Anglani Et Al_2010

download Anglani Et Al_2010

of 8

Transcript of Anglani Et Al_2010

  • 7/26/2019 Anglani Et Al_2010

    1/8

    The Regenerative Potential of the Kidney: What Can We

    Learn from Developmental Biology?

    Franca Anglani &Federica Mezzabotta &Monica Ceol &

    Rosalba Cristofaro &Dorella Del Prete &

    Angela DAngelo

    Published online: 17 August 2010# Springer Science+Business Media, LLC 2010

    Abstract Cell turnover in the healthy adult kidney is very

    slow but the kidney has a strong capacity for regenerationafter acute injury. Although many molecular aspects of this

    process have been clarified, the source of the newly-formed

    renal epithelial cells is still being debated. Several studies

    have shown, moreover, that the repair of injured renal

    epithelium starts from mature tubular cells, which enter into

    an activated proliferative state characterized by the reappear-

    ance of mesenchymal markers detectable during nephro-

    genesis, thus pointing to a marked plasticity of renal epithelial

    cells. The regenerative potential of mature epithelial cells

    might stem from their almost unique morphogenetic process.

    Unlike other tubular organs, all epithelial and mesenchymal

    cells in the kidney derive from the same germ layer, the

    mesoderm. In a fascinating view of vertebrate embryogenesis,

    the mesoderm might be seen as a cell layer capable of

    oscillating between epithelial and mesenchymal states, thus

    acquiring a remarkable plasticity that lends it an extended

    potential for innovation and a better control of three-

    dimensional body organization. The renal papilla contains a

    population of cells with the characteristic of adult stem cells.

    Mesenchymal stromal stem cells (MSC) have been found to

    reside in the connective tissue of most organs, including thekidney. Recent studies indicate that the MSC compartment

    extends throughout the body postnatally as a result of its

    perivascular location. Developmental biology suggests that

    this might be particularly true of the kidney and that the

    papilla might represent the perivascular renal stem cell niche.

    The perivascular niche hypothesis fits well with the evolving

    concept of the stem cell niche as an entity of action. It is its

    dynamic capability that makes the niche concept so important

    and essential to the feasibility of regenerative medicine.

    Keywords Renal adult stem cells . Kidney development.

    EMT . Cell plasticity . Pericytes . Perivascular niche

    Introduction

    The adult mammalian renal tubular epithelium exists in a

    relatively quiescent to slowly replicating state, and cell turnover

    in the healthy adult kidney is very slow. The kidney has a strong

    capacity for regeneration, despite the postnatal mammalian

    kidney being unable to generate new nephrons in response to

    nephron loss. No new nephrons are generated after 36 weeks of

    gestation in humans, but a renal stem cell system is supposed to

    contribute to the replacement of postnatal cell types [1].

    The anatomical and functional recovery of renal integrity

    after injury is accompanied by the activation of sophisti-

    cated processes that have yet to be fully understood, by

    means of which damaged tubular cells are replaced by

    normal, well-functioning cells that reorganize their archi-

    tecture to recreate a normal tubule. Although many of the

    molecular details of this process have been clarified over

    the last 20 years, the cellular source of the newly-formed

    renal epithelial cells is still being debated. Some studies

    F. Anglani :F. Mezzabotta :M. Ceol : R. Cristofaro

    Laboratory of Kidney Histomorphology and Molecular Biology,

    Department of Medical and Surgical Sciences, University of Padua,

    Padua, Italy

    D. Del Prete :A. DAngelo

    Division of Nephrology, Department of Medical and Surgical

    Sciences, University of Padua,

    Padua, Italy

    F. Anglani (*)

    Divisione di Nefrologia, Policlinico IV piano,

    Laboratorio di Istomorfologia e Biologia Molecolare del Rene,

    Via Giustiniani n2,

    35128 Padova, Italy

    e-mail: [email protected]

