY, Lee WH, Activation of Robo1 signaling of breast cancer cells by Slit2 from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/β-catenin pathway. Cancer Res. 72, 4652–4661 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhao S-J, Shen YF, Li Q, He YJ, Zhang YK, Hu LP, Jiang YQ, Xu NW, Wang YJ, Li J, Wang YH, Liu F, Zhang R, Yin GY, Tang JH, Zhou D, Zhang ZG, SLIT2/ROBO1 axis contributes to the Warburg effect in osteosarcoma through activation of SRC/ERK/c-MYC/PFKFB2 pathway. Cell Death Dis. 9, 390 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen X, Song E, Turning foes to friends: Targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov 18, 99–115 (2019). [DOI] [PubMed] [Google Scholar]
- 31.De Wever O, Mareel M, Role of tissue stroma in cancer cell invasion. J. Pathol 200, 429–447 (2003). [DOI] [PubMed] [Google Scholar]
- 32.Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, Chio IIC, Hwang CI, Tiriac H, Baker LA, Engle DD, Feig C, Kultti A, Egeblad M, Fearon DT, Crawford JM, Clevers H, Park Y, Tuveson DA, Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med 214, 579–596 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hanley CJ, Waise S, Parker R, Lopez MA, Taylor J, Kimbley LM, West J, Ottensmeier CH, Rose-Zerilli MJJ, Thomas GJ, Spatially discrete signalling niches regulate fibroblast heterogeneity in human lung cancer. bioRxiv 2020.2006.2008.134270 [Preprint]. 8 June 2020. 10.1101/2020.06.08.134270. [DOI] [Google Scholar]
- 34.Buchsbaum RJ, Oh SY, Breast cancer-associated fibroblasts: Where we are and where we need to go. Cancers (Basel) 8, 19 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E, Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: A metabolic survival role for tumor-associated stroma. Cancer Res. 66, 632–637 (2006). [DOI] [PubMed] [Google Scholar]
- 36.Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, Sahai E, Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol 9, 1392–1400 (2007). [DOI] [PubMed] [Google Scholar]
- 37.De Palma M, Biziato D, Petrova TV, Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer 17, 457–474 (2017). [DOI] [PubMed] [Google Scholar]
- 38.Wang F-T, Sun W, Zhang J-T, Fan Y-Z, Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol. Lett 17, 3055–3065 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Flavell RA, Sanjabi S, Wrzesinski SH, Licona-Limón P, The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol 10, 554–567 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, Preall J, Tuveson DA, IL1-induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 9, 282–301 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bhattacharjee S, Hamberger F, Ravichandra A, Miller M, Nair A, Affo S, Filliol A, Chin LK, Savage TM, Yin D, Wirsik NM, Mehal A, Arpaia N, Seki E, Mack M, Zhu D, Sims PA, Kalluri R, Stanger BZ, Olive KP, Schmidt T, Wells RG, Mederacke I, Schwabe RF, Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J. Clin. Invest 131, e146987 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bu L, Baba H, Yasuda T, Uchihara T, Ishimoto T, Functional diversity of cancer-associated fibroblasts in modulating drug resistance. Cancer Sci. 111, 3468–3477 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kopantzev EP, Vayshlya NA, Kopantseva MR, Egorov VI, Pikunov M, Zinovyeva MV, Vinogradova TV, Zborovskaya IB, Sverdlov ED, Cellular and molecular phenotypes of proliferating stromal cells from human carcinomas. Br. J. Cancer 102, 1533–1540 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Baloglu H, Yigit N, Stromal cells do not share KRAS mutations that the epithelial component harbors in colorectal carcinomas. Cancer Lett. 308, 118–121 (2011). [DOI] [PubMed] [Google Scholar]
- 45.Zhou Y, Bian S, Zhou X, Cui Y, Wang W, Wen L, Guo L, Fu W, Tang F, Single-cell multiomics sequencing reveals prevalent genomic alterations in tumor stromal cells of human colorectal cancer. Cancer Cell 38, 818–828.e815 (2020). [DOI] [PubMed] [Google Scholar]
- 46.Katarkar A, Bottoni G, Clocchiatti A, Goruppi S, Bordignon P, Lazzaroni F, Gregnanin I, Ostano P, Neel V, Dotto GP, NOTCH1 gene amplification promotes expansion of Cancer Associated Fibroblast populations in human skin. Nat. Commun 11, 5126 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hill R, Song Y, Cardiff RD, Van Dyke T, Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis. Cell 123, 1001–1011 (2005). [DOI] [PubMed] [Google Scholar]
- 48.Patocs A, Zhang L, Xu Y, Weber F, Caldes T, Mutter GL, Platzer P, Eng C, Breast-cancer stromal cells with TP53 mutations and nodal metastases. N. Engl. J. Med. 357, 2543–2551 (2007). [DOI] [PubMed] [Google Scholar]
- 49.Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, Leake D, Godden EL, Albertson DG, Nieto MA, Werb Z, Bissell MJ, Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature 436, 123–127 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hu M, Yao J, Cai L, Bachman KE, van den Brûle F, Velculescu V, Polyak K, Distinct epigenetic changes in the stromal cells of breast cancers. Nat. Genet 37, 899–905 (2005). [DOI] [PubMed] [Google Scholar]
- 51.Jiang L, Gonda TA, Gamble MV, Salas M, Seshan V, Tu S, Twaddell WS, Hegyi P, Lazar G, Steele I, Varro A, Wang TC, Tycko B, Global hypomethylation of genomic DNA in cancer-associated myofibroblasts. Cancer Res. 68, 9900–9908 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Albrengues J, Bertero T, Grasset E, Bonan S, Maiel M, Bourget I, Philippe C, Herraiz Serrano C, Benamar S, Croce O, Sanz-Moreno V, Meneguzzi G, Feral CC, Cristofari G, Gaggioli C, Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat. Commun 6, 1–15 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bhagat TD, von Ahrens D, Dawlaty M, Zou Y, Baddour J, Achreja A, Zhao H, Yang L, Patel B, Kwak C, Choudhary GS, Gordon-Mitchell S, Aluri S, Bhattacharyya S, Sahu S, Bhagat P, Yu Y, Bartenstein M, Giricz O, Suzuki M, Sohal D, Gupta S, Guerrero PA, Batra S, Goggins M, Steidl U, Greally J, Agarwal B, Pradhan K, Banerjee D, Nagrath D, Maitra A, Verma A, Lactate-mediated epigenetic reprogramming regulates formation of human pancreatic cancer-associated fibroblasts. eLife 8, e50663 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Fiegl H, Millinger S, Goebel G, Müller-Holzner E, Marth C, Laird PW, Widschwendter M, Breast cancer DNA methylation profiles in cancer cells and tumor stroma: Association with HER-2/neu status in primary breast cancer. Cancer Res. 66, 29–33 (2006). [DOI] [PubMed] [Google Scholar]
- 55.Polyak K, Haviv I, Campbell IG, Co-evolution of tumor cells and their microenvironment. Trends in genetics : TIG 25, 30–38 (2009). [DOI] [PubMed] [Google Scholar]
