Fact-checked by Grok 2 weeks ago

Fibroblast

Fibroblasts are mesenchymal cells that synthesize and maintain the (), providing structural support to and organs throughout the body. These cells, which constitute a major component of , are characterized by their elongated, spindle-shaped and ability to produce key components such as collagens, , and . Originating primarily from mesodermal precursors during embryonic development, fibroblasts can also arise through epithelial-to-mesenchymal transition () or endothelial-to-mesenchymal transition (EndMT) in specific contexts, such as cardiac formation. In healthy tissues, fibroblasts play essential roles in maintaining homeostasis by regulating ECM turnover and serving as signaling niche cells that influence neighboring cell behaviors through secreted factors like growth factors and cytokines. During wound healing, they proliferate, migrate to injury sites, and differentiate into contractile myofibroblasts expressing alpha-smooth muscle actin (α-SMA), which facilitate tissue contraction and repair by remodeling the ECM. However, in pathological conditions, dysregulated fibroblast activation leads to excessive ECM deposition, driving fibrosis—a process implicated in approximately 45% of deaths in developed countries, affecting organs like the lungs, liver, and heart. Fibroblasts exhibit remarkable heterogeneity, with transcriptional profiles varying significantly across organs; for instance, less than 20% of fibroblast genes overlap between tissues like the heart, , intestine, and in mice. This diversity is encoded by positional cues such as and contributes to their specialized functions, including , osteogenesis, and immune modulation. In cancer, fibroblasts within the , often termed cancer-associated fibroblasts (CAFs), promote tumor progression by altering stiffness, secreting pro-angiogenic factors, and suppressing anti-tumor immunity. Their plasticity and context-dependent roles make fibroblasts critical targets for and anti-fibrotic therapies.

Structure and Morphology

Cellular Features

Fibroblasts are characterized by an elongated, spindle-shaped or morphology, often with stellate features and multiple thin cytoplasmic projections that facilitate interactions with the surrounding . These cells' size and shape can vary based on the specific environment and functional state. Unlike epithelial cells, which exhibit distinct apical-basal , fibroblasts lack such organized , presenting instead a more isotropic distribution of organelles and a front-back primarily during . Ultrastructural analysis via reveals a rich in organelles adapted for synthetic activity, including abundant cisternae of rough and a well-developed Golgi apparatus, which occupy significant portions of the perinuclear region. These features underscore the 's role in protein processing and secretion, with the rough often appearing dilated in active fibroblasts. Mitochondria and other organelles are distributed throughout the , supported by a network of cytoskeletal elements. Thin, elongated cytoplasmic extensions extend from the body, enabling close apposition to adjacent fibers without forming tight junctions. A prominent cytoskeletal feature of fibroblasts is the presence of intermediate filaments, which provide structural integrity and mechanical resilience to the . These filaments, approximately 10 in diameter, form a dynamic network that interconnects organelles and adheres to the plasma membrane, contributing to the maintenance of the 's elongated form. Vimentin expression is a hallmark mesenchymal marker, distinguishing fibroblasts from other types. Fibroblasts reside predominantly in connective tissues, where they form the stromal framework of organs; common locations include the dermis of the skin, the paratenon and endotenon of tendons, and the interstitial stroma of visceral organs such as the lungs, heart, and kidneys. In these sites, their morphology adapts to the local matrix density, appearing more flattened in dense tissues like tendons and more irregular in loose areolar connective tissue. These structural attributes equip fibroblasts to synthesize and organize extracellular matrix components, influencing tissue architecture and resilience.

Distinction from Fibrocytes

Fibrocytes represent a distinct of circulating cells that serve as precursors to fibroblasts, originating from bone marrow-derived monocytes rather than the resident populations that give rise to mature fibroblasts. These cells are recruited to sites of injury or in the peripheral tissues, where they differentiate into fibroblast-like cells capable of contributing to production. Unlike resident fibroblasts, which are primarily tissue-embedded and derived from embryonic or local progenitors, fibrocytes maintain a hematopoietic lineage throughout their circulation phase. The concept of fibrocytes was first described in by Bucala et al., who identified them as a novel leukocyte subpopulation in human peripheral blood that expresses both hematopoietic and mesenchymal markers, mediating tissue repair processes. In healthy individuals, fibrocytes constitute approximately 0.1-0.5% of circulating non-erythrocytic leukocytes, though their numbers can increase under inflammatory conditions. This low baseline prevalence underscores their role as a responsive, rather than constitutive, component of the immune and repair systems. A key distinction lies in their molecular markers: fibrocytes co-express the pan-leukocyte antigen CD45, the marker , and early I, reflecting their dual hematopoietic-mesenchymal identity, whereas mature fibroblasts typically downregulate CD45 and upon tissue integration and full differentiation. This marker profile enables fibrocytes to retain immune functions, such as via , which is less prominent in differentiated fibroblasts. Morphologically, fibrocytes appear as smaller, more rounded or oval-shaped cells with spindle-like extensions suited for , often exhibiting expression (e.g., α4β1) that facilitates their into tissues; in contrast, mature fibroblasts display an elongated, branched with an elliptical optimized for in . Upon , fibrocytes undergo a functional transition, producing and other components to support , but they remain transient progenitors compared to the long-lived, resident fibroblasts that dominate sustained tissue remodeling. This precursor relationship highlights fibrocytes' role in bridging hematopoiesis and fibrogenesis, particularly in pathological contexts like .

