<<

CMLS, Cell. Mol. Life Sci. 60 (2003) 1342–1350 1420-682X/03/071342-09 DOI 10.1007/s00018-003-2328-0 CMLS Cellular and Molecular Life Sciences © Birkhäuser Verlag, Basel, 2003

Review

Fibrocytes: a unique cell population implicated in

C. N. Metz North Shore-LIJ Research Institute, 350 Community Drive, Manhasset, New York 11030 (USA), Fax: + 1 516 365 5090, e-mail: [email protected]

Received 25 November 2002; received after revision 31 December 2002; accepted 16 January 2003

Abstract. Following tissue damage, host wound healing gin remains a mystery. A unique cell population, known ensues. This process requires an elaborate interplay be- as fibrocytes, has been identified and characterized. One tween numerous cell types which orchestrate a series of of the unique features of these blood-borne cells is their regulated and overlapping events. These events include ability to home to sites of tissue damage. This article re- the initiation of an antigen-specific host immune re- views the identification and characterization of fibro- sponse, blood vessel formation, as well as the production cytes, summarizes the potential role of fibrocytes in the of critical molecules, cytokines and numerous steps of the wound-healing process and high- growth factors which mediate tissue repair and wound lights the potential role of fibrocytes in fibrotic disease closure. are considered es- pathogenesis. sential for successful wound healing; however, their ori-

Key words. Tissue repair; ; tissue remodeling; TGFb; angiogenesis; antigen presentation.

What are fibrocytes? wounds and tissues originate from the circulation or The discovery, isolation and initial characterization from the surrounding tissue areas, the concept that fi- of fibrocytes broblast-like cells found within the wound originate from the peripheral blood dates back almost 100 years (re- Wounds or tissue injuries caused by trauma, burns, in- viewed in [1]). Fibroblasts found at sites of tissue injury flammation, infection, and metabolic deficiencies result may originate from different sources depending on the in the physical disruption of the normal cellular architec- wound/injury type. For example, in minor wounds fi- ture of the tissue. In response to tissue injury, the host broblasts may migrate from surrounding undamaged tis- commences a repair process that is regulated by cellular, sue, whereas fibrocytes may be recruited to deep tissue humoral and connective tissue mediators. The cell types wounds where they differentiate into fibroblasts. implicated in the repair of tissue injury include plate- The investigations by Bucala and colleagues in the early lets, monocytes/macrophages, T lymphocytes, endothe- 1990s that led to the discovery of ‘fibrocytes’ were based lial cells and connective tissue fibroblasts. Connective on the hypothesis that specialized ‘-like cells’ tissue fibroblasts found at the sites of tissue injury and in present in experimentally implanted wound chambers areas of tissue remodeling are believed to play an essen- originated from the circulation. This discovery led to the tial role in the healing process. Although it is unclear identification and initial characterization of a distinct whether these connective tissue fibroblasts found in population of blood-borne CD34+/Col I+ fibroblast-like cells that rapidly enter sites of tissue injury [2]. Termed * Corresponding author. fibrocytes, these cells comprise ~0.1–0.5% of nonery- CMLS, Cell. Mol. Life Sci. Vol. 60, 2003 Review Article 1343 throcytic cells in the peripheral blood (as determined by Table 1. Fibrocyte-associated surface markers. spot immunofluorescence staining using anti-CD34-rho- Marker type Reference damine and anti-col I-fluorescein 5 (6)-isothiocyanate (FITC) of blood cells following erythrocyte lysis) [2]. Fi- ECM markers brocytes can be isolated from buffy coats prepared from I (2–8)] blood and cultured ex vivo [2–4]. These crude fibrocyte Collagen III [2] Fibronectin [2] preparations obtained from human or mouse blood are Vimentin [2] grown in Dulbecco’s modified Eagle medium containing CD markers fetal calf serum, without the addition of other growth fac- CD11a (LFA-1) [2–3] tors. After 10–14 days of incubation, ex vivo cultured fi- CD11b (Mac 1) [2] brocytes display an adherent, spindle-shaped morphol- CD13 (pan myeloid antigen) [2] CD34 (hemopoetic stem cell antigen) [2] ogy. Fibrocytes are then purified from this crude prepara- CD45 (leukocyte common antigen) [2] tion (70–80% pure) following negative selection for CD54 (ICAM) [2] other immune cell types (B cells, T cells and monocytes) CD58 (LFA-3) [2] (see fig. 1). The resulting fibrocyte population (>95% pure CD80 (B7-1) [3] CD86 (B7-2) [2–3] based on collagen I and CD11b staining, or collagen I and + MHC-related markers CD34 staining) has been characterized based on expres- MHC class II [2–3] sion of (i) extracellular surface markers [including cluster HLA-DP [3] of differentiation (CD) antigens, major histocompatibility HLA-DQ [3] complex (MHC)-like molecules and extracellular matrix HLA-DR [3] protein markers] (table 1) and (ii) cytokine, chemokine and Chemokine receptors (ligands) CCR3 (MCP-3, MCP-5, RANTES, MIP1a, HCC-1) [4] growth factor expression patterns (table 2). CCR5 (MIP1a, MIP1b, RANTES) [4] Fibrocytes are a unique CD45+ cell population [2] They CCR7 (ELC, SLC) [4] are distinct from monocytes, (CD14–, esterase–, CD54–), CXCR4, fusin (SDF-1) [4] dendritic cells (CD10–, CD25–, CD38–), Langerhans cells (CD1a–), T lymphocytes (CD3–, TCR–, CD4–, CD25–), B cells (CD19–), fibroblasts (collagen I+, CD34+), epithe- Table2.Fibrocytes secrete chemokines, cytokines and growth fac- – – lial cells (cytokeratin ) and endothelial cells (vWF , tors implicated in wound repair. CD11b+). In addition, when examined by electron mi- croscopy, fibrocytes exhibit unique cytoplasmic exten- Factor Comments Ref. sions intermediate in size between microvilli and a chemokines pseudopodia which further differentiate fibrocytes from MIP1a constitutive; ≠ with TGF-b1 or IL-1b [8] blood-borne leukocytes [2]. MIP1b constitutive; ≠ with TGF-b1 or IL-1b [8] Fibrocytes expressing CD34, CD11b and collagen I (but MCP-1 constitutive; ≠ with TGF-b1 or IL-1b [8] not CD14, CD3 or CD10) found in the peripheral blood, b chemokines wound sites and areas of tissue remodeling should not be IL-8 constitutive; ≠ with TGF-b1 or IL-1b [8, 31] GROa constitutive; ≠ with TGF-b1 or IL-1b [8] confused with fibrocytes or fibroblast cultures which are Cytokines TNF-a with IL-1b stimulation [8] IL-6 with IL-1b or TNF stimulation [8] IL-10 with IL-1b or TNF stimulation [8] Obtain whole blood Growth Factors Angiogenin constitutive [31] Centrifugation over Ficoll-Paque CTGF constitutive [31] IGF-1 constitutive [31] M-CSF constitutive [8, 31] Isolate buffy coat, wash and perform low speed spin (remove platelets) PDGF constitutive [31] TGFb constitutive [8] Plate resultant PBMCs in DMEM 10% FBS Other MMP-9 Constitutive, active and latent [31] aSMA ≠ with TGF-b1 [4] Culture for 10–14 days (‘crude fibrocytes’)

