mesothelial progenitor cells and their potential

Upload: josue-velazquez

Post on 04-Jun-2018

225 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    1/22

    The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    Review

    Mesothelial progenitor cells and their potentialin tissue engineering

    Sarah E. Herricka,, Steven E. Mutsaers b

    a School of Biological Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UKb Asthma&Allergy Research Institute, Department of Medicine, University of Western Australia, Nedlands, Australia

    Received 16 September 2003; received in revised form 3 November 2003; accepted 4 November 2003

    Abstract

    Themesothelium consists of a single layer of flattened mesothelial cells that lines serosal cavities and the majority of internal

    organs, playing important roles in maintaining normal serosal integrity and function. A mesothelial stem cell has not been

    identified, but evidence from numerous studies suggests that a progenitor mesothelial cell exists. Although mesothelial cells

    are of a mesodermal origin, they express characteristics of both epithelial and mesenchymal phenotypes. In addition, following

    injury, new mesothelium regenerates via centripetal ingrowth of cells from the wound edge and from a free-floating population

    of cells present in the serosal fluid, the origin of which is currently unknown. Recent findings have shown that mesothelial

    cells can undergo an epithelial to mesenchymal transition, and transform into myofibroblasts and possibly smooth muscle

    cells, suggesting plasticity in nature. Further evidence for a mesothelial progenitor comes from tissue engineering applications

    where mesothelial cells seeded onto tubular constructs have been used to generate vascular replacements and grafts to bridge

    transected nerve fibres. These findings suggest that mesothelial cell progenitors are able to switch between different cell

    phenotypes depending on the local environment. However, only by performing detailed investigations involving selective cell

    isolation, clonal analysis together with cell labelling and tracking studies, will we begin to determine the true existence of a

    mesothelial stem cell.

    2003 Elsevier Ltd. All rights reserved.

    Keywords: Peritoneum; Stem cells; Epithelial-mesenchymal transitions; Adhesions; Serosa

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622

    2. Embryology and morphology of mesothelial cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622

    3. Functions of the mesothelial cell layer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 623

    4. Mesothelial healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 625

    5. Adhesion formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627

    6. Evidence for a multipotential subserosal mesenchymal cell . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 628

    7. Epithelial-mesenchymal transition of mesothelial cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 629

    Corresponding author. Tel.: +44-161-275-6765; fax: +44-161-275-5945.

    E-mail address: [email protected] (S.E. Herrick).

    1357-2725/$ see front matter 2003 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.biocel.2003.11.002

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    2/22

    622 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    8. Tissue engineering potential of mesothelial cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 631

    8.1. Vascular grafts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 631

    8.2. Omental grafts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 632

    8.3. Nerve grafts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 633

    9. Does a mesothelial stem cell exist? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 634

    10. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 636

    Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 636

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 636

    1. Introduction

    The mesothelium lines the peritoneal, pleural and

    pericardial cavities with visceral and parietal surfaces

    covering the internal organs and body wall, respec-

    tively. It comprises a monolayer of epithelial-like

    cells resting on a thin basement membrane supported

    by sub-serosal connective tissue containing blood

    vessels, lymphatics, resident inflammatory cells and

    fibroblast-like cells (Wang, 1974; Ishihara et al., 1980;

    Albertine, Wiener-Kronish, Roos, & Staub, 1982). The

    sole function of the mesothelial layer was traditionally

    thought to provide a protective, non-adhesive surface

    to facilitate intracoelomic movement. However, it is

    now recognised as a dynamic cellular membrane with

    many physiological functions including the control

    of fluid and solute transport, immune surveillanceand the production of extracellular matrix (ECM)

    molecules, proteases, cytokines and growth factors.

    The mesothelium is bathed in serosal fluid that re-

    sembles an ultrafiltrate of plasma and contains blood

    proteins, resident inflammatory cells, sugars and var-

    ious enzymes including amylase and lactate dehydro-

    genase (Dondelinger, Boverie, & Cornet, 1982). The

    composition and volume of the serosal fluid is indica-

    tive of certain pathological states, such as peritonitis,

    tumorgenesis and endometriosis (Haney, 1993),and it

    is likely that the mesothelial layer responds as a singleunit to changes in serosal fluid composition. Indeed,

    repair of serosal tissue involves increased mesothelial

    cell proliferation at sites distant to the wound, suggest-

    ing diffuse activation of the mesothelium in response

    to mediators or cells released into the serosal fluid, or

    via cell to cell communication (Mutsaers, McAnulty

    et al., 1997; Mutsaers, Whitaker, & Papadimitriou,

    2002).

    Although local proliferation of resident cells sur-

    rounding a lesion is one source of healing cells, recent

    reports suggest that the repair of many organs in the

    adult organism also involves incorporation of multipo-

    tential stem cells and as such, has generated exciting

    prospects in cell and tissue engineering (Bianco &

    Robey, 2001; Goodell, 2001; Tuan, Boland, & Tuli,

    2003).A rich reservoir of these cells resides in specificniches within the bone marrow microenvironment as

    well as in a variety of connective tissues where they are

    maintained in an undifferentiated and quiescent state.

    At present, there is a lack of a unifying definition that

    characterises cells as stem cells. However, a general

    definition is a cell capable of extensive self-renewal

    that can give rise to successively more differentiated

    progeny cells (Wagers, Christensen, & Weissman,

    2002). Although a classic mesothelial stem cell has

    not been identified, many lines of evidence suggest that

    a mesothelial progenitor cell does exist. This reviewwill describe the mesothelial cell in terms of its embry-

    ological origin, morphological characteristics and di-

    verse functions. Subsequent sections present evidence

    to support the concept of a free-floating mesothelial

    progenitor cell present in serosal fluid and also dis-

    cuss mesothelial cell differentiation, novel tissue engi-

    neering applications for these cells and possible future

    research directions in this rapidly developing field.

    2. Embryology and morphology of mesothelial

    cells

    Bichart, in 1827 (reviewed by Whitaker, Papad-

    imitriou, & Walters, 1982a)first observed that serous

    cavities were lined by a layer of flattened cells similar

    to those of the lymphatics.Minot (1890)subsequently

    proposed the term mesothelium following a detailed

    study of its embryological origin that showed this layer

    to be the epithelial lining of mammalian mesoder-

    mic cavities. It is now understood that during human

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    3/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 623

    development, the intraembryonic mesoderm on each

    side of the neural groove differentiates into paraxial,

    intermediate and lateral mesoderm. The lateral meso-

    derm is continuous with the extraembryonic meso-derm covering the yolk sac and amnion. At the end of

    week 3, small spaces appear in the lateral mesoderm

    that fuse, dividing the mesoderm into two layers: the

    intraembryonic somatic or parietal layer and the in-

    traembryonic splanchnic or visceral layer. The somatic

    mesoderm and overlying embryonic ectoderm form

    the embryonic body wall (somatopleure), whereas the

    splanchnic mesoderm and embryonic endoderm form

    the embryonic gut wall (splanchnopleure). A contin-

    uous mesothelial membrane lines the margin of these

    two layers and so borders the intraembryonic coelom.

    Between 5 and 7 weeks, the coelom is sub-divided by

    a process of septation into a future pericardial cav-

    ity, two pleural cavities and a peritoneal cavity. In

    this phase of development, the mesothelial and sub-

    mesothelial layers of the coelom are referred to as the

    pericardium, pleura, and peritoneum respectively, and

    together as serous membranes (reviewed byThors and

    Drukker, 1997). Mesothelial cells are therefore of a

    primitive mesodermal origin, but share characteristics

    of both epithelial and mesenchymal cells (Whitaker,

    Manning, Robinson, & Shilkin, 1992).

    Morphologically, mesothelial cells are consideredgenerally similar at different serosal sites and be-

    tween different mammalian species (Whitaker et al.,

    1982a,b; Baradi & Rao, 1976; Whitaker, Papadi-

    mitriou, & Walters, 1980). In their fully differenti-

    ated state, they form a monolayer of predominantly

    squamous-like cells approximately 25 m in diame-

    ter, with characteristic surface microvilli and occa-

    sional cilia. The microvilli vary in shape, length and

    density between adjacent cells and between different

    organs (Mutsaers, Whitaker, & Papadimitriou, 1996).

    Mesothelial cells display many epithelial character-istics including a polygonal cell shape, cytokeratin

    intermediate filaments (cytokeratins 6, 8, 18 and 19)

    (Czernobilsky, Moll, Levy, & Franke, 1985), and the

    ability to secrete a basement membrane. However,

    they also show features of mesenchymal cells such

    as the presence of vimentin, desmin and upon stimu-

    lation, alpha smooth muscle actin (Afify, Al-Khafaji,

    Paulino, & Davila, 2002). Ultrastructural analysis of

    polarised mesothelial cells demonstrates well devel-

    oped cellcell junctional complexes including tight

    Fig. 1. Monolayer imprint of normal rat peritoneal mesothelialcells showing immunoreactivity for zonula occludens-1 expression,

    a plaque protein associated with tight junctions, localised to the

    plasma membrane. Bar, 10 m. Reproduced with permission from

    Foley-Comer et al. (2002).

    junctions (zonula occludens) located towards their

    luminal aspect, adherens junctions, gap junctions and

    desmosomes (Pelin, Hirvonen, & Linnainmaa, 1994)

    (Fig. 1). They also express E-, N- and P-cadherins,

    but unlike true epithelia, N-cadherin predominates

    (Simsir, Fetsch, Mehta, Zakowski, & Abati, 1999).