Origin and Development

Embryonic and Postnatal Origins

Fibroblasts primarily originate from mesodermal precursors during embryogenesis, with the majority deriving from the paraxial and , which give rise to connective tissue frameworks in various organs. In specific regions, such as craniofacial tissues, fibroblasts arise from neural crest-derived , contributing to the structural diversity of head and neck s. This mesodermal specification is regulated by key transcription factors, including Twist1, , and Foxc2, which drive the transition to mesenchymal lineages and ensure proper fibroblast differentiation. Fibroblasts first appear in human embryos during early , around weeks 7–8, coinciding with the onset of when mesenchymal cells proliferate to support tissue patterning and deposition. Throughout this period, from weeks 7 to 8 post-conception, fibroblasts play a critical role in by synthesizing provisional matrices that guide epithelial branching and vascular invasion in developing organs like the lungs, heart, and . Postnatally, fibroblast populations are maintained through self-renewal of resident pools within s, facilitated by signaling pathways such as PDGF, which promotes and in organs like the and . Additionally, recruitment from bone marrow-derived mesenchymal progenitors contributes to replenishment, particularly during turnover or mild , although this source is secondary to local self-renewal in steady-state conditions. These mechanisms ensure sustained fibroblast function across adulthood. The developmental origins of fibroblasts show strong across mammalian species, as evidenced by similar mesodermal and contributions in mice and s, enabling the use of models for studying fibroblast biology. In contrast, avian models, such as embryos, reveal variations in timing and migratory patterns of mesenchymal precursors, which have been instrumental in elucidating early fibroblast progenitor dynamics despite differences in yolk-dependent development.

Subtypes and Heterogeneity

Fibroblasts exhibit significant heterogeneity, manifesting as distinct subtypes with specialized functions that contribute to and repair. Major subtypes include matrix fibroblasts, also known as matrifibrocytes, which specialize in () production and remodeling by expressing genes such as , Cilp2, and Comp, particularly during maturation phases. Another key subtype is inflammatory fibroblasts, which secrete proinflammatory cytokines and like IL-6, IL-1, and MCP-1 in response to immune signals, thereby modulating local immune environments. These subtypes arise from shared pools but diverge through transcriptional and epigenetic programs, enabling context-specific roles across tissues. Recent advances in single-cell RNA sequencing (scRNA-seq) have illuminated this diversity through comprehensive atlases. A 2025 study on fibroblasts identified six major subtypes in healthy : superficial (papillary) fibroblasts expressing collagens like COL13A1 for epithelial support; universal (reticular) fibroblasts marked by PI16 and as potential precursors; FRC-like subtypes with immune genes such as CCL19 for niche maintenance; perivascular fibroblasts enriched in PPARG; follicle-associated subtypes (e.g., DPEP1+); and Schwann-like nerve-interfacing subtypes (e.g., NGFR+). Cross-tissue analyses from 517 human samples across 11 organs revealed nine universal subtypes, including PI16+ progenitors and LRRC15+ matrix fibroblasts, alongside six tissue-specific clusters, underscoring conserved and organ-adapted profiles. Tissue-specific variations further highlight fibroblast heterogeneity, with dermal fibroblasts showing layered distributions (e.g., papillary vs. reticular) tied to architecture, while cardiac fibroblasts express distinct markers like Tcf21 in resident populations to regulate epicardial-mesenchymal transitions and maintain . Epigenetic mechanisms, particularly patterns, drive this diversity by establishing stable, heritable marks that differentiate fibroblast phenotypes, such as hypermethylation of loci associated with genes in lung-derived subtypes. Functionally, these subtypes display differential responses to stimuli like transforming growth factor-β (TGF-β), with matrix fibroblasts showing robust activation of synthesis pathways via TGF-β1, whereas inflammatory subtypes exhibit varied sensitivity to TGF-β isoforms, influencing output and immune modulation without uniform differentiation. This subtype-specific responsiveness ensures adaptive tissue responses while linking back to developmental lineages established embryonically.

Physiological Functions

Extracellular Matrix Synthesis

Fibroblasts are the primary cellular source of () components in connective tissues, synthesizing fibrillar collagens such as types I, III, and V to provide structural integrity during tissue . , the most abundant, forms strong that confer tensile strength, while types III and V contribute to the elasticity and nucleation of , respectively. The begins intracellularly in the , where procollagen chains undergo post-translational modifications including of and residues, , and assembly into a triple helical procollagen . Upon , procollagen is processed extracellularly by procollagen peptidases, which cleave N- and C-terminal propeptides to yield mature tropocollagen, the basic unit of . The stability of these collagen fibrils is enhanced through cross-linking mediated by lysyl oxidase (LOX), a copper-dependent secreted by fibroblasts that oxidizes specific and hydroxylysine residues, forming covalent bonds between adjacent tropocollagen molecules. This enzymatic cross-linking is essential for the mechanical properties of the , preventing fibril slippage and ensuring long-term tissue resilience in steady-state conditions. In addition to s, fibroblasts produce s like , a small leucine-rich that binds to fibrils to regulate their diameter and assembly, and glycoproteins such as , which facilitates and organizes the provisional matrix by linking collagens to on the cell surface. These components interact to form a functional network that supports tissue architecture. ECM synthesis in fibroblasts is tightly regulated by growth factors, including (PDGF) and (FGF), which modulate and protein production to maintain . PDGF stimulates fibroblast and enhances the transcription of and genes via signaling, ensuring balanced matrix deposition. Similarly, FGF isoforms, such as FGF-2, promote ECM assembly by influencing expression and inhibiting excessive degradation, thereby supporting steady-state maintenance. Enzymatic remodeling of the ECM is achieved through matrix metalloproteinases (MMPs), zinc-dependent endopeptidases produced by fibroblasts that selectively degrade and other matrix components to facilitate turnover and prevent accumulation. For instance, MMP-1 (collagenase-1) initiates the cleavage of fibrillar collagens, while MMP-2 and MMP-3 target non-fibrillar elements, with their activity balanced by tissue inhibitors of metalloproteinases (TIMPs) to preserve ECM integrity. In steady-state conditions, fibroblasts dedicate a significant portion of their protein synthetic capacity to production, underscoring their role in maintaining volume and function. Variations in output among fibroblast subtypes, such as dermal versus tendon-derived cells, reflect tissue-specific demands but do not alter the core biosynthetic pathways.

