You are on page 1of 6

The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671

Cells in focus

Fibroblasts and myofibroblasts: Their source,


function and role in disease
Robin J. McAnulty ∗
Centre for Respiratory Research, University College London, Rayne Building, 5 University Street,
London, WC1E 6JJ, United Kingdom
Received 2 October 2006; received in revised form 9 November 2006; accepted 10 November 2006
Available online 23 November 2006

Abstract
Fibroblasts are found in most tissues of the body. They exhibit several phenotypes including non-contractile fibroblasts, con-
tractile myofibroblasts, and intermediate phenotypes including the protomyofibroblast. Fibroblasts are metabolically active cells
which play critical roles regulating extracellular matrices, interstitial fluid volume and pressure, and wound healing. Fibroblast
numbers can be maintained or expanded by proliferation of resident populations but in addition, recent evidence indicates they
can also be derived through epithelial-mesenchymal transition or from circulating and tissue-derived mesenchymal stem cells.
Many diseases are associated with dysregulation of the injury repair response and fibroblast function, leading to increased or
decreased deposition of extracellular matrix proteins, altered tissue architecture, impaired function and in some cases signifi-
cant morbidity and mortality. There are currently no specific therapies that target fibroblast-associated pathology but increasing
knowledge of pathological mechanisms has led to development of new agents providing hope for improved treatment of these
diseases.
© 2006 Elsevier Ltd. All rights reserved.

Cell facts

• Transdifferentiates between non-contractile and contractile phenotypes


• Major cell involved in the synthesis of soft tissue extracellular matrix proteins
• Capable of producing 3.5 million molecules of collagen per cell per day
• Major producer of MMPs capable of degrading extracellular matrix
• Important role in regulating tissue hydration/osmotic pressure
• Contraction of wounds via generation of intracellular contractile forces

Keywords: Fibroblast; Myofibroblast; Mesenchymal cells; Epithelial-mesenchymal transition; Fibrocyte; Stem cells; Extracellular matrix; Fibrosis

1. Introduction

Fibroblasts are spindle shaped cells found in the


majority of tissues and organs of the body associated
with extracellular matrix (ECM) molecules. Characteris-
∗ Tel.: +44 207 679 6971; fax: +44 207 679 6973.
tic features include expression of vimentin in the absence
E-mail address: r.mcanulty@ucl.ac.uk. of desmin and ␣-smooth muscle actin. When activated,

1357-2725/$ – see front matter © 2006 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biocel.2006.11.005
R.J. McAnulty / The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671 667