- 56.Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, de Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R, Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mizutani Y, Kobayashi H, Iida T, Asai N, Masamune A, Hara A, Esaki N, Ushida K, Mii S, Shiraki Y, Ando K, Weng L, Ishihara S, Ponik SM, Conklin MW, Haga H, Nagasaka A, Miyata T, Matsuyama M, Kobayashi T, Fujii T, Yamada S, Yamaguchi J, Wang T, Woods SL, Worthley D, Shimamura T, Fujishiro M, Hirooka Y, Enomoto A, Takahashi M, Meflin-positive cancer-associated fibroblasts inhibit pancreatic carcinogenesis. Cancer Res. 79, 5367–5381 (2019). [DOI] [PubMed] [Google Scholar]
- 58.Miyai Y, Enomoto A, Ando Y, Takahashi M, Significance of meflin-positive cancer-associated fibroblasts in predicting response to immune checkpoint inhibitors in non-small cell lung cancer. J. Clin. Oncol 38, 3118–3118 (2020). [Google Scholar]
- 59.Silzle T, Randolph GJ, Kreutz M, Kunz-Schughart LA, The fibroblast: Sentinel cell and local immune modulator in tumor tissue. Int. J. Cancer 108, 173–180 (2004). [DOI] [PubMed] [Google Scholar]
- 60.Hinz B, Lagares D, Evasion of apoptosis by myofibroblasts: A hallmark of fibrotic diseases. Nat. Rev. Rheumatol 16, 11–31 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kalluri R, The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016). [DOI] [PubMed] [Google Scholar]
- 62.Klingberg F, Hinz B, White ES, The myofibroblast matrix: Implications for tissue repair and fibrosis. J. Pathol 229, 298–309 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Castella LF, Gabbiani G, McCulloch CA, Hinz B, Regulation of myofibroblast activities: Calcium pulls some strings behind the scene. Exp. Cell Res 316, 2390–2401 (2010). [DOI] [PubMed] [Google Scholar]
- 64.Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G, The myofibroblast: One function, multiple origins. Am. J. Pathol 170, 1807–1816 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wells RG, Schwabe RF, Origin and function of myofibroblasts in the liver. Semin. Liver Dis 35, 97–106 (2015). [DOI] [PubMed] [Google Scholar]
- 66.Mori L, Bellini A, Stacey MA, Schmidt M, Mattoli S, Fibrocytes contribute to the myofibroblast population in wounded skin and originate from the bone marrow. Exp. Cell Res 304, 81–90 (2005). [DOI] [PubMed] [Google Scholar]
- 67.Park HY, Kim JH, Park CK, VEGF induces TGF-β1 expression and myofibroblast transformation after glaucoma surgery. Am. J. Pathol 182, 2147–2154 (2013). [DOI] [PubMed] [Google Scholar]
- 68.Hu B, Gharaee-Kermani M, Wu Z, Phan SH, Epigenetic regulation of myofibroblast differentiation by DNA methylation. Am. J. Pathol 177, 21–28 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Milyavsky M, Shats I, Cholostoy A, Brosh R, Buganim Y, Weisz L, Kogan I, Cohen M, Shatz M, Madar S, Kalo E, Goldfinger N, Yuan J, Ron S, MacKenzie K, Eden A, Rotter V, Inactivation of myocardin and p16 during malignant transformation contributes to a differentiation defect. Cancer Cell 11, 133–146 (2007). [DOI] [PubMed] [Google Scholar]
- 70.Yoshida T, Sinha S, Dandré F”, Wamhoff BR, Hoofnagle MH, Kremer BE, Wang DZ, Olson EN, Owens GK, Myocardin is a key regulator of CArG-dependent transcription of multiple smooth muscle marker genes. Circ. Res 92, 856–864 (2003). [DOI] [PubMed] [Google Scholar]
- 71.Kalluri R, Weinberg RA, The basics of epithelial-mesenchymal transition. J. Clin. Invest 119, 1420–1428 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, Fu X, Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp. Biol. Med. (Maywood) 241, 1–13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kim J-H, Ham S, Lee Y, Suh GY, Lee Y-S, TTC3 contributes to TGF-β1-induced epithelial–mesenchymal transition and myofibroblast differentiation, potentially through SMURF2 ubiquitylation and degradation. Cell Death Dis. 10, 92 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.De Wever O, Demetter P, Mareel M, Bracke M, Stromal myofibroblasts are drivers of invasive cancer growth. Int. J. Cancer 123, 2229–2238 (2008). [DOI] [PubMed] [Google Scholar]
- 75.De Wever O, Nguyen Q-D, Van Hoorde L, Bracke M, Bruyneel E, Gespach C, Mareel M, Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent proinvasive signals to human colon cancer cells through RhoA and Rac. FASEB J. 18, 1016–1018 (2004). [DOI] [PubMed] [Google Scholar]
- 76.Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA, Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121, 335–348 (2005). [DOI] [PubMed] [Google Scholar]
- 77.Granot D, Addadi Y, Kalchenko V, Harmelin A, Kunz-Schughart LA, Neeman M, In vivo imaging of the systemic recruitment of fibroblasts to the angiogenic rim of ovarian carcinoma tumors. Cancer Res. 67, 9180–9189 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Barcellos-Hoff MH, Lyden D, Wang TC, The evolution of the cancer niche during multistage carcinogenesis. Nat. Rev. Cancer 13, 511–518 (2013). [DOI] [PubMed] [Google Scholar]
- 79.Ren G, Zheng X, Bommarito M, Metzger S, Walia Y, Letson J, Schroering A, Kalinoski A, Weaver D, Figy C, Yeung K, Furuta S, Reduced basal nitric oxide production induces precancerous mammary lesions via ERBB2 and TGFβ. Sci. Rep 9, 6688–6688 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang X, Xiang J, Remodeling the microenvironment before occurrence and metastasis of cancer. Int. J. Biol. Sci 15, 105–113 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Schäfer M, Werner S, Cancer as an overhealing wound: An old hypothesis revisited. Nat. Rev. Mol. Cell Biol 9, 628–638 (2008). [DOI] [PubMed] [Google Scholar]
- 82.Northey JJ, Barrett AS, Acerbi I, Hayward MK, Talamantes S, Dean IS, Mouw JK, Ponik SM, Lakins JN, Huang PJ, Wu J, Shi Q, Samson S, Keely PJ, Mukhtar RA, Liphardt JT, Shepherd JA, Hwang ES, Chen YY, Hansen KC, Littlepage LE, Weaver VM, Stiff stroma increases breast cancer risk by inducing the oncogene ZNF217. J. Clin. Invest 130, 5721–5737 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Aggarwal BB, Vijayalekshmi RV, Sung B, Targeting inflammatory pathways for prevention and therapy of cancer: Short-term friend, long-term foe. Clinical cancer research: an official journal of the American Association for Cancer Research 15, 425–430 (2009). [DOI] [PubMed] [Google Scholar]
- 84.Shah M, Crane JS, in StatPearls [Internet]. (StatPearls Publishing, 2020). [Google Scholar]
- 85.Hansson LE, Nyrén O, Hsing AW, Bergström R, Josefsson S, Chow WH, Fraumeni JF Jr., H. O. Adami, The risk of stomach cancer in patients with gastric or duodenal ulcer disease. N. Engl. J. Med 335, 242–249 (1996). [DOI] [PubMed] [Google Scholar]
- 86.Suh JK, Lee J, Lee J-H, Shin S, Tchoe HJ, Kwon J-W, Risk factors for developing liver cancer in people with and without liver disease. PloS one 13, e0206374 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K, Fibroblasts in the tumor microenvironment: Shield or spear? Int. J. Mol. Sci 19, 1532 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Steenland K, Stayner L, Silica, asbestos, man-made mineral fibers, and cancer. Cancer Causes Control 8, 491–503 (1997). [DOI] [PubMed] [Google Scholar]