Immunomagnetic bead separation to isolate ‘fibrocytes’

Fibrocytes (CD 14–/CD11b+/collagen I+ or CD14–/CD34+/collagen I+) Figure 1. Schematic representation indicating the isolation and growth conditions for peripheral blood fibrocytes. Taken from [4]. 1344 C. N. Metz The role of fibrocytes in wound healing implicated in the amyloid fibrillogenesis described by that cultured peripheral blood fibrocytes require interac- Harris and colleagues [5]. Nor should they be confused tion with activated T cells to permit their early differenti- with ‘spiral fibrocytes’ located within the ear. ation. The requirement for T cell interaction is similar to Spiral ligament fibrocytes are the cells which intercon- that reported for the differentiation of CD1a+ dendritic nect with basal cells of the stria vascularis via gap junc- cells [9]. Following their interaction with T cells, it is pro- tions and are postulated to play a critical role in main- posed that they migrate to the wound site (see fig. 2 and taining cochlear homeostasis [6–7]. Both of these cell below). Within the wound, these early-differentiated fi- types appear to be unrelated to the blood-borne fibrocytes brocytes might further interact with recruited T cells and implicated in wound healing. then fully differentiate into mature fibrocytes following exposure to transforming growth factor-b (TGFb, ex- pressed in early wounds). In response to TGF-b, it is pos- Ex vivo cultured peripheral fibrocytes originate from tulated that these mature fibrocytes express increased a CD14+ cells and differentiate into a cell population actin (aSMA) which provides a contrac- with wound-healing potential tile force for wound closure and produce collagen and other critical extracellular matrix molecules that promote The precise origin of peripheral blood fibrocytes has puz- wound healing (fig. 2). zled investigators since their discovery. Fibrocytes ex- press CD45 (leukocyte common antigen), a marker of bone marrow-derived cells (see table 1). Early studies us- Where are fibrocytes found? ing sex-mismatched, bone marrow chimeric mice to- gether with DNA amplification of the male-specific SRY Although initially identified as a blood-borne cell popu- gene showed that circulating fibrocytes in vivo arise from lation (CD34+, CD11b+ and/or collagen1+), fibrocytes radioresistant bone marrow progenitor cells or an uniden- have been localized to various tissues under both normal tified tissue source [2]. and pathological conditions (see table 3). Fibrocytes ini- Recent studies by Abe and colleagues, examining the ori- tially were localized to scar tissues, as well as in im- gin and differentiation pathway of this cell population, planted wounded chambers [2]. Since the first report show that peripheral blood fibrocytes isolated from blood identifying blood-borne fibrocytes, cells with a similar differentiate ex vivo from an adherent CD14+ cell popu- phenotype (CD34+) have been described in cutaneous lation [4]. Although fibrocytes do not require the addition wounds, keloids, fibrotic tissues, normal tissue, cuta- of specific growth factors (other than those present in fe- neous tumors and mesenchymal tumors. Because CD34, tal calf serum), they do proliferate slowly in response to a marker present on endothelial cells, was the only interleukin (IL)-1b and tumor necrosis factor a (TNF-a) marker used for these studies, caution must be used in in- [8] and to basic fibroblast growth factor (bFGF) and vas- terpreting the results. In our experience, it is relatively cular endothelial growth factor (VEGF) [C. N. Metz, un- easy to differentiate between tissue fibrocytes and en- published observations]. Based on in vitro studies, it is dothelial cells because the endothelial cells are CD34Hi postulated that circulating ‘progenitor fibrocytes’ un- and fibrocytes are CD34Lo, and because fibrocytes dergo phenotypic changes that promote the differentia- (elongated) are dispersed within the tissue, whereas en- tion of these circulating precursor cells to fibrocytes that dothelial cells form ringlike structures (vessels) and ac- are recruited to wound sites where they become mature cordingly form a pronounced pattern. Staining serial sec- fibrocytes and play a role in wound contracture and tions with vWF would further help discriminate between wound healing (fig. 2). Early observations demonstrated fibrocytes (vWF–) and endothelial cells (vWF+).