    Stem Cell Rev and Rep (2010) 6:650657

    DOI 10.1007/s12015-010-9186-6

  • 7/26/2019 Anglani Et Al_2010

    2/8

    showed that novel cells derive from the division of

    differentiated cells [2, 3]; others claimed that subpopula-

    tions of renal tubular cells act as progenitor cells [ 47]; and

    yet others demonstrated that hematopoietic stem cells and

    circulating mesenchymal stem cells can repopulate the

    tubular cell system [8, 9]. Indeed, a wide variety of

    potential progenitor or adult stem cell populations have

    been identified in human and rodent kidneys on the basis oftheir functional and/or phenotypic characteristics in the

    glomerular and in the tubular/interstitial compartments

    (reviewed in [10]) (Fig.1). Many organs, particularly those

    with a high cell turnover, are believed to harbor a stem cell

    population that sustains normal organ structure and con-

    tributes to repair, and some of these organs contain several

    types of stem/precursor cells that provide new cells for

    specific locations and functions. Recent studies have shown

    in organs such as the skin [21] and liver [22] that the

    precursor cells responsible for homeostatic cell turnover

    differ from those responsible for cell replacement after

    injury. Given the complexity of the renal architecture andthe numerous cell types involved, the kidney probably has

    multiple mechanisms of cell renewal and regeneration too.

    Cell renewal and regeneration

    Most data on the origin of new tubular epithelial cells in the

    kidney derive from studies on the regeneration/repair

    process after tubular necrosis due to ischemic injury. Since

    the S3 segment of the proximal tubule is the part of the

    nephron most susceptible to injury and appears to have the

    highest proliferation rate (in young rats at least), most

    studies on regenerative nephrogenesis have focused on the

    proximal tubular epithelial cells (TECs). Vogetseder et al.

    [2325] showed that cell proliferation in the healthy kidney

    of young rats relies on differentiated TEC division. Moreimportantly, in a recent study using a refined technique

    called genetic fate mapping, Humphereys et al. [26] clearly

    demonstrated that differentiated TECs surviving after

    ischemia-reperfusion injury generated most, if not all, new

    tubular epithelial cells.

    Taking a morphological approach, Vogetseder et al. [23]

    showed that, in the healthy kidney, the slow cycling cells in

    the S3 segments (label retaining cells, LRCs)identifiable

    from their ability to retain BrdUexpressed markers of

    terminal differentiation and a typical tubular cell polarity.

    This study showed that the cell proliferation pattern in the

    S3 segment fails to display the characteristic features of astem cell system. The authors also found that the

    proliferative capacity of renal TECs comes from a large

    reserve of cells in the G1 phase, which ensures a rapid

    proliferative response when needed. This was a theory first

    advanced by Lin et al. [27] who tagged renal TECs with

    green fluorescent protein and provided compelling evidence

    of mature TECs having a leading role in the regeneration

    process in the post-ischemic kidney. The findings reported

    Fig. 1 Distribution of stem/progenitor cells within the adult kidney. The different cell types involved are indicated. Studies conducted on mouse

    or human kidney [47,1120] evidence a distributed presence of renal stem/progenitor cells

    Stem Cell Rev and Rep (2010) 6:650657 651

  • 7/26/2019 Anglani Et Al_2010

    3/8

    by Humphereys et al. [26] substantially confirm these data,

    elegantly demonstrating that no extratubular cells or intra-

    tubular stem/progenitor cells contribute to the repair

    process. They did not address, however, the possibility that

    there may be distinct differences in the regenerative

    potential among tubular cells.

    So where or what does this regenerative potential of mature

    epithelial cells come from? Developmental biology couldprovide the answer.

    Epithelial-to-mesenchymal transition in developmental

    processes

    Several studies have shown that the repair of injured renal

    epithelium begins when surviving tubular cells enter into an

    activated proliferative state characterized by the reappear-

    ance of mesenchymal markers detectable during nephro-

    genesis [28]. After injury, therefore, the conversion of

    TECs to a more mesenchymal phenotype suggests that anepithelial-to-mesenchymal transition does indeed occur.

    Epithelial-to-mesenchymal transition (EMT) is a complex,

    extreme manifestation of epithelial plasticity. It is now

    widely recognized as a fundamental process in pathological

    conditions (oncogenesis and fibrogenesis) as well as in

    physiological events such as morphogenesis [29, 30].