fibroblasts exhibit an abundant endoplasmic reticulum implications for therapy in wound healing, cancer and
and prominent Golgi associated with the synthesis and fibrosis.
secretion of ECM molecules including collagens, pro-
teoglycans and fibronectin, as well as, proteases capable 2.1. Epithelial-mesenchymal transition
of degrading the ECM. Cytoskeletal proteins in associa-
tion with cell surface integrins and the ECM facilitate cell EMT is widely believed to occur during development
motility and the generation of contractile forces impor- and in cancer progression, however its role in the tissue
tant in tissue homeostasis and wound healing. response to epithelial stress or injury, at least in vivo,
is more controversial (reviewed in Zavadil & Bottinger,
2. Cell origin and plasticity 2005). The sequence of molecular events involved in
EMT has recently been extensively reviewed by Zavadil
Fibroblasts are embryologically of mesenchymal ori- and Bottinger (2005). Briefly, EMT requires the organ-
gin with a spectrum of phenotypic entities ranging ised dedifferentiation of epithelial cells with loss of
from the non-contractile fibroblast to the contractile polarity, adherans and tight junctions through downreg-
myofibroblast with a number of intermediate pheno- ulation of proteins associated with the maintenance of
types having been described (reviewed in Eyden, 2005) these structures including ZO1, cadherins and desmo-
including that of the protomyofibroblast (Desmouliere, plakin through the upregulation of the Snail/Slug family
Darby, & Gabbiani, 2003). In addition to the fea- of transcriptional repressors and switching from epithe-
tures of active fibroblasts, prototypical myofibroblasts lial ␣6␤4 to mesenchymal ␣5␤1 integrin expression.
are distinguished by the presence of ␣-smooth mus- This leads to the release of non-polarized epithelial
cle actin containing stress fibres, linked in a linear cells with a remodelled mesenchymal stress fibre pat-
fashion through trans-membrane fibronexus junctions tern of actin localisation rather than the epithelial cortical
to protruding filamentous fibronectin fibres, increased pattern of cytoskeleton. To allow transitioning cells to
expression of ED-A fibronectin and gap junctions (for migrate into the interstitium, metalloproteinases (MMP),
extensive reviews see Desmouliere et al., 2003; Eyden, such as MMP2 and MMP9 are induced to digest the base-
2005). Myofibroblasts are further distinguished from ment membrane. A number of extracellular stimuli have
smooth muscle cells by their general lack of smooth been shown to be involved in the induction and pro-
muscle markers including desmin and smooth muscle gression of EMT including, TGF-␤, FGF-2, EGF and
myosin. Myofibroblasts may arise from the transdif- IGF-II (reviewed in Kalluri & Neilson, 2003). Whilst
ferentiation of fibroblasts and smooth muscle cells. EMT has clearly been demonstrated to occur in vitro, it
However, whether myofibroblast-like cells derived from has been more difficult to prove in vivo. Until recently, in
fibroblasts and smooth muscle cells form similar or dis- vivo evidence for EMT has relied on the demonstration
tinct phenotypic populations is debatable and whether of loss and gain of epithelial and mesenchymal mark-
fibroblasts can differentiate into smooth muscle cells and ers, respectively, in transitioning cells. Whilst there are
vice versa is uncertain, although recent studies suggest good markers for epithelial dedifferentiation (e.g. ZO-1,
that fibroblasts can differentiate into myofibroblast-like E-cadherin, desmoplakins, cytokeratin 18 and MUC1),
cells with induction of protein expression patterns pre- fibroblast markers, such as vimentin, FSP1 and ␣-smooth
viously thought to be characteristic of smooth muscle muscle actin are less specific, also being expressed in
cells (Chambers, Leoni, Kaminski, Laurent, & Heller, other cells types or only expressed in a sub-population
2003). Traditionally it was thought that replacement of fibroblasts/myofibroblasts. However, recent studies in
or expansion of fibroblast/myofibroblast populations the lungs of mice where ␤-galactosidase was specifically
homeostatically and in wound healing or disease settings expressed in epithelial cells has provided strong evidence
was from resident tissue populations of cells. However, of transitioning of these cells into fibroblasts during the
over the last 10–15 years mounting evidence suggests development of lung fibrosis and that these cells repre-
that fibroblasts/myofibroblasts, at least following injury sented the majority of the increased number of fibroblasts
and in fibrotic disease, may be derived from a variety in the lung, suggesting that EMT is an important source
of sources. These include dedifferentiation of epithe- of fibroblasts, at least in the lung (Kim et al., 2006).
lial cells by a process known as epithelial-mesenchymal
transition (EMT), as well as, bone marrow- and tissue- 2.2. Bone marrow-derived stem cells
derived mesenchymal stem cells (Fig. 1). However, the
relative contributions of each of these sources are cur- A number of studies have demonstrated the poten-
rently a topic of intense debate due to the potential tial for bone marrow-derived circulating fibrocytes to
668 R.J. McAnulty / The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671