Figure 2. The proposed differentiation pathway of fibrocytes from a circulating precursor population. Modified from [4]. CMLS, Cell. Mol. Life Sci. Vol. 60, 2003 Review Article 1345

Table3. In situ localization of fibrocytes. merous circulating cells, including monocytes, neu- trophils and T lymphocytes, migrate to tissue locations as Tissue type Reference the result of chemokine-chemokine receptor interactions. Wound tissue Based on these observations, the fibrocyte-associated Subcutaneously implanted wound chambers [2] chemokine receptor expression profile was examined [4]. Cutaneous wounds [4] These studies revealed the expression of several chemo- Scar tissue kine receptors on the surface of fibrocytes, including Cutaneous/subcutaneous [2] Red and pink [35] CCR3, CCR5, CCR7 and CXCR4. Using both in vitro Keloids/hypertrophic scars [36] and in vivo fibrocyte chemotaxis techniques, Abe and Normal tissue colleagues revealed that fibrocytes migrate in response to Normal skin [37] secondary lymphoid chemokine (SLC), the ligand for Normal pancreatic tissue [18] CCR7. SLC (also known as 6Ckine, Exodus-2 and TCA- Normal breast tissue [10, 38] 4) has been shown to play a role in the organization of Fibrotic tissue Fibrotic granulomatous liver [8] lymphoid tissues during development by attracting T lymphocytes and mature dendritic cells [11]. Interest- Tumors Basal cell carcinomas (dermal) [39] ingly, the expression of SLC at sites of inflammation Mesenchymal tumors [42–47] has been reported [12]. Thus, it appears that fibro- cytes express several chemokine receptors and can mi- grate to wound sites in response to specific chemokine Interestingly, the occurrence of CD34+ fibrocytes in the gradients. stroma surrounding skin tumors has been found to be of diagnostic significance. Kirchmann and colleagues report that CD34+ fibroblast like cells are abundant The role of fibrocytes in wound healing in benign desmoplastic trichoepitheliomas, whereas basal cell carcinomas and adnexal carcinomas lack Fibrocytes have been postulated to play a pivotal role CD34+ cells [19–20]. Similarly, the presence or absence in wound healing and tissue repair processes. Fibro- of CD34+ fibrocytes may be useful in differentiating cytes can contribute to wound healing by numerous between invasive breast carcinomas and benign breast potential mechanisms: (i) by serving as potent antigen- lesions [10]. Based on the observations that benign presenting cells (APCs); (ii) by producing important breast lesions (sclerosing adenosis) exhibit stromal cytokines, chemokines and growth factors necessary CD34+ cells, whereas stroma derived from invasive for wound repair (see table 2); (iii) by secreting essential breast carcinomas is devoid of CD34+ fibrocytes, Barth extracellular matrix proteins involved in wound repair and colleagues report that the absence of CD34+ fibro- (see table 1); (iv) by serving as a contractile force in cytes favors the diagnosis of human basal cell carcinoma wound closure via aSMA expression and (v) by promot- when distinguishing between basal cell carcinomas and ing angiogenesis, a critical step in the wound repair benign skin appendage tumors [10]. Together these ob- process. servations suggest that the presence of CD34+ fibroblast- like cells might prove to be very useful in differentiating between benign (presence of CD34+ fibrocytes) and in- Fibrocytes are potent APCs able to recruit vasive/malignant lesions (absence of CD34+ fibrocytes). and activate T cells The lack of fibrocytes with antigen-presenting capabili- The skin is the first immune defense barrier which ties (see below) in invasive tumor sites suggests that serves to protect the host against infection [13]. When these cells may play a role in local immune surveillance this critical barrier is damaged, pathogenic bacteria can and the loss of these cells may permit an invasive pheno- easily invade. One mechanism by which the body de- type. fends itself is the strategic location of APCs at specific sites to initiate antigen-specific host immune responses. Several studies support the critical role of fibrocytes in Peripheral blood fibrocytes: recruitment to wound the initiation of immunity during tissue injury and repair. sites Isolated human fibrocytes express the cell surface mole- The original report describing fibrocytes details the ap- cules required for antigen presentation, including major pearance of blood-borne fibroblast-like cells in subcuta- histocompatibility complex molecules (HLA-DP, HLA- neously implanted wound chambers in mice and in early DQ and HLA-DR), the costimulatory molecules [CD80 human cutaneous scar tissues [2]. However, the mecha- (B7-1) and CD86 (B7-2)] as well as adhesion molecules nism(s) by which peripheral blood fibrocytes migrate to [CD11a (LFA-1), CD54 (ICAM-1) and CD58 (LFA-3)] specific sites of tissue injury were not understood. Nu- [3]. The expression level of these specific markers by fi- 1346 C. N. Metz The role of fibrocytes in wound healing brocytes is similar to that expressed by monocytes. Fibrocytes secrete extracellular matrix proteins Moreover, human fibrocytes localized to cutaneous scar implicated in wound repair/remodeling and disease tissues express high levels of HLA-DR in situ [3], sug- pathogenesis gesting that fibrocytes present in