Wound Healing and Tissue Remodeling

Fibroblasts play a pivotal role in the proliferative phase of , where they migrate into the injury site and contribute to the formation of , a provisional matrix that supports tissue regeneration. During this phase, fibroblasts proliferate and deposit extracellular matrix components, such as and , to stabilize the wound bed and facilitate . This process begins shortly after the inflammatory phase, with fibroblasts originating from adjacent or circulating precursors responding to chemotactic signals to fill the defect. A key aspect of fibroblast function in closure involves their into myofibroblasts, specialized cells that express α-smooth muscle (α-SMA) and generate contractile forces through actin-myosin interactions. This is triggered by mechanical and growth factors, enabling myofibroblasts to pull edges together via , thereby accelerating closure and reducing the size of the open . The de novo expression of α-SMA enhances the contractile activity of these cells, which is essential for efficient repair in the proliferative and remodeling phases. Studies have shown that preventing closure mechanically increases α-SMA expression, underscoring the interplay between and . Cytokines such as interleukin-4 (IL-4) and transforming growth factor-β1 (TGF-β1) orchestrate fibroblast migration and proliferation during healing. TGF-β1 promotes fibroblast infiltration into the wound, stimulates their proliferation, and induces expression of genes for production, thereby supporting development. Similarly, IL-4 enhances fibroblast proliferation, migration, and synthesis, often in coordination with TGF-β signaling pathways to amplify repair responses. These cytokines are released by immune cells and , creating a microenvironment conducive to fibroblast activation. In the resolution phase of , myofibroblasts undergo to prevent excessive matrix deposition and scarring, allowing the tissue to regain normal architecture. This is induced when mechanical tension decreases and the achieves sufficient tensile strength, typically after . Failure of this apoptotic can lead to persistent myofibroblasts and abnormal repair, but in normal healing, it ensures timely termination of contraction and remodeling. Research demonstrates that factors like fibromodulin can selectively promote myofibroblast post-, reducing scar formation. Experimental models, such as scratch assays, have been instrumental in quantifying fibroblast migration dynamics during . In these assays, a confluent is scratched to simulate a , and fibroblast closure rates are measured, typically ranging from 10-20 μm/hour under standard conditions, reflecting their chemotactic response to growth factors. These models highlight how environmental cues influence migration speed and provide insights into therapeutic modulation of repair processes.

Pathological Roles

Inflammation and Fibrosis

Fibroblasts become activated in response to pro-fibrotic signals, particularly through the transforming growth factor-β (TGF-β) signaling pathway, which drives their differentiation into myofibroblasts and sustains excessive extracellular matrix (ECM) deposition. This pathway involves TGF-β binding to its receptors, leading to Smad-dependent transcription that upregulates genes for ECM components like collagen and fibronectin, resulting in persistent fibrotic remodeling in chronic inflammatory conditions. Persistent TGF-β activation prevents myofibroblast apoptosis, thereby maintaining a cycle of ECM accumulation that contributes to tissue stiffness and organ dysfunction. In fibrosis, mechanisms extend beyond collagen I overproduction to include disruptions in basement membrane integrity and dysregulation of matricellular proteins, which modulate cell-ECM interactions and exacerbate pathological remodeling. Basement membrane alterations, such as fragmentation of collagen and laminin networks, impair epithelial barrier function and facilitate fibroblast invasion into alveolar spaces, as observed in lung models. Matricellular proteins like and , secreted by activated fibroblasts, further promote ECM cross-linking and inflammatory signaling, amplifying fibrotic progression in a feed-forward manner. Fibroblasts engage in bidirectional with immune cells during , secreting to recruit immune cells and enhance pro-fibrotic responses. This recruitment sustains a local inflammatory milieu, where macrophages in turn release additional TGF-β to reinforce fibroblast activation. Additionally, fibroblast-derived interleukin-6 (IL-6) amplifies by promoting T-cell and survival, creating a self-perpetuating loop that bridges acute responses to chronic . A hallmark example of fibroblast involvement in pathological fibrosis is (IPF), where aggregates of proliferating fibroblasts known as fibroblast foci represent sites of active deposition and alveolar collapse. These foci correlate with disease severity and progression, serving as histological markers of dysregulated repair in the lung interstitium. Therapeutic strategies targeting fibroblast activation in fibrosis increasingly focus on anti-TGF-β inhibitors, with several agents advancing in clinical trials post-2023 to mitigate overproduction. For instance, bexotegrast, an integrin-based TGF-β inhibitor, showed promising safety and reductions in fibrotic biomarkers in IPF patients during phase IIa trials in 2024, but development was discontinued in 2025 due to safety concerns in phase 2b. Isoform-selective inhibitors, such as those targeting TGF-β3, are also under evaluation for systemic sclerosis-associated , aiming to block pro-fibrotic signaling while sparing TGF-β isoforms.