    During the development of the vertebrate embryo,

    EMTs are a source of mesenchyme in various places and

    stages throughout embryonic morphogenesis, but espe-

    cially in the mesodermal domains [31]. It is currently

    accepted that the epithelia precede both evolutionary and

    ontogenetically the mesenchyme, and that the mesen-

    chyme was originally an epithelial derivative [32]. Thus,

    the epithelium is the earliest type of embryonic organiza-

    tion, and the primitive epiblast represents the earliest

    epithelial state in vertebrate development. The first EMT

    occurs at gastrulation, when a subset of epiblast cells

    undergo EMT and internalize to generate mesoderm and

    endoderm, while those remaining in the epiblast become

    ectoderm [33]. Mesoderm and endoderm contribute to

    many tissues in the adult body that subsequently undergo

    several rounds of EMT and MET (the latter being reverse

    of EMT i.e. mesenchymal-to-epithelial transition). Imme-

    diately after the primary mesenchyme has formed, most

    mesoderm and all endoderm become reorganized in

    primitive epithelia. These secondary epithelia give rise to

    secondary mesenchymal cells in a series of EMTs. In

    particular, intermediate mesoderm generates the nephro-

    genic mesenchyme, which coalesces in tertiary epithelial

    structures such as nephrones and nephric ducts [34].

    In a very interesting model proposed by Perez-Pomares

    et al. [31], the vertebrate embryo is considered as a two-

    state system of which epithelium and mesenchyme repre-

    sent the stable and unstable states, respectively. In partic-

    ular, mesoderm might be seen as a cell layer capable of

    oscillating between the plastic and exploratory behavior of

    the mesenchyme and the stability of the epithelia. The

    ability to go through cycles of epithelial and mesenchymal

    states lends the mesoderm a greater plasticity, more

    potential for innovation and a better control of three-

    dimensional body organization [31]. The mesenchymederived from the chronologically earliest EMT has the

    greatest developmental potential, which decreases progres-

    sively in the subsequent EMTs. This model has a particular

    appeal for explaining the kidneys almost exclusively

    morphogenic process as well as its regenerative potential.

    In this setting, some of the developmental potential of the

    primitive epithelium may remain in the definitive epitheli-

    um derived from the mesenchyme (as in renal epithelial

    cells), enabling cells to respond quickly to micro-

    environmental changes. Indeed, very recently, two papers

    have proposed that EMT could be the process driving

    mammary epithelial cells [35] and glomerular parietalepithelial cells [36] to adopt stem cell characteristics.

    These remarks could suggest that the kidneys need for a

    real stem cell compartment is less important than the cells

    phenotypic flexibility, as in other mesenchymal tissues. The

    plasticity of TECs resembles that of adult mesenchymal cells,

    which are long-lived and constantly exposed to the extracel-

    lular matrix. The matrix provides a microenvironment that

    helps the cells to maintain a differentiated or undifferentiated

    state, i.e. the cell plasticity of this tissue seems to serve the

    same purpose as the multipotency of adult stem cells

    without the need for a real stem cell compartment [37].

    The renal papilla as a stem cell niche

    Oliver et al. [38] found a high proliferation rate in the upper

    papilla, especially laterally, adjacent to the urinary space of the

    normal kidney. By staining renal tissue sections from adult 1-

    yr old rats with Ki67, they found positive cells in the upper

    papilla with a frequency that was low (2.6% of all cells), but

    significantly higher than elsewhere in the kidney. The same

    Authors also found LRCs with several characteristics of adult

    stem cells in the papillary interstitium and collecting duct, that

    were able to generate new cells in the normal adult kidney.

    They suggested that, during normal homeostasis, interstitial

    LRCs or their immediate progeny migrate to the upper papilla

    and form a compartment of rapidly proliferating cells that may

    have a role after acute injury.

    Dekel et al. [6] also found non-tubular Sca-1+ multi-

    potent/progenitor cells in the adult mouse kidney, located

    mainly in the papillary interstitial space. These cells were

    located in the outermost part of the papilla, close to the

    tubules and sometimes adjacent to the tubular basal surface,

    652 Stem Cell Rev and Rep (2010) 6:650657

  • 7/26/2019 Anglani Et Al_2010

    4/8

    and they displayed some features of the papillary stem cells

    isolated by Oliver at al. [38].

    Unlike other areas of the kidney, the renal papilla displayed

    no apoptosis after the ischemic insult, so the presence of

    numerous LRCs within a restricted area of the papilla base

    and the rapidly-proliferating cells in the upper papilla (which

    can migrate to other parts of the kidney more susceptible to

    the low oxygen tension) evoke the architectural arrangementof the stem cell compartments of highly-regenerative organs

    such as the epidermis. Stem cells are usually located in deep,

    narrow tissue niches, where the proliferative phenomena

    behind self-maintenance take place. The transient zone (rich

    in amplifying precursor cells) and the differentiated cells in

    various stages of maturation lie respectively in the progres-

    sively more distal cell layers.