the wounded areas function as APCs. Fibrocytes produce extracellular matrix molecules Because human leukocyte antigen-D-related (HLA-DR) found during tissue repair expression is considered a prerequisite for antigen pre- Reparative cells contribute to wound healing by secreting sentation in vivo [14], Chesney and colleagues tested the extracellular matrix components. The production of ex- functional capacity of both human and mouse fibrocytes tracellular matrix molecules by these cells is regulated by to present antigen and stimulate antigen-specific T lym- numerous specific growth factors and other mediators. A phocytes in vitro [3]. Human fibrocytes induce APC-de- deficiency in these regulatory factors is postulated to pendent T cell proliferation when cultured with specific cause delayed healing (insufficient extracellular matrix), antigen, suggesting that fibrocytes play a role in the initi- whereas an excess of these factors could promote scar- ation of antigen-specific immunity. When compared with ring (excessive extracellular matrix). Ex vivo cultured fi- monocytes and dendritic cells for antigen-presenting ca- brocytes express numerous extracellular matrix mole- pacity in vitro (using the same autologous T cells), fibro- cules, including vimentin, fibronectin, collagen I and col- cytes were between monocytes (low) and dendritic cells lagen III (table 1), and fibrocytes localized to wounds in (high) when assessed by peak antigen-dependent T cell situ express collagen, suggesting their role in wound re- proliferation induced by the APCs [3]. pair. Similarly, isolated mouse fibrocytes express major histo- compatibility markers, adhesion molecules and costimu- The potential role of fibrocytes in fibrosis latory molecules (I-a, I-E, CD54 and CD86) required for and scarring antigen presentation [3]. Mouse fibrocytes cultured ex The regulation of matrix production within the wound is vivo, pulsed with foreign antigen in vitro, and then in- critical to prevent fibrosis during the tissue repair jected into mouse skin, migrate into regional lymph process. Numerous reports identify the presence of fibro- nodes where they sensitize naive T cells and/or activate cytes in scar tissue (see table 3). In the case of experi- memory T cells in vivo [3]. These observations further mental schistosomiasis, a parasitic infection where T-cell- support the role of fibrocytes in the initiation of antigen- mediated reactions against parasitic eggs sequestered in specific immunity. In addition, human fibrocytes secrete the lung and liver result in severe fibrosis, CD34+ fibro- MIP-1a and MIP-1b (see table 2), potent chemoattractant cytes were found in areas of connective tissue matrix de- molecules for CD4+ T cells. These CD4+ T cells are con- position within fibrotic livers [8]. By contrast, no CD34+ sidered essential for the generation of an antigen-specific cells were present in the normal livers of uninfected mice, response in vivo [15]. Thus, fibrocytes may contribute to suggesting that fibrocytes may contribute to the fibrotic the host defense response during tissue injury/tissue in- pathology associated with schistosomiasis. Further stud- vasion by recruiting and activating T lymphocytes to sites ies implicate fibrocytes in the pathogenic fibrosis associ- of injury. ated with radiation damage [17], Lyme disease [24–26] More recent studies by Zhu and colleagues [16] charac- (see below) as well as pulmonary fibrosis [C. N. Metz, terized fibrocytes isolated from Macaques and found unpublished observations]. Therefore, it is important to them to be phenotypically similar to human and mouse fi- understand the regulation of connective tissue molecules brocytes (i.e. CD34+ and collagen+). The Macaque fibro- by fibrocytes to prevent fibrosis while promoting wound cytes were transfected with a vector encoding green fluo- repair. rescent protein or DNA expression vectors encoding the simian immunodeficiency virus (SIVmne) structural and The presence of fibrocytes in stromal tissue: regulatory genes and then tested for their ability to aug- an indication for differentiating between malignant ment antigen presentation for SIV vaccines. These stud- and noninvasive (benign) tumor lesions ies suggest that fibrocytes are a readily accessible source Stromal remodeling is associated with chronic pancreati- of APCs capable of initiating and promoting T cell im- tis and ductal adenocarcinomas. A very recent investiga- munity. Furthermore, they highlight the potential clinical tion reported the distribution of CD34+ fibrocytes in pan- utility of fibrocytes in vaccine development for the treat- creatic diseases (pancreatic adenocarcinoma, endocrine ment of diseases such as human immunodeficiency virus tumors of the pancreas and chronic pancreatitis) [18]. (HIV) or cancer. However, further studies are required to Morphological analysis showed spindle-shaped cells with better characterize the functional capacity of fibrocytes small centrally located elongated nuclei and long slender as APCs before they can be used for clinical vaccine de- dendrite-like projections. In normal pancreatic tissue, velopment. there were few CD34+ fibrocytes, whereas there was an increased number of stromal CD34+ fibrocytes in tissue CMLS, Cell. Mol. Life Sci. Vol. 60, 2003 Review Article 1347