    Although mainly squamous in appearance, cuboidalmesothelial cells also exist at various locations includ-

    ing septal folds of the mediastinal pleura, parenchy-

    mal organs (liver, spleen), the milky spots of the

    omentum, and the peritoneal side of the diaphragm

    overlying the lymphatic lacunae (Wang, 1998). They

    also predominate following injury or stimulation of

    the serosal surface (Mutsaers et al., 2002; Whitaker &

    Papadimitriou, 1985). The two forms of mesothelial

    cell, squamous-like and cuboidal, also show differ-

    ences ultrastructurally. In particular, cuboidal cells

    have abundant mitochondria and rough endoplasmic

    reticulum (RER), a well developed Golgi apparatus,microtubules and a greater number of microfilaments

    compared with squamous cells, suggesting a more

    metabolically active state (Kluge & Hovig, 1967;

    Fukata, 1963; Baradi & Campbell, 1974).

    3. Functions of the mesothelial cell layer

    As well as providing a slippery, non-adhesive ep-

    ithelial surface, the mesothelial layer performs many

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    4/22

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    5/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 625

    has been associated with a worse prognosis and may

    represent a less differentiated tumour (Fusco et al.,

    1993).

    Mesothelial cells have also been implicated in boththe spread and inhibition of tumour growth within

    serosal cavities. It has been clearly shown that trau-

    matised mesothelial surfaces are privileged sites for

    tumour cell adhesion (Cunliffe & Sugarbaker, 1989).

    It has been suggested that this occurs due to upregula-

    tion of adhesion molecules on mesothelial cells in re-

    sponse to inflammatory mediators, promoting tumour

    cell adhesion (van der Wal et al., 1997). However,

    binding via integrins to exposed submesothelial con-

    nective tissue is likely to be the main mechanism of at-

    tachment (Sugarbaker, 1991).Tumour growth is then

    potentiated by growth factors released from activated

    mesothelial cells.

    Several studies have also shown that following

    surgical trauma, tumour growth is also increased at

    sites distal to the injury (Hofer, Shrayer, Reichner,

    Hoekstra, & Wanebo, 1998). Animal studies demon-

    strated that tumour growth was increased following

    exposure to surgical wound fluid or a combination of

    the growth factors, TGF- and FGF, suggesting that

    mediators produced after surgical trauma or by the

    tumour cells themselves, enhance local and distal tu-

    mour growth (Hofer et al., 1998). This may occur bystimulating tumour cell proliferation but also through

    upregulation of cell adhesion molecules on mesothe-

    lial cells promoting their attachment and invasion into

    serosal tissues.

    Many studies have demonstrated that adhesion of

    tumour cells to hyaluronan bound to mesothelial cells

    is important for the spread of ovarian and colorectal

    tumours (Casey & Skubitz, 2000; Harada et al., 2001;

    Lessan, Aguiar, Oegema, Siebenson, & Skubitz, 1999;

    Catterall, Jones, & Turner, 1999).However, evidence

    also suggests that secretion of hyaluronan by mesothe-lial cells into the serosal fluid may inhibit tumour cell

    adhesion (Casey & Skubitz, 2000; Jones, Gardner,

    Catterall, & Turner, 1995). Conditioned medium

    from confluent mesothelial cell cultures containing

    large amounts of hyaluronan prevented tumour cell

    attachment, but this inhibition was overcome follow-

    ing hyaluronidase treatment (Jones et al., 1995).It is

    likely that free hyaluronan in the conditioned medium

    bound to CD44 on the tumour cells and prevented

    them from binding to hyaluronan on the mesothelial

    cell surface. Removal of free hyaluronan may explain

    why tumour cells adhered to mesothelial cells in other

    studies.

    The secretion of pro-coagulants such as tissue factorand fibrin stabilisers plasminogen activator inhibitor

    (PAI)-1 and -2, as well as fibrinolytic mediators in-

    cluding the plasminogen activators (PA) urokinase

    PA (uPA) and tissue PA (tPA) by the mesothelium,

    demonstrates an importance in regulating haemostasis

    and fibrin clearance (Sitter et al., 1995). Following

    serosal injury, there is a fine balance between these

    processes, which if disrupted may result in the for-

    mation of adhesions, bands of fibrous tissue that

    occur in up to 95% of patients following surgery.

    Adhesions initially form as fibrin-rich deposits be-

    tween damaged, closely opposed serosal surfaces. If

    there is insufficient serosal fibrinolytic activity, these

    fibrin-rich adhesions persist, become organised by in-

    vading fibroblasts and endothelial cells and with sub-

    sequent collagen deposition form permanent fibrous

    adhesions within a week of injury (Sulaiman et al.,

    2000). Although the pathophysiology of adhesion

    formation is poorly understood, it is proposed that

    adhesions develop if regeneration of the mesothelial

    layer is impaired. However, there is much controversy

    regarding the mechanisms involved in normal serosal

    repair, in particular the cells involved in the regenera-tion of the mesothelium. For a more extensive review

    of mesothelial cell function seeMutsaers (2002).

    4. Mesothelial healing

    Hertzler (1919)was the first to observe that small

    and large peritoneal wounds healed in the same

    amount of time. He concluded that the mesothe-

    lium could not regenerate solely by proliferation and

    centripetal migration of cells at the wound edge asoccurs for the healing of epithelium. Since then,

    many studies involving a wide range of experimental

    model systems have been performed to elucidate the

    mechanisms regulating the regeneration process.

    It is generally agreed that the healing process be-

    gins within 24 h of injury with the appearance of

    a population of rounded cells, predominantly neu-

    trophils and macrophages, on the wound surface

    (Mutsaers et al., 2002). Mesothelial cells at the

    wound edge undergo cell division and the epithelial

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    6/22

    626 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    sheet temporally transforms into spindle-shaped fi-

    broblastic cells that migrate onto the denuded wound

    area (Whitaker & Papadimitriou, 1985; Johnson &

    Whitting, 1962; Bridges & Whitting, 1964;Mutsaers,Whitaker, & Papadimitriou, 2000). We have shown

    that proliferative factors (Mutsaers, McAnulty et al.,

    1997) and chemotactic factors, such as HGF, are

    likely to play a major role in stimulating this repair

    process (Warn et al., 2001). Under normal condi-

    tions, the mesothelium is a slowly renewing tissue

    with 0.160.5% of cells undergoing mitosis at any

    one time (Mutsaers et al., 2000; Fotev, Whitaker, &

    Papadimitriou, 1987). However, they can be stimu-

    lated to divide by a variety of agents as well as by

    direct physical damage. Watters and Buck (1973)

    showed that mesothelial cells on opposing serosal

    surfaces undergo maximal division 2 days after in-

    Fig. 2. Monolayer imprint of tritiated thymidine treated murine serosal lesions at (A) 24 h, (B) 2 days and (C) 4 days after injury. Dark

    nuclei are labelled with silver grains (small arrows) and represent cells undergoing division. The centre of the lesion (c), the margin between

    the centre and edge of the lesion defined by thick arrows, is identified by a high density of cells, many of which are inflammatory cells.

    At 24 h, few mesothelial cells surrounding the wound are undergoing division. By 2 days approximately 28% of these cells are dividing.

    At 4 days, the majority of dividing cells are at the wound centre and are characterised as mesothelial cells. Bar, 125 m. Reproduced with

    permission fromMutsaers et al. (2000).

    jury. Later, kinetic studies using [3H]-thymidine in-

    corporation in rodent models confirmed that 2860%

    of mesothelial cells at the wound edge and on the

    opposing surface were dividing 2448 h after injury(Fig. 2) (Whitaker & Papadimitriou, 1985; Mutsaers

    et al., 2000; Fotev et al., 1987). Our subsequent stud-

    ies showed that uninjured murine testicular mesothe-

    lium has a 0.25% basal mitotic activity, which upon

    stimulation by the exogenous addition of peritoneal

    inflammatory lavage cells and activated macrophages,

    increased to values greater than 12% (Mutsaers et al.,

    2002). As inflammatory cells collect on the wound

    surface within the first 24 h of injury, it is likely that

    they play a significant role in inducing mesothelial

    cell proliferation and stimulating serosal repair.

    Irrespective of the size of the damaged wound area,

    type of trauma or animal species, serosal healing is

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    7/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 627

    complete within 710 days of injury when the wound

    area is covered by cells displaying all the charac-

    teristics of mesothelial cells (Mutsaers et al., 2002;

    Whitaker & Papadimitriou, 1985; Raftery, 1973;Teranishi, Sakaguchi, & Itaya, 1977). It is unlikely

    that the processes of cell division and migration alone

    account for these similar healing times. Based on this

    evidence, a number of groups have proposed addi-

    tional sources for the regenerating mesothelial cells.

    These include: macrophage transformation (Eskeland

    & Kjaerheim, 1966; Ryan, Grobety, & Majno,

    1973), exfoliation of mature or proliferating mesothe-

    lial cells from adjacent or opposing serosal sur-

    faces (Whitaker & Papadimitriou, 1985; Johnson &

    Whitting, 1962; Mutsaers et al., 2000; Fotev et al.,

    1987; Cameron, Hassan, & De, 1957; Watters &

    Buck, 1972),pre-existing free-floating serosal progen-

    itor cells that implant on the wound and differentiate

    into mesothelial cells (Ryan et al., 1973), subserosal

    mesenchymal precursors that convert into mesothe-

    lial cells and migrate to the wound surface (Raftery,

    1973; Ellis, Harrison, & Tugh, 1965; Bolen,

    Hammar, & McNutt, 1986;Davila & Crouch, 1993),

    and bone marrow-derived circulating precursors

    (Wagner, Johnson, Brown, & Wagner, 1982).