Cancer and Tumor Microenvironment

Cancer-associated fibroblasts (CAFs) primarily originate from resident fibroblasts within the tumor stroma, which become activated in response to tumor-derived signals such as platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), fibroblast growth factor (FGF), epidermal growth factor (EGF), and hypoxia-induced factors. These resident cells, often quiescent, transdifferentiate into activated CAFs through pathways involving PDGF signaling, particularly in pancreatic and hepatic stellate cells converting to myofibroblast-like states. Additional origins include epithelial cells via epithelial-mesenchymal transition (EMT), endothelial cells via endothelial-mesenchymal transition (EndMT), bone marrow-derived mesenchymal stem cells, adipocytes, and monocytes, though local precursors predominate over circulating sources. In the , CAFs promote cancer progression by stiffening the (ECM) through excessive synthesis of collagens (types I, III, IV, and V) and , which fosters a fibrotic barrier that enhances tumor invasion and while impeding penetration and immune cell infiltration. They further support tumor growth by secreting hepatocyte growth factor (HGF), which stimulates tumor and motility, and (VEGF), which induces to sustain nutrient supply to hypoxic tumor regions. These functions are mediated by signaling cascades like IL-17a/JAK2/, contributing to stromal remodeling that favors . CAFs exhibit functional diversity, with prominent subtypes including inflammatory CAFs (iCAFs) and myofibroblastic CAFs (myCAFs), as delineated in recent reviews. iCAFs, characterized by high interleukin-1 receptor expression and low α-smooth muscle actin (α-SMA), are typically located distant from tumor cells and secrete pro-inflammatory cytokines such as CXCL-1 and IL-6 via and pathways, recruiting myeloid-derived suppressor cells (MDSCs), M2 macrophages, and regulatory T cells to suppress antitumor immunity. In contrast, myCAFs express high α-SMA, reside proximal to tumor cells, and respond to TGF-β by producing dense components like , forming physical barriers that promote therapeutic resistance and correlate with poor prognosis, though their depletion can sometimes accelerate . Single-cell RNA sequencing (scRNA-seq) studies have revealed extensive heterogeneity and across cancer types, identifying up to nine subtypes such as matrix CAFs (mCAFs), inflammatory CAFs (iCAFs), and tumor-like CAFs (tCAFs) in , , pancreatic, and other tumors, with phenotypes forming a rather than states. This enables CAFs to transition between subtypes in response to microenvironmental cues, influencing tumor progression and therapy response. Prognostic markers like fibroblast activation protein () expression, which labels myofibroblast-like CAFs involved in remodeling, are associated with aggressive disease and chemoresistance, while CD10 () in tCAFs predicts poor outcomes in non-small cell . Therapeutic strategies targeting CAFs, particularly FAP-expressing populations, include inhibitors, antibodies, CAR-T cells, and radioligands, with several in ongoing clinical trials as of 2025. For instance, 177Lu-FAPI radioligand has demonstrated an 83% disease control rate in phase I trials for and other adenocarcinomas, showing safety and efficacy in reducing tumor burden. Other agents like talabostat and BXCL701 are being evaluated in phase II trials for colorectal, pancreatic, and cancers, often combined with such as to overcome resistance, though challenges like CAF heterogeneity and suboptimal monotherapy efficacy persist. No FAP inhibitor has achieved full approval, but combination approaches hold promise for modulating the tumor .

Applications in Research and Medicine

Feeder Cells in

The use of fibroblasts as feeder cells in originated with the establishment of mouse embryonic (ES) cell lines, where mouse embryonic fibroblasts (MEFs) were employed as supportive layers to maintain pluripotency. In 1981, Evans and Kaufman derived pluripotential cells from mouse embryos by culturing them on MEF feeder layers, enabling indefinite propagation without . This approach has since become foundational for ES cell and induced pluripotent (iPS) cell maintenance, particularly in protocols involving . MEFs support ES cell pluripotency through both soluble and contact-dependent mechanisms. They secrete extracellular factors, including (LIF), which activates the JAK-STAT3 pathway to promote self-renewal and inhibit differentiation. Additional secreted factors such as fibroblast growth factor 2 (FGF2), Gremlin 1, and activins contribute to this maintenance. Contact-dependent signaling involves membrane-bound ligands that facilitate juxtacrine interactions, such as those mediated by and receptors, alongside extracellular matrix components like that enhance and prevent differentiation. To prepare MEF feeder layers, primary fibroblasts are isolated from day 13-14 mouse embryos and expanded in culture before inactivation to halt their proliferation while preserving supportive functions. Mitomycin C treatment is a standard method, typically at 10 μg/mL for 2-3 hours, which cross-links DNA and arrests cell division without disrupting secretory or adhesive capabilities. Gamma-irradiation serves as an alternative inactivation approach. Inactivated MEFs are then plated at densities of approximately 2-5 × 10^4 cells/cm² on gelatin-coated surfaces to form a confluent monolayer for seeding ES cells. A key advantage of MEF feeder layers is their cost-effectiveness in providing a natural microenvironment that sustains ES cell pluripotency over multiple passages, often outperforming feeder-free conditions in maintaining undifferentiated morphology and colony formation. They eliminate the need for complex synthetic matrices or high concentrations of recombinant growth factors, making them accessible for laboratory-scale research. However, MEFs pose limitations due to their xenogeneic origin, raising risks of immunogenic responses, viral contamination, and species-specific in human applications. These concerns have driven the development of human-derived alternatives, such as or endometrial fibroblasts, which support and cell growth under xeno-free conditions while minimizing contamination risks.