(predominantly located in areas of diffuse or nodular stro- Fibrocytes are TGF-b-responsive cells that express mal fibrosis around intralobular ducts and acini) obtained aSMA: Their potential role in wound contracture from patients with chronic pancreatitis. By contrast, Based on their presence within wounds and their expres- stroma-associated CD34+ fibrocytes were absent from sion of collagen I and collagen III, fibrocytes have been both pancreatic endocrine tumors and adenocarcinomas. postulated to mediate wound healing and/or fibrosis. These data suggest an association between CD34+ fibro- However, their functional role in these activities has not cytes in pancreatitis and stromal fibrosis, whereas pan- been well characterized. Recently, Abe and colleagues creatic endocrine tumors and adenocarcinomas lack showed that fibrocytes could differentiate into ‘myofi- CD34+ fibrocytes. broblast-like’ cells that express aSMA in response to TGF-b and exhibit a contractile force in vitro [4]. Myofi- The role of fibrocytes in disease pathogenesis broblasts are transiently found in early to mid-phase Lyme disease, transmitted by the spirochete Borrelia wound tissues and have been proposed to exert a critical burgdorferi, is the most common vector-borne illness in contractile force required to close tissue wounds (re- the United States (reviewed in [21–22]). It is a multisys- viewed in [27–28]). These cells respond to TGF-b with tem disease that affects the skin, nervous system, heart increased aSMA expression, enhanced collagen produc- and joints. Ticks deposit B. burgdorferi into the tion and increased contractile activity in vitro. Likewise, of their host, where eventually they become associated treatment of ex vivo cultured fibrocytes isolated from pe- with collagen fibers. Recent studies reveal the interac- ripheral blood with TGF-b enhanced both collagen pro- tion between decorin-binding adhesins present on the B. duction and aSMA expression by these cells. Further- burgdorferi and host tissue collagen-associated proteo- more, isolated fibrocytes contract collagen gels in vitro, glycan decorin [23]. Joint diseases associated with un- and the treatment of these cells with TGF-b increased treated B. burgdorferi infection include arthritis and fi- their contracture abilities (fig. 3) [4]. Thus, fibrocytes bromyalgia, a chronic pain syndrome with diffuse joint and appear to share many common fea- and muscle pain. However, the process by which the bac- tures, including their transient presence within the teria invade the joint tissue is not completely understood. wound, production of numerous extracellular matrix pro- Based on the production of extracellular matrix proteins teins, pro-inflammatory cytokines, chemokines, and en- (collagen I, III, fibronectin and vimentin) by fibrocytes, hanced collagen secretion and gel contracture ability fol- Grab and colleagues investigated the interaction between lowing treatment with TGF-b. Of course, the question re- isolated fibrocytes and B. burgdorferi in vitro [24]. Us- mains whether fibrocytes and myofibroblasts are two ing electron microscopy, they revealed that B. burgdor- distinct populations. In summary, the addition of TGF-b feri are not phagocytosed by fibrocytes (isolated from – a multifunctional growth factor expressed in the early humans or rhesus monkeys), but rather sequestered wounds critical for tissue repair – to fibrocytes facilitates within the cell membrane by tubelike processes extend- ing from the fibrocytes. They postulate that this semiin- ternalization of the B. burgdorferi by fibrocytes serves to enhance the infection by transporting the bacteria from the peripheral circulation to the joint while protecting the bacteria from the host immune system, and thus may play an important role in the pathogenesis of Lyme dis- ease. Based on the observations that fibrocytes localize to areas of matrix deposition in Schistosoma japonicum- infected granulomatous livers in mice (implicating per- sistent fibrocyte-T cell activation responses in the devel- opment of fibrotic liver disease [8]), and that the Lyme disease spirochete can invade peripheral blood fibro- cytes [25], Grab and colleagues hypothesize that fibro- cytes are the active immune cells that contribute to some of the pathologies observed in Lyme neuroborreliosis [26]. Furthermore, the secretion of cytokines/chemo- kines by fibrocytes (TNF-a, IL-6, MIP1a, MIP1b) Figure 3. Fibrocytes contract collagen gels in vitro. PBMCs (), which skew the CD4+ Th response could also promote cultured enriched fibrocytes (untreated = ; TGFb-treated (10 the progression of Lyme disease. Thus, fibrocytes may ng/ml) = ), or dermal fibroblasts () were resuspended in a colla- 5 be an important cell type involved in the pathogenesis of gen I solution at 10 cells/ml and subjected to a gel contraction as- say (n = 3). The data are shown as percent gel contraction (from the Lyme disease. beginning of the experiment) ± SE. * = P < 0.05, as determined by the Student’s t-test. Taken from [4]. 1348 C. N. Metz The role of fibrocytes in wound healing their differentiation toward a wound-healing phenotype mation) in vitro [31]. By contrast, fibrocytes express few similar to that exhibited by myofibroblasts (illustrated in anti-angiogenic molecules [31]. fig. 2). A critical event during the invasion stage of angiogenesis is the proteolysis of the basement membrane. Previous stud- ies have shown that matrix metalloproteinases (MMPs) Fibrocytes promote endothelial cell proliferation, mediate the dissolution of the basement membrane during migration and tube formation in vitro and early tissue repair and initiate angiogenesis. Although angiogenesis in vivo MMPs are not present in normal skin, both MMP-2 and Normal wound healing requires angiogenesis that facili- MMP-9 are strongly induced within 24 h of wounding [32, tates the removal of debris and prepares the wound bed 33]. MMP-9 is the main MMP found in wound fluid, with for development of a critical framework of granulation peak activity expressed between 2 and 4 days post-wound- tissue necessary for wound closure. These newly formed ing [34]. Consistent with these observations, fibrocytes vessels represent over 50% of the granulation tissue mass home to cutaneous wound sites in vivo within 1–4 days found in early wounds (reviewed in [29]). Wound-related [2], and ex vivo cultured fibrocytes constitutively express angiogenesis appears to be regulated by the interaction of MMP-9 messenger RNA (mRNA) and secrete high levels endothelial cells with the extracellular matrix within the of active MMP-9 [31]. Together, these data demonstrate wound space [30]. Although numerous cellular mediators that cultured fibrocytes secrete factors that promote an an- of wound healing have been identified, few studies have giogenic phenotype in endothelial cells in vitro. Clearly, focused on the role of specific cell types that mediate an- the timing of observation and the local tissue environment giogenesis during wound healing. will significantly effect the expression of MMPs, growth Based on previous observations that fibrocytes secrete factors and cytokines by fibrocytes in vivo. IL-8 and other growth factors, Hartlapp and colleagues Further studies using the Matrigel implant model of an- characterized the production of numerous pro-and anti- giogenesis in mice, show that fibrocytes (fig. 4E) (and fi- angiogenic factors by ex vivo cultured fibrocytes (table brocyte culture supernatants (fig. 4C) promote blood 2). Fibrocytes secrete VEGF, platelet-derived growth fac- vessel formation in vivo. Thus, it appears that fibrocytes tor (PDGF), angiogenin, IL-1b, granulocyte colony-stim- may play a role in blood vessel formation during the ini- ulating factor (GCSF) and bFGF. In addition to promot- tial stages of wound healing based on their early presence ing endothelial cell proliferation in vitro, culture super- within the wound and their ability to promote endothelial natants obtained from fibrocytes promote endothelial cell cell proliferation, migration, differentiation, as well as migration and endothelial cell differentiation (tube for- neovascularization in vivo.