- 89.Chandler C, Liu T, Buckanovich R, Coffman LG, The double edge sword of fibrosis in cancer. Transl. Res 209, 55–67 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Im J, Nho RS, Fibroblasts from patients with idiopathic pulmonary fibrosis are resistant to cisplatin-induced cell death via enhanced CK2-dependent XRCC1 activity. Apoptosis 24, 499–510 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Powell DW, Myofibroblasts: Paracrine cells important in health and disease. Trans. Am. Clin. Climatol. Assoc 111, 271–293 (2000). [PMC free article] [PubMed] [Google Scholar]
- 92.You H, Ding W, Rountree CB, Epigenetic regulation of cancer stem cell marker CD133 by transforming growth factor-beta. Hepatology (Baltimore, Md.) 51, 1635–1644 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Xie C-R, Sun H, Wang F-Q, Li Z, Yin Y-R, Fang Q-L, Sun Y, Zhao W-X, Zhang S, Zhao W-X, Wang X-M, Yin Z-Y, Integrated analysis of gene expression and DNA methylation changes induced by hepatocyte growth factor in human hepatocytes. Mol. Med. Rep 12, 4250–4258 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Vinel C, Rosser G, Guglielmi L, Constantinou M, Pomella N, Zhang X, Boot JR, Jones TA, Millner TO, Dumas AA, Rakyan V, Rees J, Thompson JL, Vuononvirta J, Nadkarni S, el Assan T, Aley N, Lin YY, Liu P, Nelander S, Sheer D, Merry CLR, Marelli-Berg F, Brandner S, Marino S, Comparative epigenetic analysis of tumour initiating cells and syngeneic EPSC-derived neural stem cells in glioblastoma. Nat. Commun 12, 6130 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Pickup MW, Mouw JK, Weaver VM, The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15, 1243–1253 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kaushik S, Pickup MW, Weaver VM, From transformation to metastasis: Deconstructing the extracellular matrix in breast cancer. Cancer Metastasis Rev. 35, 655–667 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Yoshida GJ, Azuma A, Miura Y, Orimo A, Activated fibroblast program orchestrates tumor initiation and progression; molecular mechanisms and the associated therapeutic strategies. Int. J. Mol. Sci 20, 2256 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Hammer DA, Weaver VM, Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005). [DOI] [PubMed] [Google Scholar]
- 99.Provenzano PP, Inman DR, Eliceiri KW, Keely PJ, Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK–ERK linkage. Oncogene 28, 4326–4343 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Karlsson M, Lindberg K, Karlén P, Öst Å, Thörn M, Winqvist O, Eberhardson M, Evidence for immunosurveillance in intestinal premalignant lesions. Scand. J. Immunol 71, 362–368 (2010). [DOI] [PubMed] [Google Scholar]
- 101.Anderson KG, Stromnes IM, Greenberg PD, Obstacles posed by the tumor microenvironment to T cell activity: A case for synergistic therapies. Cancer Cell 31, 311–325 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hynes RO, Naba A, Overview of the matrisome—An inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol 4, a004903 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Karsdal MA, Nielsen MJ, Sand JM, Henriksen K, Genovese F, Bay-Jensen A-C, Smith V, Adamkewicz JI, Christiansen C, Leeming DJ, Extracellular matrix remodeling: The common denominator in connective tissue diseases. Possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev. Technol 11, 70–92 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Streuli CH, Bailey N, Bissell MJ, Control of mammary epithelial differentiation: Basement membrane induces tissue-specific gene expression in the absence of cell-cell interaction and morphological polarity. J. Cell Sci 115, 1383–1395 (1991). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lu P, Weaver VM, Werb Z, The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol 196, 395–406 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cox TR, Erler JT, Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer. Dis. Model. Mech 4, 165–178 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lu P, Takai K, Weaver VM, Werb Z, Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol 3, a005058 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM, Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ruiz-Ojeda FJ, Méndez-Gutiérrez A, Aguilera CM, Plaza-Díaz J, Extracellular matrix remodeling of adipose tissue in obesity and metabolic diseases. Int. J. Mol. Sci 20, 4888 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yoo J, Seo BK, Park EK, Kwon M, Jeong H, Cho KR, Woo OH, Song SE, Cha J, Tumor stiffness measured by shear wave elastography correlates with tumor hypoxia as well as histologic biomarkers in breast cancer. Cancer Imaging 20, 85 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wang Y, Chaffee TS, LaRue RS, Huggins DN, Witschen PM, Ibrahim AM, Nelson AC, Machado HL, Schwertfeger KL, Tissue-resident macrophages promote extracellular matrix homeostasis in the mammary gland stroma of nulliparous mice. eLife 9, e57438 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Sagaradze GD, Basalova NA, Efimenko AY, Tkachuk VA, Mesenchymal stromal cells as critical contributors to tissue regeneration. Frontiers in cell and developmental biology 8, 576176–576176 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Campbell NE, Kellenberger L, Greenaway J, Moorehead RA, Linnerth-Petrik NM, Petrik J, Extracellular matrix proteins and tumor angiogenesis. J. Oncol 2010, 1–13 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Rozario T, DeSimone DW, The extracellular matrix in development and morphogenesis: A dynamic view. Dev. Biol 341, 126–140 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Bonnans C, Chou J, Werb Z, Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol 15, 786–801 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Sekiguchi R, Yamada KM, Basement membranes in development and disease. Curr. Top. Dev. Biol 130, 143–191 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Muschler J, Streuli CH, Cell-matrix interactions in mammary gland development and breast cancer. Cold Spring Harb. Perspect. Biol 2, a003202 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Daley WP, Gervais EM, Centanni SW, Gulfo KM, Nelson DA, Larsen M, ROCK1-directed basement membrane positioning coordinates epithelial tissue polarity. Development 139, 411–422 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Morrissey MA, Sherwood DR, An active role for basement membrane assembly and modification in tissue sculpting. J. Cell Sci 128, 1661–1668 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yurchenco PD, Basement membranes: Cell scaffoldings and signaling platforms. Cold Spring Harb. Perspect. Biol 3, a004911 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kalluri R, Basement membranes: Structure, assembly and role in tumour angiogenesis. Nat. Rev. Cancer 3, 422–433 (2003). [DOI] [PubMed] [Google Scholar]