Therapeutic Targeting and Engineering

Fibroblasts have emerged as key targets for therapeutic interventions due to their central roles in tissue , , and cancer progression. Strategies to engineer or modulate fibroblast function aim to restore regenerative capacity, reverse pathological states, and enhance treatment outcomes in various diseases. These approaches leverage genetic editing, chemical modulation, and scaffolds to reprogram or deplete aberrant fibroblasts, offering potential for . Recent advances emphasize precision targeting to minimize off-target effects while maximizing therapeutic efficacy. CRISPR-based editing has facilitated the of fibroblasts into induced pluripotent cells (iPSCs) or other lineages, bypassing traditional methods and reducing risks. For instance, genome-scale CRISPR-Cas9 screens have identified barriers to pluripotency, enabling efficient derivation of iPSCs from somatic cells like fibroblasts by targeting regulators such as USP22. Advances in scaffold-based CRISPR delivery, including non- systems using biocompatible hydrogels and nanofibers, have improved gene editing efficiency in three-dimensional environments that mimic cues. These engineered fibroblasts hold promise for autologous cell therapies in , with applications in generating patient-specific tissues for transplantation. Rejuvenation of senescent fibroblasts represents a promising anti-aging , particularly through small-molecule interventions that reverse age-related dysfunction without full . Partial chemical using cocktails of small molecules, such as the 7c , has rejuvenated fibroblasts by improving mitochondrial , reducing aging metabolites, and resetting transcriptomic and epigenomic clocks, distinct from transcription factor-based methods. In human dermal fibroblasts, partial using Yamanaka factors has reversed mesenchymal drift associated with aging, restoring youthful profiles and enhancing proliferative capacity, as demonstrated in 2025 research. These interventions, which avoid genetic alterations, offer safer applicability for treating age-related tissue decline and improving outcomes. In cancer, targeting cancer-associated fibroblasts (CAFs) via depletion therapies seeks to disrupt the tumor-supportive microenvironment. Bispecific antibodies directed against CAF markers like fibroblast activation protein () have entered early clinical trials by 2025, aiming to selectively eliminate CAFs while sparing healthy fibroblasts and enhancing immune infiltration. For example, FAP-targeted immunotherapies, including bispecific constructs linking tumor antigens to immune effectors, have shown preclinical efficacy in reducing CAF-mediated in solid tumors. Ongoing I/II trials evaluate these agents in combination with checkpoint inhibitors, reporting preliminary tolerability and tumor regression in subsets of patients with various solid tumors, including pancreatic and breast cancers, as of mid-2025. Regenerative applications utilize fibroblast sheets to accelerate closure and tissue integration, particularly in injuries where donor sites are limited. Autologous fibroblast sheets, expanded via micrografting techniques like modified MEEK, have been clinically applied to cover extensive , promoting vascularization and reducing compared to traditional split-thickness grafts. transfer-generated skin grafts incorporating fibroblasts have demonstrated improved in porcine models and early human trials. These sheet-based therapies, often combined with dermal scaffolds, have achieved over 80% graft take rates in severe patients, minimizing risks and hospitalization duration. Recent advances in anti-fibrotic therapies focus on inhibiting mechanotransduction pathways in activated fibroblasts to halt excessive deposition. According to a comprehensive review, targeting /TAZ signaling or channels disrupts fibroblast-to-myofibroblast differentiation in response to stiff matrices, as seen in where stiffness can reach 10-30 kPa or higher. Small-molecule inhibitors of RhoA/ROCK and TGFβ/Smad pathways, such as losartan, have shown efficacy in preclinical models of renal and hepatic , with phase II trials confirming reduced synthesis. These mechanosensitive targets offer disease-modifying potential, with emerging combinations integrating inhibitors to enhance resolution in chronic fibrotic conditions.