Figure 4. Fibrocytes and fibrocyte culture supernatants induce angiogenesis in vivo. Matrigel was injected into mice (n = 5 per group) con- taining (A) heparin plus aFGF (positive control); (B) heparin alone (negative control for Matrigel model); (C) heparin plus fibrocyte-con- ditioned media; (D) heparin plus unconditioned media (negative control for heparin plus fibrocyte-conditioned meda); (E) mouse fibro- cytes; or (F) NIH 3T3 cells (negative control for mouse fibrocytes). After 6 days, the Matrigel plugs were removed and examined for blood vessel formation using Masson’s Trichrome staining. Stained sections were photographed at 100 ¥. Representative Matrigel plugs are shown. Epidermis (Ep); Matrigel (Ma); skeletal muscle (SM); vessels (V); Taken from [31]. CMLS, Cell. Mol. Life Sci. Vol. 60, 2003 Review Article 1349

Future research directions 8 Chesney J., Metz C., Stavitsky A. B., Bacher M. and Bucala R. (1998) Regulated production of type I collagen and inflamma- tory cytokines by peripheral blood fibrocytes. J. Immunol. 160: Numerous studies demonstrate the potential role of fibro- 419–425 cytes in wound healing. These investigations highlight the 9 Shreedhar V., Moodycliffe A. M., Ullrich S. E., Bucana C., localization of fibrocytes at sites of tissue damage and re- Kripke M. L. and Flores-Romo L. (1999) Dendritic cells re- pair where they could (i) initiate antigen-specific immu- quire T cells for functional maturation in vivo. Immunity 11: 625–636 nity as APCs; (ii) secrete extracellular matrix proteins, cy- 10 Barth P. J., Ebrahimsade S., Ramaswamy A. and Moll R. (2002) tokines and pro-angiogenic molecules, which promote CD34+ fibrocytes in invasive ductal carcinoma, ductal carci- wound repair and (iii) express aSMA, which mediates noma in situ and benign breast lesions. Virchows Arch. 440: wound closure. Based on their ability to secrete extracellu- 298–303 11 Saeki H., Moore A. M., Brown M. J. and Hwang S. T. (1999) lar matrix molecules and their presence within scar tissue, Cutting edge: secondary lymphoid-tissue chemokine (SLC) numerous studies also implicate fibrocytes in fibrosis and and CC chemokine receptor 7 (CCR7) participate in the emi- scarring, typical in connective tissue disorders such as, gration pathway of mature dendritic cells from the skin to re- schistosomiasis, lung fibrosis, keloids and scleroderma. gional lymph nodes. J. Immunol. 162: 2472–2475 12 Hjelmstrom P., Fjell J., Nakagawa T., Sacca R., Cuff C. A. and + Also, the localization of APC-associated CD34 fibrocytes Ruddle N. H. (2000) Lymphoid tissue homing chemokines are in pancreatic tissue and stroma-associated fibrocytes in expressed in chronic inflammation. Am. J. Pathol. 156: pancreatic tissue (during pancreatitis), and the absence of 1133–1138 13 Schmitt D. (1999) Immune functions of the human skin. Mod- CD34+ fibrocytes in pancreatic endocrine tumors and ade- els of in vitro studies using Langerhans cells. Cell Biol.Toxicol. nocarcinomas suggest that these cells play a role in local 15: 41–45 immune surveillance. In other words, the loss of tissue-as- 14 Geppert T. D and Lipsky P. E. (1985) Antigen presentation by sociated fibrocytes with antigen-presenting abilities may interferon-gamma-treated endothelial cells and fibroblasts: dif- ferential ability to function as antigen-presenting cells despite contribute to the invasive nature of the tumor. comparable Ia expression. J. Immunol. 