    The origin of these regenerating cells is highly

    controversial. Nevertheless, extensive experimentalevidence suggests that a free-floating serosal pro-

    genitor is probably involved. For instance, studies

    have demonstrated that mesothelial regeneration

    is impaired following selective irradiation at the

    site of injury but recoverable after the addition of

    peritoneal lavage cells (Whitaker & Papadimitriou,

    1985). Moreover,Cleaver, Hopkins, Ng Nee Kwong,

    & Raftery (1974) showed that the healing rate of

    mesothelium was retarded following post-operative

    peritoneal lavages, possibly due to the removal of

    the free-floating serosal cells. Further evidence for afree-floating progenitor arises from peritoneal fluid

    studies where a significantly higher number of viable

    free-floating mesothelial cells were recovered from

    experimental animals 2 days following injury com-

    pared with the control uninjured animals (Whitaker &

    Papadimitriou, 1985; Fotev et al., 1987). Our own

    group has performed cell-tracking and labelling stud-

    ies in rodent models and conclusively shown that

    serosal healing involves the incorporation and prolifer-

    ation of free-floating mesothelial cells (Foley-Comer

    et al., 2002). We found that both cultured and

    lavage-derived mesothelial cells implanted onto a

    peritoneal wound surface and underwent cell division

    with subsequent incorporation into the regeneratingmesothelium as demonstrated by cell junction forma-

    tion. Peritoneal macrophages also attached to injured

    areas but failed to incorporate whereas peritoneal fi-

    broblasts failed to attach, as did mesothelial cells to un-

    injured areas. This suggests that free-floating mesothe-

    lial cells are able to adhere to exposed and deposited

    ECM substrates such as collagen, fibronectin, vit-

    ronectin and possibly fibrin following injury, undergo

    cell division and integrate into the mesothelial layer.

    It is not known whether these free-floating cells are

    desquamated mesothelial cells from the serosal lining,

    a resident peritoneal fluid sub-population or a dedi-

    cated circulating precursor cell population. However,

    cell depletion studies using whole body X-irradiation

    (Whitaker & Papadimitriou, 1985; Venables, Ellis,

    & Burns, 1967) do not appear to support the claim

    that a bone marrow-derived precursor is involved in

    mesothelial healing, but this finding still needs to be

    confirmed.

    5. Adhesion formation

    Adhesions are a common consequence of serosal

    injury in all three serosal cavities leading to serious

    complications such as intestinal obstruction, chronic

    pain and infertility in women. A detailed histological

    and ultrastructural study of human peritoneal adhe-

    sions demonstrated that they were all well vascularised

    and innervated and contained clusters of smooth mus-

    cle cells, the origin of which was unclear (Herrick

    et al., 2000; Sulaiman et al., 2001).

    It has been proposed that adhesions form as a con-

    sequence of reduced fibrinolytic activity in serosal tis-sues. This has been shown both in human studies and

    genetically modified mouse models (Holmdahl et al.,

    1997; Sulaiman, Dawson, Laurent, Bellingan, &

    Herrick, 2002).In serosal tissue, mesothelial cells are

    the major source of PA, which are proteases essential

    to the fibrinolytic pathway (Sitter et al., 1995). If

    mesothelial healing is impaired, there is a reduction in

    local PA secretion which reduces fibrinolytic activity.

    Two major therapeutic approaches have been inves-

    tigated to prevent adhesion formation: fibrinolytic

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    8/22

    628 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    agents and barrier devices such as membranes and

    gels. However, due to complications associated with

    bleeding, systemic fibrinolysis, injury to the internal

    organs and vessels, impaired wound healing and dif-ficulty of application, these approaches have shown

    limited success. The future direction in preventing

    adhesions is likely to be the application of growth

    factors and mediators designed to increase the rate of

    serosal repair and so re-establish the tissues normal

    fibrinolytic capacity.

    Another approach to increase the rate of serosal re-

    pair is through the exogenous addition of mesothelial

    cells. Several groups have demonstrated that instilla-

    tion of autologous mesothelial cells at the time of in-

    jury prevents adhesion formation (Di Paolo, Vanni, &

    Sacchi, 1990; Bertram et al., 1999). Di Paolo et al.

    (1990)found that intraperitoneal (i.p.) injection of cul-

    tured autologous omental mesothelial cells in rabbits

    with staphylococcal-induced peritonitis significantly

    reduced the formation of adhesions. In a clinical study

    by the same group, four uremic peritoneal dialysis pa-

    tients recovering from severe peritonitis were injected

    i.p. with 3108 of their own mesothelial cells, previ-

    ously cultured and frozen. At laparoscopy 3 and 6 days

    post-implantation, there were morphological signs of

    cell incorporation in peritoneal biopsies suggesting

    this technique may have important applications for theprevention of adhesions in humans (Di Paolo et al.,

    1991). In a rat surgical model, Bertram et al. (1999)

    also found that i.p. injection of cultured autologous rat

    omental mesothelial cells immediately after abrasion

    of the peritoneum reduced the number of adhesions

    compared to the control group. It is assumed from

    these studies that the addition of exogenous mesothe-

    lial cells increased serosal repair so prevented adhe-

    sion formation, although this has not been confirmed.

    These findings again support the concept that a

    free-floating progenitor mesothelial cell is involved inmesothelial repair however, they also raise a number

    of important questions. For example, it is not clear

    whether the free-floating injected cells are different

    from the resident mesothelial cells of the serosal lin-

    ing, or if their differentiation state changes during

    culture or when introduced back into the peritoneal

    cavity. Furthermore, omental mesothelial cells may

    display phenotypic characteristics that are different

    from mesothelial cell populations present in other

    locations. Our studies demonstrated incorporation of

    free-floating mesothelial cells obtained from peri-

    toneal lavage and peritoneal wall into injured serosa

    (Foley-Comer et al., 2002), suggesting that omental

    mesothelial cells alone may not be the only cellsinvolved in mesothelial regeneration.

    The concept that these free-floating mesothelial pro-

    genitor cells may have stem cell-like qualities is sup-

    ported by the findings of Lucas, Warejcka, Zhang,

    Newman, and Young (1996). They isolated and cul-

    tured mesenchymal stem cells (MSCs) from skeletal

    muscle of neonatal rats and assessed their effect on

    the formation of peritoneal adhesions. They compared

    the implantation of different concentrations of MSCs

    with dead MSCs or smooth muscle cells isolated from

    adult animals. Cells were injected i.p. immediately

    following surgical injury or at 46 h post-surgery. Ad-

    hesion number was significantly reduced in the ani-

    mals receiving living MSCs at the time of surgery in

    a concentration dependent manner, whereas adhesion

    had increased in the animals receiving MSCs 46 h af-

    ter surgery. Dead MSCs and smooth muscle cells had

    no effect on adhesion formation compared with saline

    controls. The authors proposed that MSCs have the

    capacity to differentiate into mesothelial cells capable

    of repopulating injured serosa and so prevent adhesion

    formation. Alternatively, the MSCs produce factors

    that inhibit the formation of the initial fibrin-rich adhe-sions, such as fibrinolytic proteases or growth factors

    that stimulate mesothelial healing. Cells injected 46 h

    after injury are likely to have been trapped within de-

    posited fibrin and may have differentiated into fibrob-

    lasts rather than mesothelial cells, produced collagen

    and formed stronger and more extensive adhesions.

    Cell tracking studies were not performed in this study

    so the fate of the injected cells remains unknown. It

    is crucial that future studies elucidate the origin, state

    of differentiation and ultimate fate of resident adher-

    ent and free-floating serosal cells following injury todetermine the exact roles they play in normal and ab-

    normal mesothelial repair.

    6. Evidence for a multipotential subserosal

    mesenchymal cell

    Another popular theory as to the origin of the

    regenerating mesothelial cells is that they are de-

    rived from multipotential subserosal mesenchymal

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    9/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 629

    cells, which when appropriately stimulated, begin to

    differentiate into mesothelial cells while migrating

    to the injured surface. Many groups have described

    the presence of cells with epithelial-like characteris-tics in the subserosal layer of biopsies from various

    pathological conditions (Bolen et al., 1986; Davila

    & Crouch, 1993;Bolen, Hammar, & McNutt, 1987;

    Dobbie, 1990) and from experimental animal models

    (Johnson & Whitting, 1962; Yen et al., 1996; Buoro

    et al., 1993; Pampinella et al., 1996). These findings

    have customarily been explained by the theory that

    there exists a population of subserosal multipoten-

    tial cells with the ability to differentiate along both

    mesenchymal and mesothelial pathways; a concept

    originally suggested byKlemperer and Rabin (1931).

    IndeedRaftery (1973)described the involvement of a

    subserosal precursor cell in the repair of the mesothe-

    lium, that appeared intermediate in form between

    primitive mesenchymal cells on one hand and prolif-

    erating fibroblasts or endothelial cells on the other.

    Bolen et al. (1986, 1987)provided the best support for

    a multipotential subserosal cell using light, ultrastruc-

    tural and immunohistochemical techniques to exam-

    ine intermediate filament expression in reactive and

    non-reactive human serosal tissue. The group demon-

    strated that normal surface mesothelial cells express

    low and high molecular weight cytokeratins whereassubmesothelial cells express only vimentin. However,

    in biopsies from injured serosa, submesothelial cells

    lost vimentin immunoreactivity and progressively ac-

    quired high and low molecular weight cytokeratins. It

    was suggested that these cells were differentiating to-

    wards a mesothelial cell phenotype and were respon-

    sible for the re-establishment of surface mesothelium.