References

  1. [1]
    Fibroblast - National Human Genome Research Institute
    A fibroblast is a type of cell that contributes to the formation of connective tissue, a fibrous cellular material that supports and connects other tissues ...
  2. [2]
    Fibroblasts: origins, definitions, and functions in health and disease
    Fibroblasts are diverse mesenchymal cells that participate in tissue homeostasis and disease by producing complex extracellular matrix and creating signaling ...
  3. [3]
    The biology and function of fibroblasts in cancer - Nature
    Aug 23, 2016 · Fibroblasts constitute a dominant cellular component of the tumour microenvironment (TME). The TME is composed of immune cell infiltrates, ...Missing: definition | Show results with:definition
  4. [4]
    Histology, Fibroblast - StatPearls - NCBI Bookshelf
    The fibroblast is one of the most abundant cell types present in the stroma. It has a variety of functions and composes the basic framework for tissues and ...
  5. [5]
    Guidelines and definitions for research on epithelial–mesenchymal ...
    Apr 16, 2020 · By contrast, mesenchymal cells do not contain functional epithelial junctions and present a back–front polarity in their actin stress fibres.
  6. [6]
    EM 006 Fibroblast - Histology Guide
    Transmission Electron Microscopy. Ultrathin section of a fibroblast as seen by transmission electron microscope (TEM). Examine the ultrastructure of this ...
  7. [7]
    Fibroblast: Histological structure and function | Kenhub
    The fibroblast is the most abundant cell of the connective tissue. It appears a fusiform, spindle-shaped cell with an oval euchromatic nucleus.
  8. [8]
  9. [9]
    Vimentin, the 57 000 molecular weight protein of fibroblast ... - PubMed
    Vimentin, the 57 000 molecular weight protein of fibroblast filaments, is the major cytoskeletal component in immature glia. Eur J Cell Biol. 1981 Jun;24(2): ...
  10. [10]
    Vimentin intermediate filament assembly regulates fibroblast ...
    Apr 4, 2019 · In this study, we demonstrate that inhibition of vimentin intermediate filaments (VimIFs) decreases the invasiveness of IPF fibroblasts and confers protection ...
  11. [11]
    Fibrocytes in health and disease - PMC - PubMed Central
    Fibrocytes, a group of bone marrow-derived mesenchymal progenitor cells, were first described in 1994 as fibroblast-like, peripheral blood cells that migrate ...
  12. [12]
    Fibrocytes: A Critical Review and Practical Guide - Frontiers
    Fibrocytes are hematopoietic-derived cells that directly contribute to tissue fibrosis by producing collagen following injury, during disease, and with aging.
  13. [13]
    Fibrocytes, Wound Healing, and Corneal Fibrosis - IOVS
    Results: Peripheral blood fibroblast-like cells, called fibrocytes, are primarily generated as mature collagen-producing cells in the bone marrow. They are ...
  14. [14]
    Circulating Fibrocytes Define a New Leukocyte Subpopulation That ...
    Nov 1, 1994 · Circulating Fibrocytes Define a New Leukocyte Subpopulation That Mediates Tissue Repair. Richard Bucala,; Lori A. Spiegel, ...<|separator|>
  15. [15]
    Fibrocytes in health and disease | Fibrogenesis & Tissue Repair
    Jun 6, 2012 · Fibrocytes comprise approximately 0.5% of non-erythrocytic cells in the peripheral blood, and they differentiate from CD14+ cells in culture ...Missing: prevalence | Show results with:prevalence
  16. [16]
    Elevated Circulating Fibrocytes Is a Marker of Left Atrial Fibrosis and ...
    Mar 13, 2018 · Circulating fibrocyte percentage positively associated with LA low‐voltage area in the persistent AF group, and circulating fibrocyte (≥4.05%) ...
  17. [17]
    The peripheral blood fibrocyte is a potent antigen-presenting cell ...
    Mouse fibrocytes share many of the same phenotypic properties with human fibrocytes and express both type I collagen and CD34 (4). By flow cytometry ...Materials And Methods · Fibrocyte Isolation · Results
  18. [18]
    The emerging role of fibrocytes in ocular disorders
    Apr 13, 2018 · In fact, active fibroblast morphology is different from fibrocytes. Fibroblasts have a branched cytoplasm surrounding an elliptical, speckled ...
  19. [19]
    Circulating fibrocytes: collagen-secreting cells of the peripheral blood
    For example, fibrocytes derived from wound chambers, which likely represent an inflammatory milieu, maintain CD34+ expression over time (Bucala et al., 1994).
  20. [20]
    The role of circulating fibrocytes in fibrosis
    Fibrocytes are cells that circulate in the peripheral blood and produce connective tissue proteins such as vimentin and collagens I and III.<|control11|><|separator|>
  21. [21]
  22. [22]
    Fibroblast progenitor cells of the embryonic chick limb | Development
    A population of mesenchymal cells derived from the stage-25 chick wing tip gives rise to progeny of a similar morphology and to authentic fibroblasts when ...
  23. [23]
    Cardiac and perivascular myofibroblasts, matrifibrocytes, and ...
    Matrifibrocytes and fibrocytes are two major subtypes identified as contributing to perivascular fibrosis and vascular inflammation. The major collagen ...
  24. [24]
    Cardiac Fibroblasts and Myocardial Regeneration - Frontiers
    Mar 24, 2021 · These CFs, termed as matrifibrocytes, highly express bone and cartilage-related ECM genes like Chad, Cilp2 and Comp, which are common gene ...
  25. [25]
    Mechanisms of cytokine production by fibroblasts-implications for ...
    Fibroblasts actively participate in cellular immune responses in connective tissues, when activated by signals abundant at inflammatory sites, i.e. cytokines
  26. [26]
    Essential immune functions of fibroblasts in innate host defense
    In response to innate immune stimulation, barrier tissue fibroblasts have been shown to secrete several proinflammatory cytokines and chemokines. In 1996, Noso ...Introduction · Fibroblasts respond to cytokines · The immune effector functions...
  27. [27]
    A single-cell and spatial genomics atlas of human skin fibroblasts ...
    Sep 24, 2025 · Single-cell RNA-seq reveals fibroblast heterogeneity and increased mesenchymal fibroblasts in human fibrotic skin diseases. Article Open ...
  28. [28]
  29. [29]
    Transcription Factor 21 Regulates Cardiac Myofibroblast Formation ...
    Dec 4, 2024 · TCF21 is expressed in tissue-resident fibroblasts and is downregulated in response to injury or stimuli leading to myofibroblast differentiation.Missing: dermal | Show results with:dermal