    The stem cell niche as a functional entity

    The concept of stem cell niche is changing. The mere

    location of stem cells is not sufficient to define a niche,which must have both anatomical and functional dimen-

    sions [39]. This issue is particularly important as new stem

    cell sites come to light. Accumulating data have challenged

    the conviction that heterologous cells must necessarily

    occur in niches, suggesting that for some stem cells the

    niche may consist of extracellular matrix and other non-

    cellular constituents that could regulate their function [39].

    Both paracrine and metabolic factors could serve function-

    ally as a niche, which could also be seen as a dynamic

    structure capable of being formed anew and responding to

    exogenous signals via a sensing system possibly consisting

    of the nervous or circulatory systems. Indeed, there is a

    growing body of data to indicate the perivascular site as a

    stem cell niche [4042]. Whether the components of the

    vasculature are niche cells (i.e. cells interacting with stem

    cells) or stem cells remains to be seen.

    The perivascular mesenchymal stem cell niche hypothesis

    Multipotent mesenchymal stromal cells (MSCs), typically

    defined as adherent fibroblastoid cells capable of differen-

    tiating into osteoblasts, adipocytes and chondrocytes in

    vitro, have been found to reside within the connective tissue

    of most organs, including the kidney, where they have been

    isolated from glomeruli and the kidney as a whole [40].

    Immunophenotyping indicates that MSC populations orig-

    inating from different sources share numerous surface

    markers, e.g. CD29, CD44, and express SMA, a vascular

    smooth muscle cell marker, pointing to a relationship with

    perivascular cells [43].

    These findings have prompted the suggestion that MSCs

    may derive from perivascular cells. Pericytes are good

    candidates to represent the MSC population within organs [41].

    In resting tissues, pericytes and endothelial cells (EC) are

    quiescent, slow-cycling cells, but in the angiogenic stage

    they show a highly-proliferative potential, a capacity for

    self renewal, and the ability to generate daughter cells, thus

    behaving like adult stem cells. Pericytes isolated from

    different tissues display a marked plasticity and are able to

    differentiate into osteoblasts, chondroblasts, fibroblasts,

    pre-adipocytes [42]. It is also worth mentioning thatpericytes have been involved in vascular and ectopic

    calcification [44]. That adult organ MSCs might be

    pericytes or pericyte-like cells is also supported by the fact

    that they can share some markers, such as CD146, 3G5

    [45]. Taking these findings as a whole, we could conclude

    that the MSC compartment extends through the post-natal

    body as a result of its perivascular location.

    The microvasculature of the renal papilla is particularly

    rich in pericytes, subendothelial cells that regulate micro-

    vascular integrity in the peritubular capillary network and

    give the descending vasa recta (the arteriolar segments

    supplying blood to the medulla) their contractile function.Thus in the papilla might reside that MSC compartment

    linked to the perivascular niche. Mesenchymal stromal stem

    cells have repeatedly been isolated from the kidney [14,

    4649]. Very recently Lee et al [20] have presented

    evidences that progenitor cells (MKPC) with mesenchymal

    stromal stem cell properties reside in the mouse kidney

    interstitium of the medulla and papilla in close association

    with endothelial cells. Indeed, all stem/progenitor cells

    isolated from the papilla expressed markers of MSCs such

    as CD 29 and showed positivity forSMA [5,6,20] which

    is considered a typical marker of perycites pointing to a

    possible relationship with papillary vascular pericytes [50]

    (Fig. 2).

    Could pericytes represent a subpopulation of these

    MSCs in the kidney, and in the papilla in particular? Is

    there an ancestor of the MSC natively associated with the

    perivascular cells? Once again, the answer comes from

    developmental biology.

    Embryonic origin of stromal and vascular kidney cells

    The mammalian kidney develops in three waves, only the last

    of which (the metanephros) continues to exist as the adult

    kidney. All three embryonic kidneys arise from the meso-

    derm, one of the earliest tissues to develop in the embryo.