    However, Whitaker et al. (1992) in a similar study

    were unable to reproduce these findings and suggested

    that the staining pattern seen by Bolen and colleagues

    may be a result of mature mesothelial cells migrat-ing into the subserosal connective tissue. In another

    study, Amari, Taguchi, Iwahara, Shibuya, and Naoe

    (2002) reported that cultured spheroids composed of

    free-floating multicellular clusters of rat pleural fi-

    broblasts, demonstrated differentiation of surface cells

    into that consistent with mesothelial cells. These cells

    expressed microvilli, formed adherens junctions and

    were immunoreactive for cytokeratin. This change

    in phenotype was inhibited following incubation of

    spheroids with anti-fibroblast growth factor receptor

    antibody, suggesting FGF plays a key role in the

    phenotypic conversion of fibroblasts into regenerated

    mesothelial cells.

    Further support for a multipotential submesothe-lial cell comes from experimental findings following

    short-term bladder obstruction in a rabbit model. This

    form of injury induced thickening of the subserosal

    layer with smooth muscle hypertrophy, and a tran-

    sient expression of cytokeratin 18 in subserosal mes-

    enchymal cells. At a later stage, new muscle express-

    ing smooth muscle myosin and desmin, was detected

    in the subserosal layer in the absence of mitotic ac-

    tivity in the original smooth muscle layer (Pampinella

    et al., 1996).In agreement with their previous findings

    (Buoro et al., 1993),the authors concluded that resi-

    dent keratin expressing subserosal mesenchymal cells

    transformed into myofibroblasts and subsequently into

    fetal-type smooth muscle cells a well as regenerat-

    ing mesothelial cells (Buoro et al., 1993; Pampinella

    et al., 1996). Taken together these findings would seem

    to support the view that a multipotential subserosal

    mesenchymal cell exists which can differentiate into

    myofibroblasts and possibly smooth muscle cells as

    well as mesothelial cells. However, new evidence sug-

    gests that the mesothelial cells themselves may be

    multipotential and have the ability to differentiate into

    various different cell types. Indeed, irradiation and ki-netic studies have also questioned the role of sub-

    serosal cells for mesothelial regeneration (Whitaker &

    Papadimitriou, 1985; Mutsaers et al., 2000).

    7. Epithelial-mesenchymal transition of

    mesothelial cells

    Classically, isolated mesothelial cells from normal

    serosal tissue or fluid demonstrate cobblestone ep-

    ithelioid morphology in culture. However, it has longbeen known that these cells can change to a fibroblas-

    tic phenotype with repeated passage, reducing cytok-

    eratin and increasing vimentin expression (Mackay,

    Tracy, & Craighead, 1990). Various growth factors

    can also induce mesothelial cells to change pheno-

    type and express many of the characteristics associated

    with fibroblasts such as increased motility and en-

    hanced ECM production (Fig. 3). For example, EGF

    induces the reversible change to a fibroblastic pheno-

    type that is accompanied by an increased expression

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    10/22

    630 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    Fig. 3. Primary cultures of human pericardial mesothelial cells

    representing (A) epithelioid and (B) fibroblastic phenotypes, at dif-

    ferent passage numbers of the same cell preparation. Micrographs

    courtesy of Jason Tee.

    of1 integrins, in particular21, facilitating an en-

    hanced adhesion to and migration on collagen type

    I (Leavesley, Stanley, & Faull, 1999). Furthermore,

    EGF, PDGF and IL-1 beta have also been shown to

    stimulate increased collagen production in mesothe-

    lial cells (Harvey & Amlot, 1983; Owens & Milligan,

    1994;Yang, Kim, Lee, Park, & Kim, 1999).

    Various benign disorders, including liver cirrhosis,

    endometriosis or serosal inflammation, produce effu-

    sions that often contain increased numbers of mesothe-lial cells thought to be derived from the reactive serosa.

    In culture, these cells demonstrate both fibroblastic

    and epithelioid morphologies, a pattern which is stable

    throughout early passages (Gulyas, Dobra, & Hjerpe,

    1999).It has been suggested that these two different

    cell morphologies represent mesothelial cells at differ-

    ent stages of differentiation, and it is likely that in dis-

    ease, inflammatory factors and other mediators direct

    cells down various phenotypic pathways. The expres-

    sion of Wilms tumor susceptibility gene (WT1) and

    certain proteoglycans, syndecan-4 and glypican, are

    proposed to be associated with progression through the

    differentiation process (Dobra et al., 2000;Gulyas &

    Hjerpe, 1999, 2003) with WT1 often being describedas a mesothelial lineage marker.

    Whitaker et al. (1992) first suggested that mature

    mesothelial cells could transform into fibroblast-like

    cells in vivo and invade the underlying subserosal con-

    nective tissue. Indeed, they suggested that this could

    account for the intermediate filament staining pattern

    observed by Bolen et al. (1986). This seems an un-

    usual concept because in contrast to mesenchymal

    stromal cells, epithelial-like cells infrequently convert

    into fibroblasts in mature tissue, apart from during

    wound healing or tumour progression (Hay, 1995).

    However, two recent reports investigating the patho-

    logical effects of continuous ambulatory peritoneal

    dialysis (CAPD) have provided strong evidence to sup-

    port this concept. CAPD is known to cause peritoneal

    fibrosis leading to a failure of ultrafiltration however,

    the mechanisms involved in this process are not clear.

    In the first study,Yang, Chen, and Lin (2003)demon-

    strated that transforming growth factor-1 (TGF-1)

    induced human omental mesothelial cells to transdif-

    ferentiate into myofibroblasts in vitro with the char-

    acteristic appearance of prominent RER, conspicuous

    smooth muscle actin myofilaments, intermediate andgap junctions and active deposition of ECM. Gene ex-

    pression analysis revealed a complex modulation of

    gene expression involving cytoskeletal organisation,

    cell adhesion, ECM production, cell proliferation, in-

    nate immunity, stress responses and many other es-

    sential metabolic processes as the mesothelial cells

    underwent transformation. The authors proposed that

    the differentiated epithelial cells of the mesothelium

    convert into myofibroblasts and that the pathological

    features observed following CAPD may be due to the

    recruitment of fibrogenic cells from the mesotheliumduring serosal inflammation and wound healing.

    The second study byYnez-Mo et al. (2003)also

    demonstrated that human mesothelial cells undergo a

    conversion from an epithelial to mesenchymal phe-

    notype which occurred in patients following serosal

    injury. Peritoneal mesothelial cells isolated from dial-

    ysis fluid effluents displayed a mesenchymal pheno-

    type that appeared to be related to both the duration

    of CAPD and to whether peritonitis had occurred.

    Mesothelial cells lost their epithelial morphology and

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    11/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 631

    showed a decrease in the expression of cytokeratins

    and E-cadherin through induction of the transcrip-

    tional repressorsnail. They also acquired a migratory

    phenotype with up-regulation of 2 integrins. Ma-jor profibrotic and inflammatory cytokines, such as

    TGF-1 and IL-1B, appeared to be involved in this

    process. In addition, assessment of peritoneal biopsy

    specimens from patients undergoing CAPD showed

    the presence of mesothelial markers, ICAM-1 and cy-

    tokeratins, on fibroblast-like cells embedded in the

    subserosal layer, suggesting that these cells were de-

    rived from a local conversion of mesothelial cells.

    The authors described this phenotypic conversion

    as transdifferentiation, a complex and generally

    reversible process that starts with the disruption of

    intercellular junctions and loss of apical-basolateral

    polarity typical of epithelial cells. With time, the cells

    transform into fibroblast-like cells with pseudopodial

    protrusions and increased migratory, invasive and fi-

    brogenic features (Hay, 1995).However, it is currently

    unknown whether the mesothelial cells remain as

    myofibroblasts, continue to differentiate into smooth

    muscle cells or revert back to surface mesothelial

    cells. Furthermore, it is unclear whether the mesothe-

    lial cells that undergo trandifferentiation are a resident

    population in the mesothelial layer, originate from a

    serosal fluid subpopulation or are from a circulatingblood-derived source. Nevertheless, the authors do

    suggest that in light of these recent findings, the ear-

    lier concept of a multipotential subserosal cell being

    able to convert to both epithelial mesothelial cells and

    myofibroblasts (Raftery, 1973; Bolen et al., 1986)

    should be questioned. Furthermore, it raises the inter-

    esting possibility that mesothelial transdifferentiation

    may be wholly or partly responsible for the patholog-

    ical changes that occur in the serosal layer following

    trauma caused by, for example, CAPD, irradiation,

    malignancy or surgery.

    8. Tissue engineering potential of mesothelial cells

    Although there is a lack of information regarding the

    differentiation potential of mesothelial cells, for over

    a century these cells have been used to repair damaged

    tissues and organs, as well as being employed in a

    number of new tissue engineering applications.

    8.1. Vascular grafts

    Despite considerable clinical research, no biolog-

    ical or synthetic grafts have been developed as anideal substitute for small diameter arteries (Nerem &

    Seliktar, 2001). When acellular artificial prostheses

    are used in the reconstruction of small diameter ves-

    sels, failure frequently occurs because the luminal sur-

    face is thrombogenic resulting in thrombus formation

    and re-occlusion following implantation. Cell seeding

    should decrease thrombogenicity of implanted vas-

    cular grafts but this application is hampered by the

    limited availability of autologous vascular endothelial

    cells, and so alternative cell types have been sought.