  30. [30]
    Epigenetic Regulation of Cancer-Associated Fibroblast Heterogeneity
    Here, we review epigenetic changes in CAF maturation, focusing on DNA methylation and histone modifications.
  31. [31]
    DNA Methylation of Fibroblast Phenotypes and Contributions ... - NIH
    Aug 3, 2021 · DNA methylation has been associated with different lung fibroblast populations in health and with alterations in lung disease, but to varying extents.
  32. [32]
    Heterogeneous response to TGF-β1/3 isoforms in fibroblasts of ...
    Sep 14, 2023 · In summary, our study has implications for comprehending the role of TGF-β signaling in fibroblast biology, fibrotic diseases, and cancer.
  33. [33]
    Differential Response of Cardiac Fibroblasts From Young Adult and ...
    In cardiac fibroblasts from young adult rats, ANG II significantly enhanced AT(1) mRNA levels, net collagen production, and TGF-beta production. In fibroblasts ...
  34. [34]
    Biochemistry, Collagen Synthesis - StatPearls - NCBI Bookshelf - NIH
    Collagen synthesis occurs in fibroblasts, both intracellularly and extracellularly, involving post-translational modifications, forming a triple helix, and ...
  35. [35]
    The Role of the Lysyl Oxidases in Tissue Repair and Remodeling
    Lysyl oxidase (LOX) and four lysyl oxidase-like proteins are a group of enzymes capable of catalyzing cross-linking reaction of collagen and elastin.
  36. [36]
    The small proteoglycan decorin supports adhesion and activation of ...
    Decorin is a small leucine-rich proteoglycan able to interact with several molecules of the subendothelial matrix, such as collagen and fibronectin.
  37. [37]
    Identification of extracellular matrix proteins secreted by human ...
    Mar 23, 2021 · Fibroblasts are intimately linked to the ECM and to a large extent, they are responsible for the synthesis of the fibrillar constituents of the ...
  38. [38]
    Fibroblasts in fibrosis: novel roles and mediators - Frontiers
    It acts on the mesenchyme and fibroblasts to induce proliferation, differentiation, and ECM production (Scotton and Chambers, 2007). PDGF is important for wound ...
  39. [39]
    Functions of exogenous FGF signals in regulation of fibroblast ... - NIH
    Sep 14, 2022 · Here we summarize the effects of FGFs in fibroblast to myofibroblast differentiation and ECM protein expression and discuss the underlying potential regulatory ...4. Roles Of Fgfs In... · 5. Fgf Signalling In... · 5.2. Fgf Signalling...
  40. [40]
    Biological role of matrix metalloproteinases: a critical balance
    Collectively, MMPs are able to cleave every ECM component [42]. However, different ECM components are susceptible to proteolysis by different MMPs and not all ...
  41. [41]
    Collagen I and the fibroblast: High protein expression requires a ...
    This review focuses on fibroblasts that make high levels of procollagen (type I) and how they regulate the collagen pathway.
  42. [42]
    Transition from inflammation to proliferation: a critical step during ...
    Fibroblasts play a central role in the formation of the granulation tissue, which migrate mainly from the nearby dermis to the wound in response to ...Missing: seminal | Show results with:seminal
  43. [43]
    Cellular and molecular mechanisms of repair in acute and chronic ...
    (c) Local and blood‐borne fibroblasts proliferate and migrate to form the wound granulation tissue, provide structure and signalling cues and deposit new ...Wound Granulation Tissue And... · Inflammation Is Good And Bad... · Wound Angiogenesis And...
  44. [44]
    Fibroblasts and myofibroblasts in wound healing - PMC
    Nov 6, 2014 · Notes: After damage, inflammation leads to the formation of the granulation tissue, during which myofibroblasts appear. An important ...Introduction To The... · Role In Wound Healing · Role Of Myofibroblasts In...Missing: seminal | Show results with:seminal
  45. [45]
    Akt1 Mediates α-Smooth Muscle Actin Expression and Myofibroblast ...
    Myofibroblast (MF) differentiation, marked by the de novo expression of smooth muscle α-actin (αSMA) stress fibers, plays a central role in wound healing ...Missing: myosin closure
  46. [46]
    Regulation of α-Smooth Muscle Actin Expression in Granulation ...
    Myofibroblasts are specialized contractile fibroblasts that are critical in wound closure and tissue contracture. Generation of contractile force is ...
  47. [47]
    Alpha-Smooth Muscle Actin Expression Upregulates Fibroblast ...
    Oct 13, 2017 · Our results indicate that an increased α-SMA expression is sufficient to enhance fibroblast contractile activity.
  48. [48]
    Formation and Function of the Myofibroblast during Tissue Repair
    Mechanically preventing wound closure by splinting the edges of experimental wounds accelerates expression of α-SMA compared with normally healing wounds; ...
  49. [49]
    Growth factors and cytokines in wound healing - Barrientos - 2008
    Sep 3, 2008 · This review will focus on the specific roles of these growth factors and cytokines during the wound healing process.Transforming Growth... · Platelet Derived Growth... · Chemokines
  50. [50]
    Critical Role of Transforming Growth Factor Beta in Different Phases ...
    In this review, we discuss (1) the effect of TGF-β on recruitment of inflammatory cells into the wound area, (2) fibroblast and keratinocyte migration, (3) ...Roles Of Tgf-β In Wound... · The Tgf-β Superfamily And... · The Role Of Tgf-β2 And -β3...Missing: papers | Show results with:papers
  51. [51]
    Trem2 acts as a non‐classical receptor of interleukin‐4 to promote ...
    Sep 30, 2024 · These findings identify that the IL-4/Trem2 interaction plays a critical role in diabetic wound healing. ... fibroblast proliferation, migration ...<|separator|>
  52. [52]
    Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases
    During the resolution of wound healing, myofibroblasts can follow several different cell fates. Myofibroblasts can die via apoptosis, a process induced by ...
  53. [53]
    Fibromodulin selectively accelerates myofibroblast apoptosis in ...
    Apr 12, 2025 · Here, we find that fibromodulin induces myofibroblast apoptosis after wound closure to reduce scarring in small and large animal models.
  54. [54]
    Apoptosis during wound healing, fibrocontractive diseases and ...
    During excessive scarring (hypertrophic scar or fibrosis), it is conceivable that the process of apoptosis cannot take place. After experimental endothelial ...