Fig. 1. The source and function of fibroblasts in normal and pathological states. Origin and plasticity: Fibroblasts exist as several morphological
phenotypes ranging from the extremes of the non-contractile fibroblast to the ␣-smooth muscle actin stress fibre containing contractile myofibroblast
together with an intermediate phenotype which has been termed the protomyofibroblast. Fibroblasts can transdifferentiate into myofibroblasts and
there is some evidence to suggest the process may be reversible to at least some extent. Fibroblast populations can be maintained or expanded
by proliferation of existing populations, or derived from epithelial-mesenchymal transition, circulating bone marrow-derived fibrocytes or from
tissue-derived stem cells. There is also evidence that fibroblasts can undergo mesenchymal-epithelial transition and it has recently been shown that
genetic programmes can be induced in fibroblasts to convert them into pluripotent stem cells. Function: Major functions of fibroblasts/myofibroblasts
include: synthesis and degradation of the multitude of glycoproteins which make up the specialised extracellular matrices of tissues and organs of the
body which contribute to their specific functions; regulation through cell–matrix interactions of interstitial fluid volume, pressure and appropriate
levels of tissue contraction for optimum function; playing a critical role in wound healing through cell–cell and cell–matrix interactions, production
and response to mediators, modulation of extracellular matrix metabolism, wound contraction and scar resolution. Pathology: Dysregulated or
inappropriate fibroblast function is associated with pathologies in which diminished or excess extracellular matrix deposition, or inappropriate tissue
contraction is a feature. Such conditions affect almost all tissues and organs of the body, examples of which are included in the figure.
R.J. McAnulty / The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671 669

enter tissues following injury and contribute to wound 3. Functions


healing and pathological scarring (Abe, Donnelly, Peng,
Bucala, & Metz, 2001; Bucala, Spiegel, Chesney, Hogan, One of the major functions of fibroblasts is the pro-
& Cerami, 1994; Quan, Cowper, Wu, Bockenstedt, & duction and homeostatic maintenance of the ECM of the
Bucala, 2004). Fibrocytes represent approximately 0.5% tissue or organ in which they reside. They are metabol-
of the peripheral blood leukocyte population that have ically highly active cells, being capable of synthesising
an adherant spindle shape morphology in culture. They and secreting most ECM components, including col-
express markers of haematopoietic cells (CD34), leuko- lagens, proteoglycans, fibronectin, tenascin, laminin
cytes (CD11b, CD13 and CD45) as well as fibroblast and fibronectin. Fibroblasts continually synthesise ECM
products (collagens I, III, and fibronectin) and are dis- proteins and it has been estimated that each cell
tinguished from monocytes/macrophages, dendritic cells can synthesise approximately 3.5 million procollagen
and B cells by their lack of specific markers for these cell molecules/cell/day (McAnulty, Campa, Cambrey, &
types. They appear to be derived from the differentia- Laurent, 1991). However, the amount they secrete is
tion of CD14+ peripheral blood mononuclear cells (Yang regulated by lysosomal enzymes, such as cathepsins B,
et al., 2002) largely under the control of TGF-␤ which D and L, with between 10% and 90% of all procolla-
together with endothelin-1 also stimulates proliferation gen molecules being degraded intracellularly prior to