    It has long been recognised that foreign objects

    introduced into the peritoneal cavity of the rat, rab-

    bit or mouse, initiate an inflammatory response with

    the resultant granulation tissue covered by a layer of

    mesothelium (Ryan et al., 1973; Campbell & Ryan,

    1983; Mosse, Campbell, & Ryan, 1985). Eskeland

    and Kjaerheim (1966) were first to demonstrate that

    a mesothelial membrane could be grown on the outer

    surface of a free-floating diffusion chamber placed

    in the peritoneal cavity of rats. Later ultrastructural

    studies showed that mesothelial cells deposited and

    organised ECM; including thick collagen fibres, the

    amorphous components of elastic fibres and base-ment membrane-like structures restricted to the basal

    region of the cell layer (Rennard et al., 1984). Based

    on these observations, in addition to the known fibri-

    nolytic and antithrombotic properties of mesothelial

    cells (Louagie et al., 1986), Clarke, Pittilo, Machin,

    and Woolf (1984)proposed that autologous mesothe-

    lial cells may represent a practical alternative to

    endothelial cells in vascular grafts. Subsequently,

    many groups have investigated the efficacy of using

    mesothelial cells, mainly derived from the omentum,

    as endothelial replacements (Sparks et al., 2002; Bullet al., 1988; Bearn et al., 1992; Verhagen et al., 1998;

    Theuer et al., 1996).

    Studies by Bull et al. (1988) showed that Dacron

    arterial grafts seeded with autologous mesothe-

    lial cells promoted luminal cell cover, displayed

    anti-thrombogenic activity, inhibited platelet aggre-

    gation and released more prostacyclin than unseeded

    grafts in canine abdominal aorta replacements. How-

    ever, later studies using digested omental extract

    seeded onto knitted Dacron scaffolds and implanted

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    12/22

    632 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    as bilateral femoral artery replacements, suggested

    that the mesothelial cells were not retained on the

    graft 24h later (Bearn et al., 1992). Furthermore,

    fibronectin coated small diameter polytetrafluoroethy-lene (PTFE) scaffolds seeded with cultured omental

    mesothelial cells showed poor patency and increased

    neointimal thickening compared with non-seeded

    grafts following implantation into the carotid artery

    of the same dog (Verhagen et al., 1998). Other studies

    in which the infrarenal inferior vena cava was re-

    placed with interposition grafts of either a peritoneal

    tube, PTFE or PTFE lined peritoneum, demonstrated

    that peritoneal lined grafts maintained a continuous

    circumferential cellular lining but showed no im-

    provement in short term patency compared to PTFE

    alone (Theuer et al., 1996).

    Despite these disappointing findings, Campbell,

    Efendy, and Campbell (1999) using an alternative

    seeding method, have produced more favourable re-

    sults. Free-floating silastic tubing was implanted into

    the peritoneal cavity of rats and rabbits and after two

    weeks, the ones that remained free-floating were re-

    moved and processed. When the tubes were everted

    and histologically assessed they consisted of an in-

    tima of non-thrombotic mesothelial cells, a media of

    smooth muscle-like cells or myofibroblasts embedded

    in a collagen and elastic matrix, and an outer collage-nous adventitia. The grafts remained patent, showed

    reasonable tensile strength and were responsive to

    contractile agonists for at least 4 months. The role

    of haemodynamic stress, active stretch and neuronal

    imput on the differentiation of the cells within the

    mesothelial tubes was investigated in a subsequent

    study. Following end-to-end anastomosis with the

    aorta, there was a progressive increase in myofilament

    expression (evidence of smooth muscle phenotype) in

    the grafts over time, which was also observed by cycli-

    cally stretching the tubes in vitro (Efendy, Campbell,& Campbell, 2000). In contrast, innervation of the

    tubes following transplantation into the rat anterior

    eye chamber appeared to have little effect on the

    differentiation of cells towards a smooth muscle cell

    phenotype. The authors state that these grafts have

    several advantages over others in that they are biocom-

    patible with the host tissue, need no artificial mesh as

    part of the wall, have a nonthrombogenic surface and

    develop elastic lamellae. Moreover, they have demon-

    strated patency for at least 4 months with 1020%

    contractile responses compared with the control artery

    after transplantation. However, many questions re-

    main unanswered such as; how similar the inner sur-

    face lining of mesothelial cells are to true endothelialcells, and are mesothelial cells in the intimal layer

    subsequently replaced by local ingrowth of endothe-

    lial cells following transplantation to high pressure

    arterial sites. Indeed, if the free-floating mesothelial

    cells of the peritoneal cavity are able to provide all

    the cell types found in the transplanted graft, is this

    through a transdifferentiation process as described

    previously?

    Many authors remain to be convinced of the use

    of the peritoneal cavity as a feasible environment for

    growing functional bioartificial vascular grafts as re-

    viewed byMoldovan and Havemann (2002).Cebotari,

    Walles, Sorrentino, Haverich, and Mertsching (2002)

    repeated the work of Campbell et al. (1999) using

    decellularised allogenic scaffolds and, although they

    found repopulation of the implanted grafts in the

    given time period, they also showed extensive denat-

    uration of collagen and graft degeneration. Whether

    prior seeding vascular scaffolds with mesothelial cells

    isolated from the omentum (Pearce et al., 1987; Pasic

    et al., 1994; Salacinski, Punshon, Krijgsman,

    Hamilton, & Seifalian, 2001) or peritoneal fluid

    (Tiwari et al., 2003) is a better method for generatingtissue engineered grafts, awaits further investigation.

    Until then, the use of mesothelial cells as endothe-

    lial cell replacements still remains a possibility and

    may prove important in, for example, the develop-

    ment of autologous coronary artery bypass grafts

    or arteriovenous access fistulae for hemodialysis

    patients.

    8.2. Omental grafts

    The scientific community has neglected the omen-

    tum for many years, although recent interest has

    stemmed from its multiple uses in reconstructive

    surgery (Liebermann-Meffert, 2000). The omentum

    is essentially composed of two mesothelial sheets

    which enclose predominately adipocytes embed-

    ded in a highly vascularised connective tissue. The

    greater part of the omentum is associated with the

    stomach, small intestines and transverse colon and

    forms an apron-like structure covering abdominal

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    13/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 633

    organs. The omentum is particularly susceptible to

    forming adhesions as it floats passively within the

    peritoneal cavity but rapidly adheres to inflamed or

    damaged tissues. In a post-mortem study, Weibel andManjo (1973) found that the omentum was the or-

    gan most frequently involved in adhesion formation

    and many workers have suggested that omental ad-

    hesions offer protection against more severe com-

    plications such as peritonitis and ischaemic bowel

    disease (Williams & White, 1986; Hasgood, 1990).

    Part of the omentums ability to rescue injured tis-

    sue is likely to be due to its angiogenic (Goldsmith,

    Griffith, Kupferman, & Catsimpoolas, 1984; Gold-

    smith, Griffith, & Catsimpoolas, 1986) and neu-

    rotrophic (Chamorro et al., 1993) properties; hence,

    its use as a pedicle graft tissue for clinical condi-

    tions involving revascularisation of ischaemic parts

    of the brain, kidney, spleen, heart and spinal cord

    (Goldsmith, Chen, & Duckett, 1973; Goldsmith,

    Duckett, & Chen, 1975).

    Free omental grafts have been used in the treatment

    of numerous human disorders including neurodegen-

    erative diseases such as Alzheimers disease, chronic

    leg ulcers and gastric ulcers (Weinzweig, Schlechter,

    Baraniewski, & Schuler, 1997). Piano et al. (1998)

    used free omental grafts to treat severe necrotising

    fasciitis and observed that necrotic tissue becamerevascularised resulting in acceptance of the graft

    and healing of the defect. The exact mechanism of

    this early revascularisation is unknown, however, it

    has been suggested that various growth factors such

    as FGF (Chamorro et al., 1993) and VEGF (Zhang

    et al., 1997; Mandl-Weber, Cohen, Haslinger,

    Kretzler, & Sitter, 2002), which are present in high

    levels and can be isolated from the omentum, are in-

    volved. In another study,Chamorro et al. (1993)used

    free omental grafts to facilitate nerve graft regenera-

    tion in rats by surrounding the nerve graft with omen-tum. Early revascularisation and directional growth

    of sprouting axons was encouraged, thus increasing

    the efficiency of nerve regeneration. It is worth noting

    that the fate and role of the mesothelial cells was not

    determined in any of these studies and therefore it is

    not clear whether they were in part responsible for

    the success of these grafts, through either the release

    of growth factors or themselves being incorporated

    into the repairing tissue.

    8.3. Nerve grafts

    Regeneration of severed peripheral nerves is of-

    ten incomplete due to loss or misdirection of nervefibres and neuroma formation. The use of nerve re-

    placements composed of artificial tubes seeded with

    isolated mesothelial cells as an alternative to primary

    nerve suture has been introduced as a biological ap-

    proach to nerve injuries. Initial studies in a rat model

    by Lundborg et al. (1982) investigated the regen-

    eration of a transected sciatic nerve through either

    preformed mesothelial chambers or autologous nerve

    grafts bridging a 10 mm gap. Within the mesothelial

    chambers, an organised multifascicular nerve trunk

    formed between proximal and distal stumps. After 3

    months there was no difference with respect to ax-

    onal density or distribution of axons between the two

    grafts. Furthermore, the conduction velocities across

    the gaps were similar. In the mesothelial chambers,

    the regenerating nerve was surrounded by a loose

    cellular stroma and a small amount of interstitial

    fluid, which was found to contain trophic activity for

    cultured rodent sensory neurons.