  55. [55]
    Role of fibroblasts in wound healing and tissue remodeling on Earth ...
    It has been shown that myofibroblast apoptosis occurs when the wound closes and the tissue recovers, at least in part, its tensile strength, suggesting that ...
  56. [56]
    An on-chip wound healing assay fabricated by xurography ... - Nature
    Oct 1, 2020 · The overall average cell migration rate for HDFs was calculated to be 27 μm/h. Moreover, the cell migration within 20 h was also split in ...
  57. [57]
    Methods to study differences in cell mobility during skin wound ...
    May 24, 2016 · A monolayer scratch assay is used to study random fibroblast and endothelial cell migration in response to EGF and bFGF respectively and a ...2. Materials And Methods · 2.2. Fibroblast And... · 3. Results
  58. [58]
    Stimulation of Skin and Wound Fibroblast Migration by ... - NIH
    In this study we have used an in vitro scratch assay procedure incorporating labeled MSCs and fibroblasts derived from normal donors and chronic wound patients ...
  59. [59]
    TGF-β signaling in health, disease and therapeutics - Nature
    Mar 22, 2024 · TGF-β is found to induce JAK1 and JAK2 activation respectively in hepatic stellate cells (HSCs) and fibroblasts. In these cases, activated JAK ...
  60. [60]
    Transforming growth factor–β in tissue fibrosis
    Feb 20, 2020 · This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions,
  61. [61]
    Organ fibrosis: beyond collagen I expression. Fibroblast phenotype ...
    A summary of some recent advances in uncovering critical fibroblast activation states, ECM organization, and composition of fibrotic ECM, including basement ...Cardiac Fibrosis · Basement Membrane Proteins... · Collagen Iv In Fibrosis
  62. [62]
    ECM formation and degradation during fibrosis, repair, and ... - Nature
    Jun 10, 2025 · Here, we review our current understanding of the structure of the ECM, the mechanisms behind organ-specific fibrosis, resolution, healing and regeneration.Missing: matricellular | Show results with:matricellular
  63. [63]
    CXCL12+ dermal fibroblasts promote neutrophil recruitment and ...
    Feb 23, 2024 · These findings show that CXCL12+ dermal immune acting fibroblast subsets play a critical role in cutaneous neutrophil recruitment and host defense.Introduction · Results · Discussion · Materials and methods
  64. [64]
    Fibroblast-macrophage reciprocal interactions in health, fibrosis ...
    May 11, 2021 · Here, we review the current understanding of the molecular mechanisms that underlie fibroblast-macrophage interactions in health, fibrosis, and cancer.
  65. [65]
    IL-6 in the infarcted heart is preferentially formed by fibroblasts ... - JCI
    Mar 21, 2023 · We show that purinergic metabolic cooperation between CFs and T cells is a mechanism that modulates IL-6 formation by the heart and has therapeutic potential.
  66. [66]
    Three-dimensional characterization of fibroblast foci in idiopathic ...
    Apr 21, 2016 · In idiopathic pulmonary fibrosis (IPF), the fibroblast focus is a key histological feature representing active fibroproliferation.
  67. [67]
    Revisiting pulmonary fibrosis: inflammatory dynamics of ... - Frontiers
    The balance between inflammation and fibrosis appears to be a tightly regulated process, where inflammatory fibroblasts serve as a key intermediary linking ...
  68. [68]
    Bexotegrast in Patients with Idiopathic Pulmonary Fibrosis
    Mar 25, 2024 · Conclusions: Bexotegrast demonstrated a favorable safety and tolerability profile, up to 12 weeks for the doses studied. Exploratory analyses ...
  69. [69]
    Isoform-selective TGF-β3 inhibition for systemic sclerosis
    Feb 9, 2024 · TGF-β has been implicated as a central mediator of fibrogenesis, yet pan-TGF-β inhibition has led to severe adverse effects in clinical studies, ...Missing: post- | Show results with:post-
  70. [70]
    Cancer‐Associated Fibroblasts: Origin, Classification ...
    Oct 28, 2025 · This review explores the multifaceted roles of cancer‐associated fibroblasts (CAFs) in tumor biology. It examines their diverse origins, dynamic ...
  71. [71]
    Cancer-associated fibroblasts in cancer drug resistance ... - Nature
    Jul 24, 2025 · In this review, I focused on the functions of CAFs in the development of therapeutic resistance in cancers and the progression of cancer from ...Myofibroblastic Cafs... · Inflammatory Cafs (icafs) · Caf-Targeting Therapies
  72. [72]
    Cancer-associated fibroblast classification in single-cell and spatial ...
    Jul 18, 2023 · To define CAF phenotypes, we analyse a single-cell RNA sequencing (scRNA-seq) dataset of over 16,000 stromal cells from tumours of 14 breast ...
  73. [73]
    Cancer-associated fibroblasts as therapeutic targets for cancer
    Jan 9, 2025 · In this review, we provide a summary of markers for identifying CAFs and their subtypes. Importantly, we conduct a comprehensive review of both ...
  74. [74]
    Establishment in culture of pluripotential cells from mouse embryos
    Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981 Jul 9;292(5819):154-6. doi: 10.1038/292154a0. Authors. M J Evans, M H Kaufman.
  75. [75]
    Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult ...
    Dec 26, 2004 · Here, we dem- onstrate induction of pluripotent stem cells from mouse embryonic or adult fibroblasts by introducing four factors, Oct3/4, Sox2, ...
  76. [76]
    LIF signal in mouse embryonic stem cells - PMC - PubMed Central
    The dependency on exogenous LIF for the maintenance of pluripotency is obvious in feeder-free culture but it is somewhat obscure in the presence of MEF-feeder ...
  77. [77]
    Feeder Layer Cell Actions and Applications - PMC - NIH
    Feeder cells consist in a layer of cells unable to divide, which provides extracellular secretions to help another cell to proliferate.
  78. [78]
    Mouse Embryonic Fibroblasts (MEF Feeder Cells)
    ### Summary of MEF Feeder Cell Preparation and Inactivation
  79. [79]
    ATCC Stem Cell Culture Guide
    The drawbacks with using feeders are 1) it can be more labor intensive, 2) it may pose a risk of transmitting animal pathogens to human stem cells if MEFs are ...
  80. [80]
    Toward xeno-free culture of human embryonic stem cells - PMC
    In this review, we describe various methods tested to culture cells in the absence of MEF feeder layers and other advances in eliminating xenogeneic products ...Missing: xenogenic | Show results with:xenogenic