135: 3750–3762 In the setting of wound healing, one can predict that too 15 Schall T. J., Bacon K., Camp R. D., Kaspari J. W. and Goeddel few fibrocytes (or underactive fibrocytes) at the site of D. V. (1993) Human macrophage inflammatory protein alpha tissue damage may result in a weak host immune re- (MIP-1 alpha) and MIP-1 beta chemokines attract distinct pop- ulations of lymphocytes. J. Exp. Med. 177: 1821–1826 sponse, permitting further pathogenic invasion and a de- 16 Zhu Y., Koo K., Bradshaw J. D., Sutton W. F., Kuller L. R., Bu- layed wound healing response. By contrast, too many fi- cala R. et al. (2000) Macaque blood-derived antigen-presenting brocytes or prolonged, persistent fibrocyte-mediated T cells elicit SIV-specific immune responses. J. Med. Primatol. cell activation within the damaged tissue region may con- 29: 182–192 17 Burger A., Loffler H., Bamberg M. and Rodemann H. P. (1998) tribute to fibrosis and scarring. Future studies are re- Molecular and cellular basis of radiation fibrosis. Int. J. Radiat. quired to determine how the delicate balance between the Bol. 73: 401–408 wound-healing properties and pro-fibrotic abilities of fi- 18 Barth P. J., Ebrahimsade S., Hellinger A., Moll R. and Ra- + brocytes is achieved. maswamy A. (2002) CD34 fibrocytes in neoplastic and in- flammatory pancreatic lesions. Virchows Arch. 440: 128–133 19 Kirchmann T. T., Prieto V. G. and Smoller B. R. (1994) CD34 Acknowledgements. This work was supported by a grant from the staining pattern distinguishes basal cell carcinoma from tri- Picower Foundation. choepithelioma. Arch. Dermatol. 130: 589–592 20 Kirchmann T. T., Prieto V. G. and Smoller B. R. (1995) Use of CD34 in assessing the relationship between stroma and tumor 1 Dunphy J. E. The fibroblast: a ubiquitous ally for the surgeon. in desmoplastic keratinocytic neoplasms. J. Cutan. Pathol. 22: (1963) N. Engl. J. Med. 268: 1367–1371 422–426 2 Bucala R., Spiegel L. A., Chesney J., Hogan M. and Cerami A. 21 Steere A. C. (2001) Lyme disease. N. Engl. J. Med. 345: 115– 125 (1994) Circulating fibrocytes define a new leukocyte subpopu- 22 Steere A. C. (1995) Musculoskeletal manifestations of Lyme lation that mediates tissue repair. Mol. Med. 1: 71–81 disease. Am. J. Med. 98: 44S–48S 3 Chesney J., Bacher M., Bender A. and Bucala R. (1997) The pe- 23 Guo B. P., Brown E. L., Dorward D. W., Rosenberg L. C. and ripheral blood fibrocyte is a potent antigen-presenting cell ca- Hook M. (1998) Decorin-binding adhesins from Borrelia pable of priming naive T cells in situ. Proc. Natl. Acad. Sci. burgdorferi. Mol. Microbiol. 30: 711–723 USA 94: 6307–6312 24 Grab D. J., Lanners H., Martin L. N., Chesney J., Cai C., Ad- 4 Abe R., Donnelly S. C., Peng T., Bucala R. and Metz C. N. kisson H. D. et al. (1999) Interaction of Borrelia burgdorferi (2001) Peripheral blood fibrocytes: differentiation pathway and with peripheral blood fibrocytes, antigen-presenting cells with migration to wound sites. J. Immunol. 166: 7556–7562 the potential for connective tissue targeting. Mol. Med. 5: 46– 5 Harris D. L., King E., Ramsland P. A. and Edmundson A. B. 54 (2000) Binding of nascent collagen by amyloidogenic light 25 Grab D. J., Lanners H. N., Williams W. L. and Bucala R. (1999) chains and amyloid fibrillogenesis in monolayers of human fi- Peripheral blood fibrocytes with foamy virus infection-like brocytes. J. Mol. Recognit.. 13: 198–212 morphology. Hum. Pathol. 30: 1395–1396 6Takahashi T. and Kimura R. S. (1970) The ultrastructure of the 26 Grab D., Salim M., Chesney J., Bucala R. and Lanners H. spiral ligament in the Rhesus monkey. Acta Otolaryngol. 69: (2002) A role for peripheral blood fibrocytes in Lyme disease? 46–60 Med. Hypotheses. 59: 1 7 Ichimiya I., Yoshida K., Hirano T., Suzuki M. and Mogi G. 27 Powell D. W., Mifflin R. C., Valentich J. D., Crowe S. E., Saada (2000) Significance of spiral ligament fibrocytes with cochlear J. I. and West A. B. (1999) Myofibroblasts. I. Paracrine cells im- inflammation. Int. J. Pediatr. Otorhinolaryngol. 56: 45–51 portant in health and disease. Am. J Physiol. 277: C1–C9 1350 C. N. Metz The role of fibrocytes in wound healing