    In a subsequent study, nerve regrowth occurred

    when a preformed mesothelial tube bridged the gap

    between left and right sciatic nerves that had been

    transferred to the backs of rats (Danielsen, Dahlin,Lee, & Lundborg, 1983). When the gap was 10 mm or

    less, a well developed nerve structure was generated

    in the chamber between the nerve ends, and axons

    from the left sciatic nerve reinnervated muscles in the

    right limb via the right sciatic nerve. Additional stud-

    ies demonstrated that when rabbit hypoglossal nerves

    were repaired using mesothelial chambers, a sig-

    nificantly faster migration of radio-labelled proteins

    in the distal nerve segment was observed compared

    to sutured nerves (Danielsen, Lundborg, & Frizell,

    1986).Remarkably, the thin mesothelial lining foundaround the tube lacked primary inflammatory signs

    at follow-up after 1 year and showed no signs of

    compression (Dahlin & Lundborg, 2001). Similar to

    the studies ofChamorro et al. (1993),Castaneda and

    Kinne (2002) performed siatic nerve transections in

    rats and found that 2530 mm defects bridged by an

    omental graft were fully healed with increased func-

    tional recovery and less scarring than end to end repair.

    It was suggested that grafts incorporating mesothelial

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    14/22

    634 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    cells may have an advantage as they allow sliding of

    the repair site against surrounding tissues due to the

    secretion of surfactants (Dahlin & Lundborg, 2001).

    Although the origin, fate or function of the mesothe-lial cells was not described in these studies, artificial

    tubes lined by mesothelial cells appear to be impor-

    tant alternatives to conventional repair techniques for

    primary nerve repair and reconstruction of segmental

    defects.

    9. Does a mesothelial stem cell exist?

    The biology of adult stem cells remains remark-

    ably poorly understood and in general, there is a lack

    of a unifying definition as well as specific markers

    to define them. A rich reservoir of adult stem cells

    resides in specific niches within the bone marrow mi-

    croenvironment as well as in a variety of connective

    tissues, where they are maintained in an undiffer-

    entiated and quiescent state. The ability to produce

    cells that can progress down a variety of distinct

    cell lineages, even as clonally isolated cells, is one

    of the main characteristics of stem cells. For exam-

    ple, when appropriately induced, mesenchymal stem

    cells (MSCs) have the potential to differentiate along

    specific mesenchymal lineages (multipotency) andform tissues that include endothelium, muscle, bone,

    cartilage and fat (reviewed by Tuan, Boland & Tuli,

    2003). Although a mesothelial stem cell has not been

    identified, growing evidence based on its primitive

    embryological origin and ability to transdifferenti-

    ate strongly supports the idea that a population of

    mesothelial progenitor cells exist. Indeed,Donna and

    Betta (1986)proposed that the mesothelial cell was

    not only totipotent but represented real mesoderm that

    retained the potential to differentiate along embryonic

    developmental lines including to cartilage and bone.Thus, they suggested the term mesoderma instead

    of mesothelioma to recognise the mesodermal ori-

    gin of associated mesothelial tumours. Since then, as

    previously described, tissue culture and animal exper-

    imental studies have convincingly demonstrated that

    adult mesothelial cells are capable of transdifferenti-

    ating from an epithelial to mesenchymal phenotype

    and this seems to depend on the presence of cer-

    tain growth factors or cytokines (Yang et al., 2003;

    Yanez-Mo et al., 2003). However, conclusive evidence

    demonstrating that adult human mesothelial cells are

    capable of differentiating along specific mesenchymal

    cell lineages is still lacking.

    Munoz-Chapuli et al. (1999)recently hypothesisedthat hemangioblasts, the common progenitor of the

    endothelial and hematopoietic cell lineages, originated

    from embryonic splanchic mesothelium, and that the

    differentiation of endothelial and blood cells was

    therefore from a common mesothelial-derived progen-

    itor. A later study provided evidence to support this

    theory. Using cell-labelling techniques and quail-chick

    chimeras,Perez-Pomares and Munoz-Chapuli (2002)

    showed that during development, epicardial mesothe-

    lium differentiates into endothelium or smooth muscle

    through an epithelial-mesenchymal transition (EMT)

    process. This finding raised the interesting question

    of whether the coelomic mesothelium retains its abil-

    ity to transform into multipotent mesenchymal cells

    in the adult. Based on this assumption, Wada, Osler,

    Reese, and Bader (2003) recently showed that in

    culture, explants of adult rat epicardial mesothelium

    retain the ability to produce mesenchyme including

    smooth muscle cells in response to specific growth

    factors. The authors suggest that a cell line derived

    from rat epicardial mesothelial cells acts in a sim-

    ilar manner to the bipotential vascular progenitor

    cells, a stem cell population originally described byYamashita et al. (2000).

    As well as the intriguing possibility that adult

    mesothelial progenitor cells are able to produce en-

    dothelium and smooth muscle, findings from several

    experimental models suggest that these cells may also

    form skeletal muscle and cartilage. For instance, dur-

    ing the healing phase of a chemical-induced peritoni-

    tis, skeletal muscle fibres were found to develop de

    novo in the peritoneal lining of the adult rat diaphragm.

    The location and orientation of the fibres suggested

    an origin from mesothelial or submesothelial cellsin granulation tissue rather than intrinsic diaphrag-

    matic muscle satellite cells (Levine & Saltzman,

    1994; Drakontides, Danon, & Levine, 1999). How-

    ever, more extensive studies are required to confirm

    these findings. If the mesothelium is the source of new

    skeletal muscle fibres, as the authors state, it will be

    important to determine if the diaphragmatic mesothe-

    lium is different from mesothelium in other locations.

    Indeed, it would be desirable to imitate the environ-

    ment of the inflamed diaphragmatic peritoneum in

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    15/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 635

    other areas of skeletal muscle damage where regener-

    ation is needed.

    Although rare, it is of no surprise that biopsies taken

    from human malignant mesothelioma express markersof osseous and cartilaginous differentiation (Donna

    & Betta, 1986; Yousem & Hochholzer, 1987; Kiyo-

    zuka et al., 1999; Andrion, Mazzucco, Bernardi, &

    Mollo, 1989). Furthermore, in experimental models,

    bone and cartilage were found in peritoneal malig-

    nant mesotheliomas that were induced by i.p. injec-

    tion of asbestos fibres (Rittinghausen, Ernst, Muhle,

    & Mohr, 1992). Surprisingly, however, Fadare,

    Bifulco, Carter and Parkash (2002) found evidence

    of cartilaginous differentiation in human peritoneal

    tissue biopsies which did not appear to be associated

    with an intra-abdominal malignancy. Indeed, in the

    human peritoneum, several other well-documented

    cases of mesenteric heterotopic ossification (or os-

    seous metaplasia) and/or cartilaginous differentiation

    have been reported (Lemeshev, Lahr, Denton, Kent, &

    Diethelm, 1983; Wilson, Montague, Salcuni, Bordi, &

    Rosai, 1999; Yannopoulos, Katz, Flesher, Geller,

    & Berroya, 1992).The source of the cells that undergo

    this differentiation process remains controversial but

    the traditional view is that they are derived from a

    population of subserosal multipotential cells as de-

    scribed earlier. However, in light of recent findings,it is also possible that a population of mesothelial

    cells may have the ability to form cells of different

    mesenchymal lineages. These progenitor cells may

    be resident in the mesothelial layer, free-floating in

    the serosal fluid or alternatively, may be derived from

    a circulating multipotential cell population which en-

    ters serosal cavities via the vasculature. This matter is

    further complicated by the observation that cells of a

    haemopoietic origin, identified through bone marrow

    transplant and Ly5 antigen expression, are able to

    differentiate into myofibroblasts and smooth musclecells in response to a foreign body implanted into the

    peritoneal cavity (Campbell, Efendy, Han, Girjes, &

    Campbell, 2000). Depending on the local environ-

    ment these progenitor cells may be able to progress

    down various differentiation pathways. Growth fac-

    tors levels, cellcell interactions, cell density and

    physical and mechanical stimuli may all contribute

    to the end product of differentiation. In addition, the

    mesothelial layer and free-floating cells are in con-

    tinuous communication with peritoneal fluid and so

    Fig. 4. Hypothetical representation of a mesothelial progenitor

    cell residing in the serosal monolayer. Following injury, these

    cells may transdifferentiate into subserosal progenitor cells with

    the capacity to further differentiate into myofibroblasts, smooth

    muscle cells and endothelial cells. In addition, they may detach

    from the basement membrane and become free-floating progenitor

    cells in the serosal fluid before repopulating serosal lesions.

    any changes in, for example, levels of cytokines and

    growth factors, proteases, oxygen, and pH, may also

    affect ultimate progenitor cell fate (Fig. 4).

    As well as an ability to differentiate along specific

    lineages upon stimulation, other key features of stem

    cells are to remain in a quiescent undifferentiated state

    until provided with the signal to divide asymmetri-

    cally and undergo many more replicative cycles than

    normal. Future studies have yet to determine if theseare characteristics of mesothelial progenitor cells. At

    present, little is know regarding aspects of ageing or

    senescence of mesothelial cells, except that in culture

    they senesce 2.5-fold faster than fibroblasts and in vivo

    senesce following exposure to dialysis fluids (Thomas

    et al., 1997; Gotloib, Wajsbrot, & Shostak, 2003).

    Furthermore, whether serosal fluid contains survival

    factors that allow mesothelial progenitors to remain

    viable and proliferative or if it contains chemoattrac-

    tants that cause circulating progenitor cells to home to

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    16/22

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    17/22

    S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642 637

    sheeps visceral pleura. American Journal of Anatomy, 165(3),

    277294.

    Amari, M., Taguchi, K., Iwahara, M., Shibuya, K., & Naoe, S.