and differentiation of fibrocytes to myofibroblast-like ␣- secretion, depending on tissue and age. Regulation of
smooth muscle actin positive cells (Li & Huard, 2002; this process appears to provide a mechanism for rapid
Schmidt, Sun, Stacey, Mori, & Mattoli, 2003; Yang et adaptation of the amount of collagen secreted following
al., 2002). However, this does not appear to be the case injury (McAnulty & Laurent, 1995). In addition, fibrob-
in all situations as Hashimoto, Jin, Liu, Chensue, & Phan lasts produce matrix metalloproteinases (MMP) and
(2004) found that bone marrow-derived lung fibroblasts their inhibitors, tissue inhibitors of metalloproteinase
were unresponsive to TGF-␤ and failed to differentiate (TIMP), which regulate extracellular degradation of the
into myofibroblasts. The contribution of bone marrow- ECM. Fibroblast ECM metabolism is regulated by com-
derived stem cells to increased fibroblast/myofibroblast plex mechanisms including cell–cell and cell–matrix
number during wound healing and fibrosis is uncertain interactions, as well as a multitude of stimulatory and
with estimates ranging from a few percent up to approx- inhibitory mediators, which may be present in their
imately 80%. local environment (McAnulty & Laurent, 2002). These
include mediators derived from other resident cells in the
2.3. Tissue-derived mesenchymal stem cells local environment, fibroblasts themselves and mediators
derived from the circulation or infiltrating inflammatory
Studies in several tissues have also suggested that cells.
mesenchymal stem cells reside in tissues (Gharzi, Fibroblasts also play an important role in regulating
Reynolds, & Jahoda, 2003; Li & Huard, 2002) and one tissue interstitial fluid volume and pressure by interac-
recent extensive study suggests they may be localised to tion of ␤1 integrin receptors which anchor them to ECM
vessel walls (da Silva Mereilles, Chagastelles, & Nardi, proteins, and particularly the collagen- and laminin-
2006). These cells lack haematopoietic and leukocyte binding ␣2␤1 integrin via intracellular forces generated
markers but express ␣SMA, a marker associated with through the cytoskeleton (Wiig, Rubin, & Reed, 2003).
myofibroblasts and smooth muscle cells. Furthermore, In vitro modelling of these cell–matrix interactions indi-
previous studies in the lung have suggested that cate that these processes can be modulated by PDGF
following injury myofibroblasts may originate from and endothelin which enhance contraction and IL-1
perivascular and peribronchial sources (Zhang, Rekhter, and TNF-␣ which reduce contraction. Consistent with
Gordon, & Phan, 1994). Whether these cells derive this, interstitial fluid pressure has been shown to be
from a population of mesenchymal stem cells similar increased in vivo by PDGF-BB and ␣-trinositol, and
to those described by Mereilles and co-workers is not decreased by TNF-␣, nicotinomide and dexametha-
known. sone.
Together these findings demonstrate a high degree Following tissue injury fibroblasts/myofibroblasts
of plasticity and a diversity of origins of fibrob- play a central role in wound healing and repair (reviewed
last/myofibroblast populations. This diversity may also in Desmouliere et al., 2003). The initial processes follow-
begin to explain the heterogeneous phenotypic character- ing injury include clot formation and platelet degranu-
istics of fibroblast/myofibroblast populations observed lation, releasing mediators to attract inflammatory cells
both between tissues and within tissues. to the wound site which produce additional mediators
670 R.J. McAnulty / The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671