    Nephrogenesis involves a carefully-controlled series of

    morphogenic and differentiation events that start with the

    interaction between two different primordial epithelial and

    mesenchymal tissues both originating from the intermediate

    mesoderm, i.e. the ureteric bud of Wolffian derivation grows

    and invades the metanephric blastema. Factors secreted by the

    ureteric bud induce this mesenchyme first to form clusters and

    Stem Cell Rev and Rep (2010) 6:650657 653

  • 7/26/2019 Anglani Et Al_2010

    5/8

    then to change into epithelium and the specialized structures

    constituting the nephron. The induced mesenchyme in turn

    sends signals back to the ureteric bud to make it divide and

    grow (reviewed in [51]). This process of reciprocal induction

    proceeds in an orderly manner, from the deep to the outer

    cortex, producing branches of the collecting tree and neph-

    rons, progressing from the glomerulus to the distal tubule.

    The development of most tubular organs relies on signaling

    between epithelial and mesenchymal stromal progenitor

    populations. These two populations often derive from different

    germ layers that are specified during gastrulation, well in

    advance of organ condensation [52]. In kidney development,

    however, the mesenchymal and epithelial lineages both derive

    from the intermediate mesoderm. Moreover, the branching

    morphogenesis of most tubular organs arises from pre-existing

    epithelial sheets or tubules, whereas in the kidney not only do

    the mesenchymal cells of metanephric blastema generate all

    the epithelia of the nephron through mesenchymal-to-

    epithelial transition (MET), they also drive the branching of

    the ureteric bud. The metanephric blastema (also known as

    metanephric mesenchyme) is composed of a non-

    homogeneous population of cells that sends signals to the

    Wolffian duct, which gives rise to the ureteric bud in response

    [53]. One of the fundamental questions of kidney develop-

    mental biology is how many cell lineages there are in the

    metanephric blastema before and after it is induced by the

    ureteric bud. The traditionally held view is that mesenchymal

    cells that do not regain an epithelial state through MET may

    differentiate and give rise to connective tissue and stromal

    cells [54].

    To date, the origin of renal stromal cells has remained

    unknown, though there is some evidence of these cells

    developing from a separate cell lineage within the meta-

    nephric blastema [55]. Using fate mapping techniques,

    Guillaume et al. [52] very recently demonstrated that large

    populations of renal stromal cell originate from a different

    part of the mesodermthe paraxial mesodermthus

    indicating that renal development (like that of all other

    tubular organs) depends on the integration of progenitors

    from different embryonic tissues into a single rudiment. It is

    noteworthy that bones, skeletal muscle and tendons

    originate from paraxial mesoderm. In this light, the ability

    of MSCs to differentiate into osteoblasts and chondrocytes

    must be a commitment received in the very early

    embryological stages.

    During the reciprocal inductive stage of metanephric

    development, two distinct regions of metanephric mesen-

    chyme are morphologically distinguishable, i.e. the cap

    mesenchymal cells, which are Pax2 positive and will form

    nephrons; and the region peripheral to the cap mesenchyme,

    where cells expressing Foxd1 may represent stromal progen-

    itor cells, and cells that are Foxd1-negative, but Six2-, Gdnf-

    and Cited1-positive, may be mesenchymal stem or progenitor

    cells. When branching of the ureteric bud has begun and

    nephron induction takes place, the stromal cells surrounding

    the branches of the ureteric bud and the induced nephrons

    Fig. 2 a The different stem cell

    types isolated from the papillary

    interstitium are indicated, fig-

    ures were drawn from the

    articles cited within parethesis. b

    Histology of the renal papilla,

    EE staining, 200x magnifica-

    tion. c -smooth muscle stain-

    ing of the renal papilla, 200X

    magnification

    654 Stem Cell Rev and Rep (2010) 6:650657

  • 7/26/2019 Anglani Et Al_2010

    6/8

    constitute the primitive interstitium [56] (Fig. 3). By late

    gestation, a secondary interstitium is created with two distinct

    populations, the cortical stroma and the medullary stroma,

    which then differentiate to form the adult kidney interstitium,

    with the cortical interstitium comprising fibroblasts and

    lymphocyte-like cells, and the medullary interstitium mainly

    comprising lipid-laden interstitial cells, lymphocyte-like cells

    and pericytes [56,57].

    From these studies, one might argue thatwhatever

    their true origin (from the intermediate or paraxial meso-

    derm)the mesenchymal stromal cells contain different

    cell lineages, including the precursor of the vasculature.

    Angioblasts (individual endothelial progenitors) are

    integral to the development of the glomerular tuft, and they

    are identified in the developing kidney because they

    express typical vascular markers, such as vascular endo-

    thelial growth factor receptor 2 (VEGFR2). Although

    metanephric mesenchyme contains VEGFR2-positive cells

    from the early stages of development, cross-species

    transplantation studies have suggested that extrarenal cells

    also contribute to the glomerular vasculature [58].