    (2002). Immunohistochemical and ultrastructural study on effect

    of fibroblast growth factor on transformation of fibroblasts toregenerated mesothelial cells. Medical Electron Microscopy,

    35(4), 225233.

    Andrion, A., Mazzucco, G., Bernardi, P., & Mollo, F. (1989).

    Sarcomatous tumor of the chest wall with osteochondroid

    differentiation. Evidence of a mesodermal origin. The American

    Journal of Surgical Pathology, 13, 707712.

    Baradi, A. F., & Campbell Jr., W. G. (1974). Exudative

    peritonitis induced in mice by bovine serum albumin. Archives

    of Pathology, 97(1), 212.

    Baradi, A. F., & Rao, S. N. (1976). A scanning electron microscope

    study of mouse peritoneal mesothelium. Tissue & Cell, 8(1),

    159162.

    Bearn, P. E., Miller, K., Bull, H., Seddon, A. M., McCollum,C. N., & Marston, A. (1992). An immunohistochemical study

    of mesothelial cell seeding for knitted Dacron. International

    Journal of Experimental Pathology, 73(6), 801808.

    Bellingan, G. J., Xu, P., Cooksley, H., Cauldwell, H., Shock, A.,

    & Bottoms, S. et al., (2002). Adhesion molecule-dependent

    mechanisms regulate the rate of macrophage clearance during

    the resolution of peritoneal inflammation. The Journal of

    Experimental Medicine, 196(11), 15151521.

    Bertram, P., Tietze, L., Hoopmann, M., Treutner, K. H.,

    Mittermayer, C., & Schumpelick, V. (1999). Intraperitoneal

    transplantation of isologous mesothelial cells for prevention of

    adhesions.The European Journal of Surgery, 165(7), 705709.

    Betjes, M. G., Tuk, C. W., Struijk, D. G., Krediet, R. T.,Arisz, L., & Hart, M. et al., (1993). Interleukin-8 production

    by human peritoneal mesothelial cells in response to tumor

    necrosis factor-alpha, interleukin-1, and medium conditioned

    by macrophages cocultured with Staphylococcus epidermidis.

    The Journal of Infectious Diseases, 168(5), 12021210.

    Bianco, P., & Robey, P. G. (2001). Stem cells in tissue engineering.

    Nature, 414(6859), 118121.

    Bolen, J. W., Hammar, S. P., & McNutt, M. A. (1986). Reactive and

    neoplastic serosal tissue. A light-microscopic, ultrastructural,

    and immunocytochemical study. The American Journal of

    Surgical Pathology, 10(1), 3447.

    Bolen, J. W., Hammar, S. P., & McNutt, M. A. (1987).

    Serosal tissue: reactive tissue as a model for understanding

    mesotheliomas. Ultrastructural Pathology, 11(23), 251262.

    Bridges, J. B., & Whitting, H. W. (1964). Parietal peritoneal healing

    in the rat. Journal of Pathology and Bacteriology, 87, 123.

    Bull, H. A., Pittilo, R. M., Drury, J., Pollock, J. G., Clarke, J. M.,

    & Woolf, N. et al., (1988). Effects of autologous mesothelial

    cell seeding on prostacyclin production within Dacron arterial

    prostheses. The British Journal of Surgery, 75(7), 671674.

    Buoro, S., Ferrarese, P., Chiavegato, A., Roelofs, M., Scatena,

    M., & Pauletto, P. et al., (1993). Myofibroblast-derived

    smooth muscle cells during remodelling of rabbit urinary

    bladder wall induced by partial outflow obstruction. Laboratory

    Investigation, 69(5), 589602.

    Cameron, G. R., Hassan, S. M., & De, S. N. (1957). Repair of

    Glissons capsule after tangential wounds of the liver. Journal

    of Pathology Bacteriology, 73, 110.

    Campbell, G. R., & Ryan, G. B. (1983). Origin of myofibroblasts in

    the avascular capsule around free-floating intraperitoneal bloodclots. Pathology, 15(3), 253264.

    Campbell, J. H., Efendy, J. L., & Campbell, G. R. (1999). Novel

    vascular graft grown within recipients own peritoneal cavity.

    Circulation Research, 85(12), 11731178.

    Campbell, J. H., Efendy, J. L., Han, C.-L., Girjes, A. A., &

    Campbell, G. R. (2000). Haemopoietic origin of myofibroblasts

    formed in the peritoneal cavity in response to a foreign body.

    Journal of Vascular Research, 37, 364371.

    Casey, R. C., & Skubitz, A. P. (2000). CD44 and beta1 integrins

    mediate ovarian carcinoma cell migration toward extracellular

    matrix proteins. Clinical & Experimental Metastasis, 18(1),

    6775.

    Castaneda, F., & Kinne, R. K. (2002). Omental graft improvesfunctional recovery of transected peripheral nerve. Muscle

    Nerve, 26(4), 527532.

    Catterall, J. B., Jones, L. M., & Turner, G. A. (1999). Membrane

    protein glycosylation and CD44 content in the adhesion

    of human ovarian cancer cells to hyaluronan. Clinical &

    Experimental Metastasis, 17(7), 583591.

    Cebotari, S., Walles, T., Sorrentino, S., Haverich, A., & Mertsching,

    H. (2002). Guided tissue regeneration of vascular grafts in the

    peritoneal cavity. Circulation Research, 90(8), e71.

    Chamorro, M., Carceller, F., Llanos, C., Rodriguez-Alvarino, A.,

    Colmenero, C., & Burgueno, M. (1993). The effect of omental

    wrapping on nerve graft regeneration. British Journal of Plastic

    Surgery, 46(5), 426429.Chang, K., & Pastan, I. (1996). Molecular cloning of mesothelin, a

    differentiation antigen present on mesothelium, mesotheliomas,

    and ovarian cancers. Proceedings of the National Academy of

    Sciences of the United States of America, 93(1), 136140.

    Chung-Welch, N., Patton, W. F., Shepro, D., & Cambria, R.

    P. (1997). Human omental microvascular endothelial and

    mesothelial cells: characterization of two distinct mesodermally

    derived epithelial cells. Microvascular Research, 54(2), 108

    120.

    Clarke, J. M., Pittilo, R. M., Machin, S. J., & Woolf, N. (1984).

    A study of the possible role of mesothelium as a surface for

    flowing blood. Thrombosis and Haemostasis, 51(1), 5760.

    Cleaver, C. L., Hopkins, A. D., Ng Nee Kwong, K. C., & Raftery,

    A. T. (1974). The effect of postoperative peritoneal lavage

    on survival, peritoneal wound healing and adhesion formation

    following fecal peritonitis: an experimental study in the rat.

    The British Journal of Surgery, 61, 601604.

    Cunliffe, W. J., & Sugarbaker, P. H. (1989). Gastrointestinal

    malignancy: rationale for adjuvant therapy using early

    postoperative intraperitoneal chemotherapy.The British Journal

    of Surgery, 76(10), 10821090.

    Czernobilsky, B., Moll, R., Levy, R., & Franke, W. W.

    (1985). Co-expression of cytokeratin and vimentin filaments in

    mesothelial, granulosa and rete ovarii cells of the human ovary.

    European Journal of Cell Biology, 37, 175190.

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    18/22

    638 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    Dahlin, L. B., & Lundborg, G. (2001). Use of tubes in peripheral

    nerve repair. Neurosurgery Clinics of North America, 12(2),

    341352.

    Danielsen, N., Dahlin, L. B., Lee, Y. F., & Lundborg, G. (1983).

    Axonal growth in mesothelial chambers. The role of thedistal nerve segment. Scandinavian Journal of Plastic and

    Reconstructive Surgery and Hand Surgery, 17(2), 119125.

    Danielsen, N., Lundborg, G., & Frizell, M. (1986). Nerve repair

    and axonal transport: outgrowth delay and regeneration rate

    after transection and repair of rabbit hypoglossal nerve. Brain

    Research, 376(1), 125132.

    Davila, R. M., & Crouch, E. C. (1993). Role of mesothelial and

    submesothelial stromal cells in matrix remodeling following

    pleural injury.American Journal of Pathology,142(2), 547555.

    Di Paolo, N., Vanni, L., & Sacchi, G. (1990). Autologous

    implant of peritoneal mesothelium in rabbits and man. Clinical

    Nephrology, 34(4), 179184.

    Di Paolo, N., Sacchi, G., Vanni, L., Corazzi, S., Terrana, B.,

    Rossi, P., Gaggiotti, E., & Buoncristiani, U. (1991). Autologous

    peritoneal mesothelial cell implant in rabbits and peritoneal

    dialysis patients. Nephron, 57(3), 323331.

    Dobbie, J. W. (1990). New concepts in molecular biology and

    ultrastructural pathology of the peritoneum: their significance

    for peritoneal dialysis. American Journal of Kidney Diseases,

    15(2), 97109.

    Dobra, K., Andang, M., Syrokou, A., Karamanos, N. K., & Hjerpe,

    A. (2000). Differentiation of mesothelioma cells is influenced by

    the expression of proteoglycans. Experimental Cell Research,

    258(1), 1222.

    Dondelinger, R. F., Boverie, J. H., & Cornet, O. (1982). Diagnosis

    of pancreatic injury: a need to improve performance. JBR-BTR,

    83(4), 160166.

    Donna, A., & Betta, P. G. (1986). Differentiation towards cartilage

    and bone in a primary tumour of pleura. Further evidence in

    support of the concept of mesodermoma.Histopathology,10(1),

    101108.