28 Powell D. W., Mifflin R. C., Valentich J. D., Crowe S. E., Saada 38 Yamazaki K. and Eyden B. P. (1995) Ultrastructural and im- J. I. and West A. B. (1999) Myofibroblasts. II. Intestinal subepi- munohistochemical observations on intralobular fibroblasts of thelial myofibroblasts. Am. J. Physiol. 277: C183–C201 human breast, with observations on the CD34 antigen. J. Sub- 29 Davidson J. (1992) Wound repair. In: Inflammation: Basic Prin- microsc. Cytol. Pathol. 27: 309–323 ciples and Clinical Correlates, pp. 809–819, Gallin J. and 39 Humphreys T. R., Monteiro M. R. and Murphy G. F. (2000) Goldstein I. (eds), Raven Press, New York Mast cells and dendritic cells in basal cell carcinoma stroma. 30 Tonnesen M. G., Feng X. and Clark R. A. (2000) Angiogenesis Dermatol. Surg. 26: 200–203 in wound healing. J. Investig. Dermatol. Symp. Proc. 5: 40–46 40 Luttges J., Mentzel T., Hubner G. and Kloppel G. (1999) Soli- 31 Hartlapp I., Abe R., Saeed R. W., Peng T., Voelter W., Bucala R. tary fibrous tumour of the pancreas: a new member of the small et al. (2001) Fibrocytes induce an angiogenic phenotype in cul- group of mesenchymal pancreatic tumours. Virchows Arch. tured endothelial cells and promote angiogenesis in vivo. 435: 37–42 FASEB J. 15: 2215–2224 41 Seidal T. and Edvardsson H. (2000) Diagnosis of gastrointesti- 32 Agren M. S. (1994) Gelatinase activity during wound healing. nal stromal tumor by fine-needle aspiration biopsy: a cytologi- Br. J. Dermatol. 131: 634–640 cal and immunocytochemical study. Diagn. Cytopathol. 23: 33 Agren M. S. (1999) Matrix metalloproteinases (MMPs) are re- 397–401 quired for re-epithelialization of cutaneous wounds. Arch. Der- 42 Seidal T. and Edvardsson H. (1999) Expression of c-kit matol. Res. 291: 583–590 (CD117) and Ki67 provides information about the possible cell 34 Salo T., Makela M., Kylmaniemi M., Autio-Harmainen H. and of origin and clinical course of gastrointestinal stromal tu- Larjava H. (1994) Expression of matrix metalloproteinase-2 mours. Histopathology 34: 416–424 and -9 during early human wound healing. Lab. Invest. 70: 43 Silverman J. S. and Tamsen A. (1996) Mammary fibroadenoma 176–182 and some phyllodes tumour stroma are composed of CD34+ fi- 35 Ueda K., Furuya E., Yasuda Y., Oba S. and Tajima S. (1999) broblasts and factor XIIIa+ dendrophages. Histopathology 29: Keloids have continuous high metabolic activity. Plast. Re- 411–419 constr. Surg. 104: 694–698 44 Sircar K., Hewlett B. R., Huizinga J. D., Chorneyko K., Berezin 36 Abergel R. P., Pizzurro D., Meeker C. A., Lask G., Matsuoka L. I. and Riddell R. H. (1999) Interstitial cells of Cajal as precur- Y., Minor R. R. et al. (1985) Biochemical composition of the con- sors of gastrointestinal stromal tumors. Am. J. Surg. Pathol. 23: nective tissue in keloids and analysis of collagen metabolism in 377–389 keloid fibroblast cultures. J. Invest. Dermatol. 84: 384– 390 45 Suster S. and Fisher C. (1997) Immunoreactivity for the human 37 Narvaez D., Kanitakis J., Faure M. and Claudy A. (1996) Im- hematopoietic progenitor cell antigen (CD34) in lipomatous tu- munohistochemical study of CD34-positive dendritic cells of mors. Am. J. Surg. Pathol. 21: 195–200 human dermis. Am. J. Dermatopathol. 18: 283–288