    Mesoangioblasts are considered vessel-associated meso-

    dermal stem cells. Originally identified in the mouse

    embryonic aorta, similar but not identical cells have been

    identified in postnatal vessels of several organs, such as the

    skeletal muscle and heart. Interestingly, they have in common

    the expression of endothelial and/or pericyte markers [59].

    Mesangioblasts may be the ancestors of postnatal vessel-

    associated progenitors, such as pericytes [60].

    Taken together, these findings may indicate: 1) that MSCs

    andpericytes are identical in that they have a common ancestor

    and share the same phenotypic markers; 2) that the distribution

    of MSCs throughout the body post-natally is related to their

    existence in a perivascular niche; and 3) that interstitial MSCs

    might reside in the microvasculature in the kidney papilla.

    Conclusion

    Our growing understanding of the cellular and molecular

    mechanisms behind kidney regeneration and repair pro-

    cessestogether with a knowledge of the embryonic origin

    of renal cellsshould induce us to bear in mind that the

    kidneys need for a real stem cell compartment is less

    important than the cells phenotypic flexibility. One of the

    major tasks of regenerative medicine will be to disclose the

    molecular mechanisms underlying renal tubular plasticity

    and to exploit its biological and therapeutic potential.

    On the other hand, data are emerging that capillary and

    microvessel walls all over the body may harbor a reserve of

    MSCs; in the kidney, this might be located in the papilla.

    The perivascular niche hypothesis fits well with the

    evolving concept of the stem cell niche as an entity of

    action. It is its dynamic capability that makes the niche

    concept so important and essential to the feasibility of

    regenerative medicine.

    Acknowledgements This study was supported by Grant No.

    CPDA085494 from the University of Padua.

    Conflicts of interest The authors declare no potential conflicts of

    interest.

    Fig. 3 Schematic representation of renal development during em-

    bryogenesis showing the different cellular components involved in theprocess of nephrogenesis and collecting duct branching. Figure is

    based on Figure 4 in the Journal of Cellular and Molecular Medicine

    by authors Anglani F, Forino M, Del Prete D, Tosetto E, TorregrossaR., DAngelo A [51]

    Stem Cell Rev and Rep (2010) 6:650657 655

  • 7/26/2019 Anglani Et Al_2010

    7/8

    References

    1. Little, M. H. (2006). Regrow or repair: potential regenerative

    therapies for the kidney. Journal of the American Society of

    Nephrology, 17, 2390401.

    2. Witzgall, R., Brown, D., Schwarz, C., & Bonventre, J. V. (1994).

    Localization of proliferating cell nuclear antigen, vimentin, c-Fos,

    and clusterin in the postischemic kidney. Evidence for a

    heterogenous genetic response among nephron segments, and alarge pool of mitotically active and dedifferentiated cells. Journal

    of Clinical Investigation, 93, 21752188.

    3. Bonventre, J. V. (2003). Dedifferentiation and proliferation of

    surviving epithelial cells in acute renal failure. Journal of the

    American Society of Nephrology, suppl 1, S55S61.

    4. Maeshima, A., Yamashita, S., & Nojima, Y. (2003). Identification

    of renal progenitor-like tubular cells that participate in the

    regeneration processes of the kidney. Journal of the American

    Society of Nephrology, 14, 31383146.

    5. Oliver, J. A., Maarouf, O., Cheema, F. H., Martens, T. P., & Al-

    Awqati, Q. (2004). The renal papilla is a niche for adult kidney

    stem cells. Journal of Clinical Investigation, 114, 795804.

    6. Dekel, B., Zangi, L., Shezen, E., et al. (2006). Isolation and

    characterization of non tubular sca-1 + lin- multipotent stem/

    progenitor cells from adult mouse kidney. Journal of theAmerican Society of Nephrology, 17, 33003314.

    7. Gupta, S., Verfaillie, C., Chmielewski, D., et al. (2006). Isolation

    and characterization of kidney-derived stem cells. Journal of the

    American Society of Nephrology, 17, 30283040.

    8. Poulsom, R., Alison, M. R., Cook, T., et al. (2003). Bone marrow

    stem cells contribute to healing of the kidney. Journal of the

    American Society of Nephrology, suppl 1, S48S54.