- 122.Crest J, Diz-Muñoz A, Chen DY, Fletcher DA, Bilder D, Organ sculpting by patterned extracellular matrix stiffness. eLife 6, e24958 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Chang J, Chaudhuri O, Beyond proteases: Basement membrane mechanics and cancer invasion. J. Cell Biol 218, 2456–2469 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Frantz C, Stewart KM, Weaver VM, The extracellular matrix at a glance. J. Cell Sci 123, 4195–4200 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mouw JK, Ou G, Weaver VM, Extracellular matrix assembly: A multiscale deconstruction. Nat. Rev. Mol. Cell Biol 15, 771–785 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Horiguchi M, Ota M, Rifkin DB, Matrix control of transforming growth factor-β function. J. Biochem 152, 321–329 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Bissell MJ, Hall HG, Parry G, How does the extracellular matrix direct gene expression? J. Theor. Biol 99, 31–68 (1982). [DOI] [PubMed] [Google Scholar]
- 128.Bornstein P, McPherson J, Sage H. (New York: Academic Press, 1982). [Google Scholar]
- 129.Maniotis AJ, Chen CS, Ingber DE, Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. Proc. Natl. Acad. Sci. U.S.A 94, 849–854 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang N, Tytell JD, Ingber DE, Mechanotransduction at a distance: Mechanically coupling the extracellular matrix with the nucleus. Nat. Rev. Mol. Cell Biol 10, 75–82 (2009). [DOI] [PubMed] [Google Scholar]
- 131.Muiznieks LD, Keeley FW, Molecular assembly and mechanical properties of the extracellular matrix: A fibrous protein perspective. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1832, 866–875 (2013). [DOI] [PubMed] [Google Scholar]
- 132.Barczyk M, Carracedo S, Gullberg D, Integrins. Cell Tissue Res. 339, 269–280 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Carter WG, Kaur P, Gil SG, Gahr PJ, Wayner EA, Distinct functions for integrins alpha 3 beta 1 in focal adhesions and alpha 6 beta 4/bullous pemphigoid antigen in a new stable anchoring contact (SAC) of keratinocytes: Relation to hemidesmosomes. J. Cell Biol 111, 3141–3154 (1990). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Katsumi A, Orr AW, Tzima E, Schwartz MA, Integrins in mechanotransduction. J. Biol. Chem 279, 12001–12004 (2004). [DOI] [PubMed] [Google Scholar]
- 135.Zhang H, Landmann F, Zahreddine H, Rodriguez D, Koch M, Labouesse M, A tension-induced mechanotransduction pathway promotes epithelial morphogenesis. Nature 471, 99–103 (2011). [DOI] [PubMed] [Google Scholar]
- 136.Schwartz MA, Shattil SJ, Signaling networks linking integrins and rho family GTPases. Trends Biochem. Sci 25, 388–391 (2000). [DOI] [PubMed] [Google Scholar]
- 137.Legate KR, Wickström SA, Fässler R, Genetic and cell biological analysis of integrin outside-in signaling. Genes Dev. 23, 397–418 (2009). [DOI] [PubMed] [Google Scholar]
- 138.Lawson C, Schlaepfer DD, Integrin adhesions: Who's on first? What's on second? Connections between FAK and talin. Cell adh. mig 6, 302–306 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Goñi GM, Epifano C, Boskovic J, Camacho-Artacho M, Zhou J, Bronowska A, Martín MT, Eck MJ, Kremer L, Gräter F, Gervasio FL, Perez-Moreno M, Lietha D, Phosphatidylinositol 4,5-bisphosphate triggers activation of focal adhesion kinase by inducing clustering and conformational changes. Proc. Natl. Acad. Sci 111, E3177–E3186 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Welf ES, Naik UP, Ogunnaike BA, A spatial model for integrin clustering as a result of feedback between integrin activation and integrin binding. Biophys. J 103, 1379–1389 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Balaban NQ, Schwarz US, Riveline D, Goichberg P, Tzur G, Sabanay I, Mahalu D, Safran S, Bershadsky A, Addadi L, Geiger B, Force and focal adhesion assembly: A close relationship studied using elastic micropatterned substrates. Nat. Cell Biol 3, 466–472 (2001). [DOI] [PubMed] [Google Scholar]
- 142.Choquet D, Felsenfeld DP, Sheetz MP, Extracellular matrix rigidity causes strengthening of integrin–cytoskeleton linkages. Cell 88, 39–48 (1997). [DOI] [PubMed] [Google Scholar]
- 143.Riveline D, Zamir E, Balaban NQ, Schwarz US, Ishizaki T, Narumiya S, Kam Z, Geiger B, Bershadsky AD, Focal contacts as mechanosensors: Externally applied local mechanical force induces growth of focal contacts by an mDia1-dependent and ROCK-independent mechanism. J. Cell Biol 153, 1175–1186 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Walko G, Castañón MJ, Wiche G, Molecular architecture and function of the hemidesmosome. Cell Tissue Res. 360, 529–544 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Zhang H, Labouesse M, The making of hemidesmosome structures in vivo. Dev. Dyn 239, 1465–1476 (2010). [DOI] [PubMed] [Google Scholar]
- 146.Rousselle P, Beck K, Laminin 332 processing impacts cellular behavior. Cell Adh. Migr 7, 122–134 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Tran M, Rousselle P, Nokelainen P, Tallapragada S, Nguyen NT, Fincher EF, Marinkovich MP, Targeting a tumor-specific laminin domain critical for human carcinogenesis. Cancer Res. 68, 2885–2894 (2008). [DOI] [PubMed] [Google Scholar]
- 148.Zhang H, Berg JS, Li Z, Wang Y, Lång P, Sousa AD, Bhaskar A, Cheney RE, Strömblad S, Myosin-X provides a motor-based link between integrins and the cytoskeleton. Nat. Cell Biol 6, 523–531 (2004). [DOI] [PubMed] [Google Scholar]
- 149.Fletcher DA, Mullins RD, Cell mechanics and the cytoskeleton. Nature 463, 485–492 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Dahl KN, Ribeiro AJ, Lammerding J, Nuclear shape, mechanics, and mechanotransduction. Circ. Res 102, 1307–1318 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Huber F, Schnauß J, Rönicke S, Rauch P, Müller K, Fütterer C, Käs J, Emergent complexity of the cytoskeleton: From single filaments to tissue. Adv. Phys 62, 1–112 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Waterman-Storer CM, Worthylake RA, Liu BP, Burridge K, Salmon ED, Microtubule growth activates Rac1 to promote lamellipodial protrusion in fibroblasts. Nat. Cell Biol 1, 45–50 (1999). [DOI] [PubMed] [Google Scholar]
- 153.Lee SH, Dominguez R, Regulation of actin cytoskeleton dynamics in cells. Mol. Cell 29, 311–325 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kuo JC, Mechanotransduction at focal adhesions: Integrating cytoskeletal mechanics in migrating cells. J. Cell. Mol. Med 17, 704–712 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Byfield FJ, Reen RK, Shentu TP, Levitan I, Gooch KJ, Endothelial actin and cell stiffness is modulated by substrate stiffness in 2D and 3D. J. Biomech 42, 1114–1119 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Prasain N, Stevens T, The actin cytoskeleton in endothelial cell phenotypes. Microvasc. Res 77, 53–63 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Tojkander S, Gateva G, Lappalainen P, Actin stress fibers—Assembly, dynamics and biological roles. J. Cell Sci 125, 1855–1864 (2012). [DOI] [PubMed] [Google Scholar]