involved in the recruitment of fibroblastic cells derived peutic strategies primarily revolve around the use of anti-
from several potential sources as described above. The inflammatory corticosteroids, immunosuppressants and
fibroblastic cells present in this early phase are highly treatment of tissue or disease specific symptoms. How-
active synthetically replacing the provisional matrix with ever, at best these have limited therapeutic effects. Over
a more mature ECM including collagens and fibronectin the last 30 years a number of approaches to modulate
under the control of mediators produced by inflamma- extracellular matrix synthesis or degradation, including
tory cells, injured and regenerating epithelial cells, and use of prolyl-4-hydroxylase inhibitors to limit collagen
fibroblasts themselves. As granulation tissue deposition synthesis and TIMP inhibitors to promote ECM degra-
proceeds the fibroblasts develop characteristics of myofi- dation have been developed, although none have yet
broblasts, including the appearance of ␣-smooth muscle progressed the clinic. The rapid expansion of our knowl-
actin containing stress fibres. The appearance of these edge over the last 15–20 years of mediators involved in
myofibroblasts correlates with contraction and closure of the regulation of the normal injury repair process and
the wound through focal adhesions between myofibrob- in the pathogenesis of diseases associated with aberrant
lasts and the extracellular matrix. During the final phases repair has identified novel therapeutic targets and led
of remodelling and resolution the production of MMPs to the development of a number of specific inhibitors
and TIMPs by cells including fibroblasts changes from a for mediators or their receptors. These include small
balance favouring ECM deposition to a matrix degrading molecule antagonists and biological inhibitors target-
environment and myofibroblasts are removed by apopto- ing TGF␤, endothelin-1, interferon-␥, EGF, and IL-13
sis. Remaining fibroblasts exhibit a more quiescent, non- which are currently in or about to enter clinical trials.
contractile phenotype which may have reverted from a For example, inhibitors of TGF-␤ activity including neu-
myofibroblast phenotype or be derived from a source of tralizing antibodies and antisense vaccines are currently
cells not involved in differentiation into myofibroblasts. undergoing clinical trials and small molecule receptor
Dysregulation of the injury repair response may lead to antagonists are in pre-clinical development (reviewed in
ineffective or over exuberant and pathological wound Howell & McAnulty, 2006). These are likely to impact
healing. on a number of processes, including inhibition of the
conversion of fibrocytes to myofibroblasts, EMT, and
4. Associated pathologies modulation of ECM metabolism and deposition. In addi-
tion, the recent development of siRNA technologies may
Many diseases associated with diminished or excess allow for more rapid drug development with the potential
deposition of ECM are likely to be related to dysregula- to dramatically reduce the time from target identifica-
tion of the injury repair response and fibroblast function. tion to clinical trials. For example, siRNAs developed
In this context ‘injury’ is broad ranging including envi- for use in other disease settings by Sirna Therapeutics
ronmental, infectious, cancerous, traumatic/mechanical, (Sirna-027, which targets vascular endothelial growth
autoimmune and drug-induced insults. Thus, diseases factor receptor 1, VEGFR-1) and Alnylam (ALN-
in which fibroblasts, in their various phenotypic guises, RSV01, which targets respiratory syncytial virus) took
play a central role may affect almost all tissues and less than 2 years from development to Phase I clinical
organs of the body (illustrated in Fig. 1). Their impor- trials.
tance is further highlighted by the suggestion that almost There is therefore renewed optimism that increased
half of all deaths are associated with fibrosing conditions. knowledge of the pathogenesis of disease, which has led
Diseases associated with either increased or decreased to an influx of compounds in development or in clini-
ECM deposition, or contraction of tissues result in dis- cal trials across the spectrum of diseases associated with
torted tissue architecture, impaired function and in many aberrant fibroblast function, together with the potential
cases, particularly where the vital organs are involved, for more rapid development of drugs against newly iden-
significant morbidity and mortality. Dysregulation of tified targets, will lead to improved treatments in the not
several phases of the injury repair response, including too distant future.
chronic or repetitive injury, an inappropriate inflamma-
tory response, an altered balance of ECM metabolism
and deposition, altered phenotypic profiles or persis- Acknowledgements
tence of myofibroblasts contribute to aberrant tissue
repair. RJMs research is supported by grants from the
At present there are no treatments which specifically Wellcome Trust, British Lung Foundation and Asthma
target fibroblast-associated pathologies. Current thera- UK.
R.J. McAnulty / The International Journal of Biochemistry & Cell Biology 39 (2007) 666–671 671