    9. McTaggart, S. J., & Atkinson, K. (2007). Mesenchymal stem

    cells: immunobiology and therapeutic potential in kidney disease.

    Nephrology, 12, 4452.

    10. Little, M. H., & Bertram, J. F. (2009). Is there such a thing as a

    renal stem cell? Journal of the American Society of Nephrology,

    20, 21122117.

    11. Hishikawa, K., Marumo, T., Miura, S., Nakanishi, A., Matsuzaki,

    Y., & Shibata, K. (2005). Musculin/MyoR is expressed in kidneyside population cells and can regulate their function. The Journal

    of Cell Biology, 169, 921928.

    12. Challen, G. A., Bertoncello, I., Deane, J. A., Ricardo, S. D., &

    Little, M. H. (2006). Kidney side population reveals multilineage

    potential and renal functional capacity but also cellular heterogene-

    ity. Journal of the American Society of Nephrology, 17, 18961912.

    13. Lazzeri, E., Crescioli, C., Ronconi, E., et al. (2007). Regenerative

    potential of embryonic renal multipotent progenitors in acute renal

    failure.Journal of the American Society of Nephrology, 18, 3128

    3138.

    14. Sagrinati, C., Netti, G. S., Mazzinghi, B., et al. (2006). Isolation

    and characterization of multipotent progenitor cells from the

    Bowmans capsule of adult human kidneys. Journal of the

    American Society of Nephrology, 17, 24432456.

    15. Bruno, S., Bussolati, B., Grange, C., et al. (2009). Isolation and

    characterization of resident mesenchymal stem cells in human

    glomeruli.Stem Cells and Development, 18, 867880.

    16. Bussolati, B., Bruno, S., Grange, C., et al. (2005). Isolation of

    renal progenitor cells from adult human kidney. The American

    Journal of Pathology, 166, 545555.

    17. Kitamura, S., Yamasaki, Y., Kinomura, M., et al. (2005).

    Establishment and characterization of renal progenitor like cells

    from S3 segment of nephron in rat adult kidney. The FASEB

    Journal, 2005(19), 17891797.

    18. Yamashita, S., Maeshima, A., & Nojima, Y. (2005). Involvement

    of renal progenitor tubular cells in epithelial-to-mesenchymal

    transition in fibrotic rat kidneys. Journal of the American Society

    of Nephrology, 16, 20442051.

    19. Markovc-Lipkovsky, J., Muller, C. A., Klein, G., et al. (2007).

    Neural cell adhesion molecule expression on renal interstitial

    cells.Nephrology, Dialysis, Transplantation, 22, 15581566.

    20. Lee, P. T., Lin, H. H., Jiang, S. T., et al. (2010). Mouse kidney

    progenitor cells accelerate renal regeneration and prolong survival

    after ischemic injury. Stem Cells, 28(3), 57384.

    21. Ito, M., Liu, Y., Yang, Z., et al. (2005). Stem cells in the hair

    follicle bulge contribute to wound repair but not to homeostasis ofthe epidermis. Natural Medicines, 2005(11), 13511354.

    22. Crosby, H. A., & Strain, A. J. (2001). Adult liver stem cells: bone

    marrow, blood, or liver derived? Gut, 48, 153154.

    23. Vogetseder, A., Karadeniz, A., Kaissling, B., & Le Hir, M. (2005).

    Tubular cell proliferation in the healthy rat kidney. Histochemistry

    and Cell Biology, 124, 97104.

    24. Vogetseder, A., Palan, T., Bacic, D., Kaissling, B., & Le Hir, M.

    (2007). Proximal tubular epithelial cells are generated by division

    of differentiated cells in the healthy kidney. American Journal of

    Physiology. Cell Physiology, 292, C807C813.

    25. Vogetseder, A., Picard, N., Gaspert, A., Walch, M., Kaissling, B.,

    & Le Hir, M. (2008). The proliferation capacity of the renal

    proximal tubule involves the bulk of differentiated epithelial cells.

    American Journal of Physiology. Cell Physiology, 294, C22C28.

    26. Humphreys, B. D., Valerius, M. T., Kobayashi, A., et al. (2008).

    Intrinsic epithelial cells repair the kidney after injury. Cell Stem

    Cell, 2, 284291.

    27. Lin, F., Moran, A., & Igarashi, P. (2005). Intrarenal cells, not bone

    marrow-derived cells, are the major source for regeneration in

    postischemic kidney.Journal of Clinical Investigation, 115, 1756

    1764.