- 158.Hernandez P, Tirnauer JS, Tumor suppressor interactions with microtubules: Keeping cell polarity and cell division on track. Dis. Model. Mech 3, 304–315 (2010). [DOI] [PubMed] [Google Scholar]
- 159.Müsch A, Microtubule organization and function in epithelial cells. Traffic 5, 1–9 (2004). [DOI] [PubMed] [Google Scholar]
- 160.Gomes ER, Gundersen GG, Real-time centrosome reorientation during fibroblast migration. Methods Enzymol. 406, 579–592 (2006). [DOI] [PubMed] [Google Scholar]
- 161.Myers KA, Applegate KT, Danuser G, Fischer RS, Waterman CM, Distinct ECM mechanosensing pathways regulate microtubule dynamics to control endothelial cell branching morphogenesis. J. Cell Biol 192, 321–334 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Herrmann H, Aebi U, Intermediate filaments: Molecular structure, assembly mechanism, and integration into functionally distinct intracellular scaffolds. Annu. Rev. Biochem 73, 749–789 (2004). [DOI] [PubMed] [Google Scholar]
- 163.Goldman RD, Khuon S, Chou YH, Opal P, Steinert PM, The function of intermediate filaments in cell shape and cytoskeletal integrity. J. Cell Biol 134, 971–983 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Garrod D, Chidgey M, Desmosome structure, composition and function. Biochem. Biophys. Acta 1778, 572–587 (2008). [DOI] [PubMed] [Google Scholar]
- 165.Herrmann H, Bär H, Kreplak L, Strelkov SV, Aebi U, Intermediate filaments: From cell architecture to nanomechanics. Nat. Rev. Mol. Cell Biol 8, 562–573 (2007). [DOI] [PubMed] [Google Scholar]
- 166.Chang W, Worman HJ, Gundersen GG, Accessorizing and anchoring the LINC complex for multifunctionality. J. Cell Biol 208, 11–22 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Osmanagic-Myers S, Dechat T, Foisner R, Lamins at the crossroads of mechanosignaling. Genes Dev. 29, 225–237 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PCDP, Pinter J, Pajerowski JD, Spinler KR, Shin JW, Tewari M, Rehfeldt F, Speicher DW, Discher DE, Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Goldberg MW, Huttenlauch I, Hutchison CJ, Stick R, Filaments made from A- and B-type lamins differ in structure and organization. J. Cell Sci 121, 215–225 (2008). [DOI] [PubMed] [Google Scholar]
- 170.Denais CM, Gilbert RM, Isermann P, McGregor AL, te Lindert M, Weigelin B, Davidson PM, Friedl P, Wolf K, Lammerding J, Nuclear envelope rupture and repair during cancer cell migration. Science 352, 353–358 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Miroshnikova YA, Nava MM, Wickström SA, Emerging roles of mechanical forces in chromatin regulation. J. Cell Sci 130, 2243–2250 (2017). [DOI] [PubMed] [Google Scholar]
- 172.Shah P, Cheng S, Hobson CM, Colville Marshall, Paszek M, Superfine R, Lammerding J, Nuclear deformation causes DNA damage by increasing replication stress. bioRxiv 2020. 2006.2012.148890 [Preprint]. 21 October 2020. 10.1101/2020.06.12.148890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Wipff P-J, Rifkin DB, Meister J-J, Hinz B, Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix. J. Cell Biol 179, 1311–1323 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Munger JS, Sheppard D, Cross talk among TGF-β signaling pathways, integrins, and the extracellular matrix. Cold Spring Harb. Perspect. Biol 3, a005017 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Verzijl N, DeGroot J, Zaken CB, Braun-Benjamin O, Maroudas A, Bank RA, Mizrahi J, Schalkwijk CG, Thorpe SR, Baynes JW, Bijlsma JWJ, Lafeber FPJG, TeKoppele JM, Crosslinking by advanced glycation end products increases the stiffness of the collagen network in human articular cartilage: A possible mechanism through which age is a risk factor for osteoarthritis. Arthritis Rheum. 46, 114–123 (2002). [DOI] [PubMed] [Google Scholar]
- 176.Rojas A, Añazco C, González I, Araya P, Extracellular matrix glycation and receptor for advanced glycation end-products activation: A missing piece in the puzzle of the association between diabetes and cancer. Carcinogenesis 39, 515–521 (2018). [DOI] [PubMed] [Google Scholar]
- 177.Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME, Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J. Clin. Invest 108, 1853–1863 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Turner DP, Advanced glycation end-products: A biological consequence of lifestyle contributing to cancer disparity. Cancer Res. 75, 1925–1929 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Cox TR, Rumney RMH, Schoof EM, Perryman L, Høye AM, Agrawal A, Bird D, Latif NA, Forrest H, Evans HR, Huggins ID, Lang G, Linding R, Gartland A, Erler JT, The hypoxic cancer secretome induces pre-metastatic bone lesions through lysyl oxidase. Nature 522, 106–110 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 180.Ulrich P, Cerami A, Protein glycation, diabetes, and aging. Recent Prog. Horm. Res 56, 1–22 (2001). [DOI] [PubMed] [Google Scholar]
- 181.Gkogkolou P, Böhm M, Advanced glycation end products: Key players in skin aging? Dermatoendocrinol 4, 259–270 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Waghela BN, Vaidya FU, Ranjan K, Chhipa AS, Tiwari BS, Pathak C, AGE-RAGE synergy influences programmed cell death signaling to promote cancer. Mol. Cell. Biochem 476, 585–598 (2021). [DOI] [PubMed] [Google Scholar]
- 183.Baker EL, Lu J, Yu D, Bonnecaze RT, Zaman MH, Cancer cell stiffness: Integrated roles of three-dimensional matrix stiffness and transforming potential. Biophys. J 99, 2048–2057 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Chaudhuri O, Koshy ST, Branco da Cunha C, Shin JW, Verbeke CS, Allison KH, Mooney DJ, Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater 13, 970–978 (2014). [DOI] [PubMed] [Google Scholar]
- 185.Tung JC, Barnes JM, Desai SR, Sistrunk C, Conklin MW, Schedin P, Eliceiri KW, Keely PJ, Seewaldt VL, Weaver VM, Tumor mechanics and metabolic dysfunction. Free Radic. Biol. Med 79, 269–280 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zheng J, Energy metabolism of cancer: Glycolysis versus oxidative phosphorylation (Review). Oncol. Lett 4, 1151–1157 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, Zanconato F, le Digabel J, Forcato M, Bicciato S, Elvassore N, Piccolo S, Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011). [DOI] [PubMed] [Google Scholar]