References Li, Y., & Huard, J. (2002). Differentiation of muscle-derived cells into
myofibroblasts in injured skeletal muscle. American Journal of
Abe, R., Donnelly, S. C., Peng, T., Bucala, R., & Metz, C. N. (2001). Pathology, 161, 895–907.
Peripheral blood fibrocytes: Differentiation pathway and migration McAnulty, R. J., & Laurent, G. J. (1995). Pathogenesis of lung fibrosis
to wound sites. Journal of Immunology, 166, 7556–7562. and potential new therapeutic strategies. Experimental Nephrology,
Bucala, R., Spiegel, L. A., Chesney, J., Hogan, M., & Cerami, 3, 96–107.
A. (1994). Circulating fibrocytes define a new leukocyte sub- McAnulty, R. J., & Laurent, G. J. (2002). Fibroblasts. In P. Barnes, J.
population that mediates tissue repair. Molecular Medicine, 1, Drazen, S. Rennard, & N. Thomson (Eds.), Asthma and COPD:
71–81. Basic mechanisms and clinical management (pp. 139–144). Lon-
Chambers, R. C., Leoni, P., Kaminski, N., Laurent, G. J., & Heller, R. don: Academic Press.
A. (2003). Global expression profiling of fibroblast responses to McAnulty, R. J., Campa, J. S., Cambrey, A. D., & Laurent, G. J. (1991).
transforming growth factor-␤1 reveals the induction of inhibitor of The effect of transforming growth factor ␤ on rates of procollagen
differentiation-1 and provides evidence of smooth muscle cell phe- synthesis and degradation in vitro. Biochimica et Biophysica Acta,
notypic switching. American Journal of Pathology, 162, 533–546. 1091, 231–235.
da Silva Mereilles, L., Chagastelles, P. C., & Nardi, N. B. (2006). Metz, C. N. (2003). Fibrocytes: A unique cell population impli-
Mesenchymal stem cells reside in virtually all post-natal organs cated in wound healing. Cellular and Molecular Life Sciences,
and tissues. Journal of Cell Science, 119, 2204–2213. 60, 1342–1350.
Desmouliere, A., Darby, I. A., & Gabbiani, G. (2003). Normal and Quan, T. E., Cowper, S., Wu, S.-P., Bockenstedt, L. K., & Bucala,
pathologic soft tissue remodelling: Role of the myofibroblast, with R. (2004). Circulating fibrocytes: Collagen-secreting cells of the
special emphasis on liver and kidney fibrosis. Laboratory Investi- peripheral blood. International Journal of Biochemistry and Cell
gation, 83, 1689–1707. Biology, 36, 598–606.
Eyden, B. (2005). The myofibroblast: A study of normal, reactive and Schmidt, M., Sun, G., Stacey, M. A., Mori, L., & Mattoli, S. (2003).
neoplastic tissues, with an emphasis on ultrastructure. Part 1 – Identification of circulating fibrocytes as precursors of bronchial
Normal and reactive cells. Journal of Submicroscopic Cytology myofibroblasts in asthma. Journal of Immunology, 170, 380–
and Pathology, 37, 109–204. 389.
Gharzi, A., Reynolds, A. J., & Jahoda, C. A. B. (2003). Plasticity of hair Wiig, H., Rubin, K., & Reed, R. K. (2003). New and active role of
follicle dermal cells in wound healing and induction. Experimental interstitium in control of interstitial fluid pressure: Potential ther-
Dermatology, 12, 126–136. apeutic consequences. Acta Anaesthesiologica Scandinavica, 47,
Hashimoto, N., Jin, H., Liu, T., Chensue, S. W., & Phan, S. H. (2004). 111–121.
Bone marrow-derived progenitor cells in pulmonary fibrosis. Jour- Yang, L., Scott, P. G., Giuffre, J., Shankowsky, H. A., Ghahary, A.,
nal of Clinical Investigation, 113, 243–252. & Tredget, E. E. (2002). Peripheral blood fibrocytes from burn
Howell, J. E., & McAnulty, R. J. (2006). TGF-␤: Its role in asthma and patients: Identification and quantification of fibrocytes in adherent
therapeutic potential. Current Drug Targets, 7, 547–565. cells cultured from peripheral blood mononuclear cells. Laboratory
Kalluri, R., & Neilson, E. G. (2003). Epithelial-mesenchymal tran- Investigation, 82, 1183–1192.
sition and its implications for fibrosis. Journal of Clinical Zavadil, J., & Bottinger, E. P. (2005). TGF-␤ and epithelial-to-
Investigation, 112, 1776–1784. mesenchymal transitions. Oncogene, 24, 5764–5774.
Kim, K. K., Kugler, M. C., Wolters, P. J., Robillard, L., Galvez, M. Zhang, K., Rekhter, M. D., Gordon, D., & Phan, S. H. (1994).
G., Brumwell, A. N., Sheppard, D., & Chapman, H. A. (2006). Co-expression of ␣-smooth muscle actin and type I collagen
Alveolar epithelial cell mesenchymal transition develops in vivo in fibroblast-like cells of rat lungs with bleomycin-induced pul-
during pulmonary fibrosis and is regulated by the extracellular monary fibrosis: A combined immunohistochemical and in situ
matrix. Proceeding of the National Academy of Sciences USA, 103, hybridization study. American Journal of Pathology, 145, 114–
13180–13185. 125.

You might also like