    Donna, A., Betta, P. G., Chiodera, P., Bellingeri, D., Libener, R.,

    & Zorzi, F. et al., (1997). Newly marketed tissue markers for

    malignant mesothelioma: immunoreactivity of rabbit AMAD-2

    antiserum compared with monoclonal antibody HBME-1 and

    a review of the literature on so-called antimesothelioma

    antibodies. Human Pathology, 28(8), 929937.

    Drakontides, A. B., Danon, M. J., & Levine, S. (1999). Heterotopic

    neogenesis of skeletal muscle induced in the adult rat

    diaphragmatic peritoneum: ultrastructural and transplantation

    studies. Histology and Histopathology, 14(4), 11351143.Efendy, J. L., Campbell, G. R., & Campbell, J. H. (2000).

    The effect of environmental cues on the differentiation of

    myofibroblasts in peritoneal granulation tissue. The Journal of

    Pathology, 192(2), 257262.

    Ellis, H., Harrison, W., & Tugh, T. B. (1965). The healing of

    peritoneum under normal and pathological conditions. The

    British Journal of Surgery, 52, 471476.

    Eskeland, G., & Kjaerheim, A. (1966). Regeneration of parietal

    peritoneum in rats. 2. An electron microscopical study. Acta

    Pathologica et Microbiologica Scandinavica, 68(3), 379395.

    Fadare, O., Bifulco, C., Carter, D., & Parkash, V. (2002).

    Cartilaginous differentiation in peritoneal tissues: a report of

    two cases and a review of the literature. Modern Pathology,

    15(7), 777780.

    Faris, R. A., McBride, A., Yang, L., Affigne, S., Walker, C., & Cha,

    C. J. (1994). Isolation, propagation, and characterization of rat

    liver serosal mesothelial cells. American Journal of Pathology,145(6), 14321443.

    Fetsch, P. A., Simsir, A., & Abati, A. (2001). Comparison

    of antibodies to HBME-1 and calretinin for the detection

    of mesothelial cells in effusion cytology. Diagnostic

    Cytopathology, 25(3), 158161.

    Foley-Comer, A. J., Herrick, S. E., Al-Mishlab, T., Prele, C.

    M., Laurent, G. J., & Mutsaers, S. E. (2002). Evidence for

    incorporation of free-floating mesothelial cells as a mechanism

    of serosal healing. Journal of Cell Science, 115(Pt 7), 1383

    1389.

    Fotev, Z., Whitaker, D., & Papadimitriou, J. M. (1987). Role of

    macrophages in mesothelial healing. The Journal of Pathology,

    151(3), 209219.Fukata, H. (1963). Electron microscopic study on normal rat

    peritoneal mesothelium and its changes in absorption of

    particulate iron dextran complex. Acta Pathologica Japonica,

    13, 309325.

    Fusco, V., Ardizzoni, A., Merlo, F., Cinquegrana, A., Faravelli, B.,

    & De Palma, M. et al., (1993). Malignant pleural mesothelioma.

    Multivariate analysis of prognostic factors on 113 patients.

    Anticancer Research, 13, 683689.

    Goldsmith, H. S., Chen, W. F., & Duckett, S. W. (1973). Brain

    vascularization by intact omentum. Archives of Surgery,106(5),

    695698.

    Goldsmith, H. S., Duckett, S., & Chen, W. F. (1975). Prevention

    of cerebral infarction in the dog by intact omentum. American

    Journal of Surgery, 130(3), 317320.Goldsmith, H. S., Griffith, A. L., Kupferman, A., & Catsimpoolas,

    N. (1984). Lipid angiogenic factor from omentum. The Journal

    of the American Medical Association, 252(15), 20342036.

    Goldsmith, H. S., Griffith, A. L., & Catsimpoolas, N. (1986).

    Increased vascular perfusion after administration of an omental

    lipid fraction.Surgical Gynecology and Obstetrics,162(6), 579

    583.

    Goodell, M. A. (2001). Stem cells: is there a future in plastics?

    Current Opinion in Cell Biology, 13(6), 662665.

    Gotloib, L., Wajsbrot, V., & Shostak, A. (2003). Icodextrin-induced

    lipid peroxidation disrupts the mesothelial cell cycle engine.

    Free Radical Biology & Medicine, 34(4), 419428.

    Gulyas, M., Dobra, K., & Hjerpe, A. (1999). Expression of genes

    coding for proteoglycans and Wilms tumour susceptibility gene

    1 (WT1) by variously differentiated benign human mesothelial

    cells. Differentiation, 65(2), 8996.

    Gulyas, M., & Hjerpe, A. (2003). Proteoglycans and WT1

    as markers for distinguishing adenocarcinoma, epithelioid

    mesothelioma, and benign mesothelium. The Journal of

    Pathology, 199(4), 479487.

    Haney, A. F. (1993). Endometriosis, macrophages, and adhesions.

    Progress in Clinical and Biological Research, 381, 1944.

    Harada, N., Mizoi, T., Kinouchi, M., Hoshi, K., Ishii, S., &

    Shiiba, K. et al., (2001). Introduction of antisense CD44S

    CDNA down-regulates expression of overall CD44 isoforms

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    19/22

  • 8/13/2019 Mesothelial Progenitor Cells and Their Potential

    20/22

    640 S.E. Herrick, S.E. Mutsaers/ The International Journal of Biochemistry & Cell Biology 36 (2004) 621642

    Mandl-Weber, S., Cohen, C. D., Haslinger, B., Kretzler, M., &

    Sitter, T. (2002). Vascular endothelial growth factor production

    and regulation in human peritoneal mesothelial cells. Kidney

    International, 61(2), 570578.

    Marshall, B. C., Santana, A., Xu, Q. P., Petersen, M. J., Campbell,E. J., & Hoidal, J. R. et al., (1993). Metalloproteinases

    and tissue inhibitor of metalloproteinases in mesothelial

    cells. Cellular differentiation influences expression. Journal of

    Clinical Investigation, 91(4), 17921799.

    Martin, P., Hopkinson-Woolley, J., & McCluskey, J. (1992). Growth

    factors and cutaneous wound repair. Progress in Growth Factor

    Research, 4(1), 2544.

    Minot, C. S. (1890). The mesoderm and the coelom of vertebrates.

    The American Naturalist, 24, 877898.

    Moldovan, N. I., & Havemann, K. (2002). Transdifferentiation,

    a potential mechanism for covering vascular grafts grown

    within recipients peritoneal cavity with endothelial-like cells.

    Circulation Research, 91(3), e1.Mosse, P. R., Campbell, G. R., & Ryan, G. B. (1985). A

    comparison of the avascular capsule surrounding free floating

    intraperitoneal blood clots in mice and rabbits. Pathology,17(3),

    401407.

    Muir, T. E., Cheville, J. C., & Lager, D. J. (2001). Metanephric

    adenoma, nephrogenic rests, and Wilms tumor: a histologic

    and immunophenotypic comparison. The American Journal of

    Surgical Pathology, 25, 12901296.

    Munoz-Chapuli, R., Perez-Pomares, J. M., Macias, D., Garcia-

    Garrido, L., Carmona, R., & Gonzalez, M. (1999).

    Differentiation of hemangioblasts from embryonic mesothelial

    cells? A model on the origin of the vertebrate cardiovascular

    system. Differentiation, 64(3), 133141.

    Mutsaers, S. E. (2002). Mesothelial cells: their structure, functionand role in serosal repair. Respirology, 7(3), 171191.

    Mutsaers, S. E., Whitaker, D., & Papadimitriou, J. M. (1996).

    Changes in the concentration of microvilli on the free surface of

    healing mesothelium are associated with alterations in surface

    membrane charge. The Journal of Pathology, 180(3), 333339.

    Mutsaers, S. E., Bishop, J. E., McGrouther, G., & Laurent, G.

    J. (1997). Mechanisms of tissue repair: from wound healing

    to fibrosis. The International Journal of Biochemistry & Cell

    Biology, 29(1), 517.

    Mutsaers, S. E., McAnulty, R. J., Laurent, G. J., Versnel, M.

    A., Whitaker, D., & Papadimitriou, J. M. (1997). Cytokine

    regulation of mesothelial cell proliferation in vitro and in vivo.

    European Journal of Cell Biology, 72(1), 2429.

    Mutsaers, S. E., Whitaker, D., & Papadimitriou, J. M. (2000).

    Mesothelial regeneration is not dependent on subserosal cells.

    The Journal of Pathology, 190(1), 8692.

    Mutsaers, S. E., Whitaker, D., & Papadimitriou, J. M. (2002).

    Stimulation of mesothelial cell proliferation by exudate

    macrophages enhances serosal wound healing in a murine

    model. American Journal of Pathology, 160(2), 681692.

    Nagy, J. A., Shockley, T. R., Masse, E. M., Harvey, V. S., Hoff,

    C. M., & Jackman, R. W. (1995). Systemic delivery of a

    recombinant protein by genetically modified mesothelial cells

    reseeded on the parietal peritoneal surface. Gene Therapy, 2,

    402410.

    Nerem, R. M., & Seliktar, D. (2001). Vascular tissue engineering.

    Annual Review of Biomedical Engineering, 3, 225243.

    Offner, F. A., Feichtinger, H., Stadlmann, S., Obrist, P., Marth, C.,

    & Klingler, P. et al., (1996). Transforming growth factor-beta

    synthesis by human peritoneal mesothelial cells. Induction byinterleukin-1. American Journal of Pathology, 148(5), 1679

    1688.

    Owens, M. W., & Milligan, S. A. (1994). Growth factor

    modulation of rat pleural mesothelial cell mitogenesis and

    collagen synthesis. Effects of epidermal growt