- 188.Munson JM, Shieh AC, Interstitial fluid flow in cancer: Implications for disease progression and treatment. Cancer Manag. Res 6, 317–328 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Knudson W, Biswas C, Toole BP, Interactions between human tumor cells and fibroblasts stimulate hyaluronate synthesis. Proc. Natl. Acad. Sci. U.S.A 81, 6767–6771 (1984). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Nallanthighal S, Heiserman JP, Cheon D-J, The role of the extracellular matrix in cancer stemness. Frontiers in cell and developmental biology 7, 86–86 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Lemmon MA, Freed DM, Schlessinger J, Kiyatkin A, The dark side of cell signaling: Positive roles for negative regulators. Cell 164, 1172–1184 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Baschieri F, Dayot S, Elkhatib N, Ly N, Capmany A, Schauer K, Betz T, Vignjevic DM, Poincloux R, Montagnac G, Frustrated endocytosis controls contractility-independent mechanotransduction at clathrin-coated structures. Nat. Commun 9, 3825 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Klein EA, Yin L, Kothapalli D, Castagnino P, Byfield FJ, Xu T, Levental I, Hawthorne E, Janmey PA, Assoian RK, Cell-cycle control by physiological matrix elasticity and in vivo tissue stiffening. Current biology: CB 19, 1511–1518 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Aragona M, Panciera T, Manfrin A, Giulitti S, Michielin F, Elvassore N, Dupont S, Piccolo S, A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell 154, 1047–1059 (2013). [DOI] [PubMed] [Google Scholar]
- 195.Kim J-H, Asthagiri AR, Matrix stiffening sensitizes epithelial cells to EGF and enables the loss of contact inhibition of proliferation. J. Cell Sci 124, 1280–1287 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ingallina E, Sorrentino G, Bertolio R, Lisek K, Zannini A, Azzolin L, Severino LU, Scaini D, Mano M, Mantovani F, Rosato A, Bicciato S, Piccolo S, del Sal G, Mechanical cues control mutant p53 stability through a mevalonate-RhoA axis. Nat. Cell Biol 20, 28–35 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Deng M, Lin J, Nowsheen S, Liu T, Zhao Y, Villalta PW, Sicard D, Tschumperlin DJ, Lee SB, Kim JJ, Lou Z, Extracellular matrix stiffness determines DNA repair efficiency and cellular sensitivity to genotoxic agents. Sci. Adv 6, eabb2630 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Mouw JK, Yui Y, Damiano L, Bainer RO, Lakins JN, Acerbi I, Ou G, Wijekoon AC, Levental KR, Gilbert PM, Hwang ES, Chen YY, Weaver VM, Tissue mechanics modulate microRNA-dependent PTEN expression to regulate malignant progression. Nat. Med 20, 360–367 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Vachon PH, Integrin signaling, cell survival, and anoikis: Distinctions, differences, and differentiation. J. Signal Transduct 2011, 738137 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Friedland JC, Lee MH, Boettiger D, Mechanically activated integrin switch controls alpha5beta1 function. Science 323, 642–644 (2009). [DOI] [PubMed] [Google Scholar]
- 201.Paoli P, Giannoni E, Chiarugi P, Anoikis molecular pathways and its role in cancer progression. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 1833, 3481–3498 (2013). [DOI] [PubMed] [Google Scholar]
- 202.Marconi A, Dallaglio K, Lotti R, Vaschieri C, Truzzi F, Fantini F, Pincelli C, Survivin identifies keratinocyte stem cells and is downregulated by anti-beta1 integrin during anoikis. Stem Cells 25, 149–155 (2007). [DOI] [PubMed] [Google Scholar]
- 203.Ebata T, Mitsui Y, Sugimoto W, Maeda M, Araki K, Machiyama H, Harada I, Sawada Y, Fujita H, Hirata H, Kawauchi K, Substrate stiffness influences doxorubicin-induced p53 activation via ROCK2 expression. Biomed. Res. Int 2017, 1–10 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Reddel RR, Telomere maintenance mechanisms in cancer: Clinical implications. Curr. Pharm. Des 20, 6361–6374 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Shay JW, Wright WE, Senescence and immortalization: Role of telomeres and telomerase. Carcinogenesis 26, 867–874 (2005). [DOI] [PubMed] [Google Scholar]
- 206.Kusoglu A, Bagca BG, Ay NPO, Gunduz C, Avci CB, Telomerase inhibition regulates EMT mechanism in breast cancer stem cells. Gene 759, 145001 (2020). [DOI] [PubMed] [Google Scholar]
- 207.Park J, Kim DS, Shim TS, Lim CM, Koh Y, Lee SD, Kim WS, Kim WD, Lee JS, Song KS, Lung cancer in patients with idiopathic pulmonary fibrosis. Eur. Respir. J 17, 1216–1219 (2001). [DOI] [PubMed] [Google Scholar]
- 208.Soysal SD, Ng CKY, Costa L, Weber WP, Paradiso V, Piscuoglio S, Muenst S, Genetic alterations in benign breast biopsies of subsequent breast cancer patients. Front. Med 6, 166–166 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Knouse KA, Lopez KE, Bachofner M, Amon A, Chromosome segregation fidelity in epithelia requires tissue architecture. Cell 175, 200–211.e213 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Tse HT, Weaver WM, Di Carlo D, Increased asymmetric and multi-daughter cell division in mechanically confined microenvironments. PloS one 7, e38986 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Irianto J, Xia Y, Pfeifer CR, Athirasala A, Ji J, Alvey C, Tewari M, Bennett RR, Harding SM, Liu AJ, Greenberg RA, Discher DE, DNA damage follows repair factor depletion and portends genome variation in cancer cells after pore migration. Current biology : CB 27, 210–223 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Bennett RR, Pfeifer CR, Irianto J, Xia Y, Discher DE, Liu AJ, Elastic-fluid model for dna damage and mutation from nuclear fluid segregation due to cell migration. Biophys. J 112, 2271–2279 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Peiris-Pagès M, Sotgia F, Lisanti MP, Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget 6, 10728–10745 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Ruan Y, Ogana H, Gang E, Kim HN, Kim Y-M, Wnt signaling in the tumor microenvironment. Adv. Exp. Med. Biol 1270, 107–121 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Dzobo K, Senthebane DA, Ganz C, Thomford NE, Wonkam A, Dandara C, Advances in therapeutic targeting of cancer stem cells within the tumor microenvironment: An updated review. Cell 9, 1896 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, Hynes RO, Jain RK, Janowitz T, Jorgensen C, Kimmelman AC, Kolonin MG, Maki RG, Powers RS, Puré E, Ramirez DC, Scherz-Shouval R, Sherman MH, Stewart S, Tlsty TD, Tuveson DA, Watt FM, Weaver V, Weeraratna AT, Werb Z, A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 20, 174–186 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Cosse JP, Michiels C, Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer Agents Med Chem. 8, 790–797 (2008). [DOI] [PubMed] [Google Scholar]
- 218.Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, True L, Nelson PS, Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat. Med 18, 1359–1368 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, du J, Davis A, Mongare MM, Gould J, Frederick DT, Cooper ZA, Chapman PB, Solit DB, Ribas A, Lo RS, Flaherty KT, Ogino S, Wargo JA, Golub TR, Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kuo MT, Liu Z, Wei Y, Lin-Lee YC, Tatebe S, Mills GB, Unate H, Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-κB signaling. Oncogene 21, 1945–1954 (2002). [DOI] [PubMed] [Google Scholar]
- 221.Pramanik KC, Makena MR, Bhowmick K, Pandey MK, Advancement of NF-κB signaling pathway: A novel target in pancreatic cancer. Int. J. Mol. Sci 19, 3890 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Giannoni E, Bianchini F, Calorini L, Chiarugi P, Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness. Antioxid. Redox Signal 14, 2361–2371 (2011). [DOI] [PubMed] [Google Scholar]
- 223.Singh A, Settleman J, EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Lu L, Zhou D, Jiang X, Song K, Li K, Ding W, Loss of E-cadherin in multidrug resistant breast cancer cell line MCF-7/Adr: Possible implication in the enhanced invasive ability. Eur. Rev. Med. Pharmacol. Sci 16, 1271–1279 (2012). [PubMed] [Google Scholar]
- 225.Yu T, Yang G, Hou Y, Tang X, Wu C, Wu X.-a., Guo L, Zhu Q, Luo H, Du Y.-e., Wen S, Xu J, Yin J, Tu G, Liu M, Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene 36, 2131–2145 (2017). [DOI] [PubMed] [Google Scholar]
- 226.Marcucci F, Rumio C, Glycolysis-induced drug resistance in tumors—A response to danger signals? Neoplasia 23, 234–245 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Apicella M, Giannoni E, Fiore S, Ferrari KJ, Fernández-Pérez D, Isella C, Granchi C, Minutolo F, Sottile A, Comoglio PM, Medico E, Pietrantonio F, Volante M, Pasini D, Chiarugi P, Giordano S, Corso S, Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies. Cell Metab. 28, 848–865.e846 (2018). [DOI] [PubMed] [Google Scholar]
- 228.Feig C, Jones JO, Kraman M, Wells RJB, Deonarine A, Chan DS, Connell CM, Roberts EW, Zhao Q, Caballero OL, Teichmann SA, Janowitz T, Jodrell DI, Tuveson DA, Fearon DT, Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. U.S.A 110, 20212–20217 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Monteran L, Erez N, The dark side of fibroblasts: Cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front. Immunol 10, 1835 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, Sirven P, Magagna I, Fuhrmann L, Bernard C, Bonneau C, Kondratova M, Kuperstein I, Zinovyev A, Givel A-M, Parrini M-C, Soumelis V, Vincent-Salomon A, Mechta-Grigoriou F, Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 33, 463–479.e410 (2018). [DOI] [PubMed] [Google Scholar]
- 231.Smith BN, Bhowmick NA, Role of EMT in metastasis and therapy resistance. J. Clin. Med 5, 17 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Shibue T, Weinberg RA, EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol 14, 611–629 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Lee A-F, Chen M-C, Chen C-J, Yang C-J, Huang M-S, Liu Y-P, Reverse epithelial-mesenchymal transition contributes to the regain of drug sensitivity in tyrosine kinase inhibitor-resistant non-small cell lung cancer cells. PloS one 12, e0180383 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Williams ED, Gao D, Redfern A, Thompson EW, Controversies around epithelial-mesenchymal plasticity in cancer metastasis. Nat. Rev. Cancer 19, 716–732 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Reddy TP, Rosato RR, Li X, Moulder S, Piwnica-Worms H, Chang JC, A comprehensive overview of metaplastic breast cancer: Clinical features and molecular aberrations. Breast Cancer Res. 22, 121 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Giroux V, Rustgi AK, Metaplasia: Tissue injury adaptation and a precursor to the dysplasia-cancer sequence. Nat. Rev. Cancer 17, 594–604 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Adamson IY, Bowden DH, Bleomycin-induced injury and metaplasia of alveolar type 2 cells. Relationship of cellular responses to drug presence in the lung. Am. J. Pathol 96, 531–544 (1979). [PMC free article] [PubMed] [Google Scholar]
- 238.Du B, Shim JS, Targeting epithelial-mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules 21, 965 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Ashrafizadeh M, Zarrabi A, Hushmandi K, Kalantari M, Mohammadinejad R, Javaheri T, Sethi G, Association of the epithelial-mesenchymal transition (EMT) with cisplatin resistance. Int. J. Mol. Sci 21, 4002 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Valkenburg KC, de Groot AE, Pienta KJ, Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol 15, 366–381 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Bissell MJ, Radisky DC, Rizki A, Weaver VM, Petersen OW, The organizing principle: Microenvironmental influences in the normal and malignant breast. Differentiation 70, 537–546 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Lampi MC, Reinhart-King CA, Targeting extracellular matrix stiffness to attenuate disease: From molecular mechanisms to clinical trials. Sci. Transl. Med 10, eaao0475 (2018). [DOI] [PubMed] [Google Scholar]
- 243.Liu L, Zhang SX, Liao W, Farhoodi HP, Wong CW, Chen CC, Ségaliny AI, Chacko JV, Nguyen LP, Lu M, Polovin G, Pone EJ, Downing TL, Lawson DA, Digman MA, Zhao W, Mechanoresponsive stem cells to target cancer metastases through biophysical cues. Sci. Transl. Med 9, eaan2966 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Wang J, Kaplan JA, Colson YL, Grinstaff MW, Mechanoresponsive materials for drug delivery: Harnessing forces for controlled release. Adv. Drug Deliv. Rev 108, 68–82 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Philippeos C, Telerman SB, Oulès B, Pisco AO, Shaw TJ, Elgueta R, Lombardi G, Driskell RR, Soldin M, Lynch MD, Watt FM, Spatial and single-cell transcriptional profiling identifies functionally distinct human dermal fibroblast subpopulations. J. Invest. Dermatol 138, 811–825 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Tabib T, Morse C, Wang T, Chen W, Lafyatis R, SFRP2/DPP4 and FMO1/LSP1 define major fibroblast populations in human skin. J. Invest. Dermatol 138, 802–810 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Solé-Boldo L, Raddatz G, Schütz S, Mallm JP, Rippe K, Lonsdorf AS, Rodríguez-Paredes M, Lyko F, Single-cell transcriptomes of the human skin reveal age-related loss of fibroblast priming. Communications Biology 3, 188 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, Sivajothi S, Armstrong TD, Engle DD, Yu KH, Hao Y, Wolfgang CL, Park Y, Preall J, Jaffee EM, Califano A, Robson P, Tuveson DA, Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Friedman G, Levi-Galibov O, David E, Bornstein C, Giladi A, Dadiani M, Mayo A, Halperin C, Pevsner-Fischer M, Lavon H, Mayer S, Nevo R, Stein Y, Balint-Lahat N, Barshack I, Ali HR, Caldas C, Nili-Gal-Yam E, Alon U, Amit I, Scherz-Shouval R, Cancer-associated fibroblast compositions change with breast cancer progression linking the ratio of S100A4+ and PDPN+ CAFs to clinical outcome. Nature Cancer 1, 692–708 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Bartoschek M, Oskolkov N, Bocci M, Lövrot J, Larsson C, Sommarin M, Madsen CD, Lindgren D, Pekar G, Karlsson G, Ringnér M, Bergh J, Björklund Å, Pietras K, Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat. Commun 9, 5150 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Li H, Courtois ET, Sengupta D, Tan Y, Chen KH, Goh JJL, Kong SL, Chua C, Hon LK, Tan WS, Wong M, Choi PJ, Wee LJK, Hillmer AM, Tan IB, Robson P, Prabhakar S, Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat. Genet 49, 708–718 (2017). [DOI] [PubMed] [Google Scholar]
- 252.Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, Rodman C, Luo CL, Mroz EA, Emerick KS, Deschler DG, Varvares MA, Mylvaganam R, Rozenblatt-Rosen O, Rocco JW, Faquin WC, Lin DT, Regev A, Bernstein BE, Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624.e1624 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, Bassez A, Decaluwé H, Pircher A, van den Eynde K, Weynand B, Verbeken E, de Leyn P, Liston A, Vansteenkiste J, Carmeliet P, Aerts S, Thienpont B, Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med 24, 1277–1289 (2018). [DOI] [PubMed] [Google Scholar]
- 254.Kuppe C, Ibrahim MM, Kranz J, Zhang X, Ziegler S, Perales-Patón J, Jansen J, Reimer KC, Smith JR, Dobie R, Wilson-Kanamori JR, Halder M, Xu Y, Kabgani N, Kaesler N, Klaus M, Gernhold L, Puelles VG, Huber TB, Boor P, Menzel S, Hoogenboezem RM, Bindels EMJ, Steffens J, Floege J, Schneider RK, Saez-Rodriguez J, Henderson NC, Kramann R, Decoding myofibroblast origins in human kidney fibrosis. Nature 589, 281–286 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Young MD, Mitchell TJ, Vieira Braga FA, Tran MGB, Stewart BJ, Ferdinand JR, Collord G, Botting RA, Popescu DM, Loudon KW, Vento-Tormo R, Stephenson E, Cagan A, Farndon SJ, del Castillo Velasco-Herrera M, Guzzo C, Richoz N, Mamanova L, Aho T, Armitage JN, Riddick ACP, Mushtaq I, Farrell S, Rampling D, Nicholson J, Filby A, Burge J, Lisgo S, Maxwell PH, Lindsay S, Warren AY, Stewart GD, Sebire N, Coleman N, Haniffa M, Teichmann SA, Clatworthy M, Behjati S, Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science 361, 594–599 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Habermann AC, Gutierrez AJ, Bui LT, Yahn SL, Winters NI, Calvi CL, Peter L, Chung M-I, Taylor CJ, Jetter C, Raju L, Roberson J, Ding G, Wood L, Sucre JMS, Richmond BW, Serezani AP, Donnell WJM, Mallal SB, Bacchetta MJ, Loyd JE, Shaver CM, Ware LB, Bremner R, Walia R, Blackwell TS, Banovich NE, Kropski JA, Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci. Adv. 6, eaba1972 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N, Ahangari F, Chu SG, Raby BA, De Iuliis G, Januszyk M, Duan Q, Arnett HA, Siddiqui A, Washko GR, Homer R, Yan X, Rosas IO, Kaminski N, Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci. Adv 6, eaba1983 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Valenzi E, Bulik M, Tabib T, Morse C, Sembrat J, Trejo Bittar H, Rojas M, Lafyatis R, Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann. Rheum. Dis 78, 1379–1387 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Dobie R, Wilson-Kanamori JR, Henderson BEP, Smith JR, Matchett KP, Portman JR, Wallenborg K, Picelli S, Zagorska A, Pendem SV, Hudson TE, Wu MM, Budas GR, Breckenridge DG, Harrison EM, Mole DJ, Wigmore SJ, Ramachandran P, Ponting CP, Teichmann SA, Marioni JC, Henderson NC, Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep. 29, 1832–1847.e1838 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Mederacke I, Dapito DH, Affò S, Uchinami H, Schwabe RF, High-yield and high-purity isolation of hepatic stellate cells from normal and fibrotic mouse livers. Nat. Protoc 10, 305–315 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, Shi X, Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res 37, 324 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Affo S, Yu L-X, Schwabe RF, The role of cancer-associated fibroblasts and fibrosis in liver cancer. Annual Review of Pathology: Mechanisms of Disease 12, 153–186 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Yavuz BG, Pestana RC, Abugabal YI, Krishnan S, Chen J, Hassan MM, Wolff RA, Rashid A, Amin HM, Kaseb AO, Origin and role of hepatic myofibroblasts in hepatocellular carcinoma. Oncotarget 11, 1186–1201 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Ramachandran P, Dobie R, Wilson-Kanamori JR, Dora EF, Henderson BEP, Luu NT, Portman JR, Matchett KP, Brice M, Marwick JA, Taylor RS, Efremova M, Vento-Tormo R, Carragher NO, Kendall TJ, Fallowfield JA, Harrison EM, Mole DJ, Wigmore SJ, Newsome PN, Weston CJ, Iredale JP, Tacke F, Pollard JW, Ponting CP, Marioni JC, Teichmann SA, Henderson NC, Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Xu J, Cong M, Park TJ, Scholten D, Brenner DA, Kisseleva T, Contribution of bone marrow-derived fibrocytes to liver fibrosis. Hepatobiliary Surgery and Nutrition 4, 34–47 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Barrera LN, Evans A, Lane B, Brumskill S, Oldfield FE, Campbell F, Andrews T, Lu Z, Perez-Mancera PA, Liloglou T, Ashworth M, Jalali M, Dawson R, Nunes Q, Phillips PA, Timms JF, Halloran C, Greenhalf W, Neoptolemos JP, Costello E, Fibroblasts from distinct pancreatic pathologies exhibit disease-specific properties. Cancer Res. 80, 2861–2873 (2020). [DOI] [PubMed] [Google Scholar]
- 267.Dominguez CX, Müller S, Keerthivasan S, Koeppen H, Hung J, Gierke S, Breart B, Foreman O, Bainbridge TW, Castiglioni A, Senbabaoglu Y, Modrusan Z, Liang Y, Junttila MR, Klijn C, Bourgon R, Turley SJ, Single-cell rna sequencing reveals stromal evolution into LRRC15(+) myofibroblasts as a determinant of patient response to cancer Immunotherapy. Cancer Discov. 10, 232–253 (2020). [DOI] [PubMed] [Google Scholar]
- 268.Bernard V, Semaan A, Huang J, San Lucas FA, Mulu FC, Stephens BM, Guerrero PA, Huang Y, Zhao J, Kamyabi N, Sen S, Scheet PA, Taniguchi CM, Kim MP, Tzeng CW, Katz MH, Singhi AD, Maitra A, Alvarez HA, Single-cell transcriptomics of pancreatic cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. Clin. Cancer Res 25, 2194–2205 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Marangoni RG, Korman BD, Wei J, Wood TA, Graham LV, Whitfield ML, Scherer PE, Tourtellotte WG, Varga J, Myofibroblasts in murine cutaneous fibrosis originate from adiponectin-positive intradermal progenitors. Arthritis Rheumatol. 67, 1062–1073 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Kim HS, Shin MS, Cheon MS, Kim JW, Lee C, Kim WH, Kim YS, Jang BG, GREM1 is expressed in the cancer-associated myofibroblasts of basal cell carcinomas. Plos One 12, e0174565 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Shook BA, Wasko RR, Rivera-Gonzalez GC, Salazar-Gatzimas E, López-Giráldez F, Dash BC, Muñoz-Rojas AR, Aultman KD, Zwick RK, Lei V, Arbiser JL, Miller-Jensen K, Clark DA, Hsia HC, Horsley V, Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science 362, eaar2971 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]