    28. Anglani, F., Ceol, M., Mezzabotta, F., et al. (2008). The renal stem

    cell system kidney repair and regeneration. Frontiers in Biosci-

    ence, 13, 63956405.

    29. Zavadil, J., & Bttinger, E. P. (2005). TGF-beta and epithelial-to-

    mesenchymal transitions. Oncogene, 24, 57645774.

    30. Kalluri, R., & Weinberg, R. A. (2009). The basic of epithelial-

    mesenchymal transition. Journal of Clinical Investigation, 119,

    14201428.

    31. Perez-Pomares, J. M., & Munoz-Chapuli, R. (2002). Epithelial-

    mesenchymal transition: A mesodermal cell strategy for evolutive

    innovation in metazoans. The Anatomical Record, 268, 343351.

    32. Acloque, H., & Adams, M. S. (2009). Epithelial-mesenchymal

    transition: the importance of changing cell state in development

    and disease. Journal of Clinical Investigation, 119, 14381449.

    33. Hay, E. D. (1968). Organization and fine structure of epithelium

    and mesenchyme in the developing chick embryo. In R.

    Fleischmajer & R. E. Billingham (Eds.), Epithelial-mesenchymal

    interactions(pp. 3155). Baltimore: Williams and Wilkins.

    34. Gilbert, S. F. (1995). Developmental biology (p. 894). Sunderland:

    Sinauer Associates.

    35. Mani, S. A., Guo, W., Liao, M. J., et al. (2008). The epithelial-

    mesenchymal transition generates cells with properties of stem

    cells. Cell, 133(4), 704

    15.36. Swetha, G., Chandra, V., Phadnis, S., & Bhonde, R. (2009).

    Glomerular parietal epithelial cells of adult murine kidney

    undergo EMT to generate cells with traits of renal progenitors. J

    Cell Mol Med. Oct 16 [Epub ahead of print]

    37. Bianchi, G., Muraglia, A., Daga, A., Corte, G., Cancedda, R., &

    Quarto, R. (2001). Microenvironment and stem properties of

    bone-marrow derived mesenchymal cells. Wound Repair and

    Regeneration, 9, 460466.

    38. Oliver, J. A., Klinakis, A., Cheema, F. K., et al. (2009).

    Proliferation and migration of label-retaining cells of the kidney

    papilla. Journal of the American Society of Nephrology, 20(11),

    23152327.

    656 Stem Cell Rev and Rep (2010) 6:650657

  • 7/26/2019 Anglani Et Al_2010

    8/8

    39. Scadden, D. T. (2006). The stem-cell niche as an entity of action.

    Nature, 441, 10751079.

    40. da Silva Meirelles, L., Chagastelles, P. C., & Nardi, N. B. (2006).

    Mesenchymal stem cells in virtually all post-natal organs and

    tissues.Journal of Cell Science, 119, 22042213.

    41. Crisan, M., Yap, S., Casteilla, L. A., et al. (2008). A perivascular

    origin for mesenchymal stem cells in multiple human organs. Cell

    Stem Cell, 3, 301313.

    42. Diaz-Flores, L., Gutirrez, R., Madrid, J. F., et al. (2009).

    Pericytes. Morphofunction, interaction and pathology in aquiescent and activated mesenchymal cell niche. Histol Histo-

    patol, 24, 909969.

    43. Phinney, D. G., & Prockop, D. J. (2007). Concise review:

    mesenchymal stem/multipotent stromal cells: the state of trans-

    differentiation and modes of tissue repaircurrent views. Stem

    Cells, 25, 28962902.

    44. Johnson, R. C., Leopold, J. A., & Loscalzo, J. (2006). Vascular

    calcification: pathobiological mechanisms and clinical implica-

    tions.Circulation Research, 99, 10441059.

    45. Covas, D. T., Panepucci, R. A., Fontes, A. M., et al. (2008).

    Multipotent mesenchymal stromal cells obtained from diverse

    human tissues share functional properties and geneexpression

    profile with CD146+ perivascular cells and fibroblasts. Experi-

    mental Hematology, 36, 642654.

    46. Plotkin, M. D., & Goligorsky, M. S. (2006). Mesenchymal cells

    from adult kidney support angiogenesis and differentiate into

    multiple interstitial cell types including erytropoietin-producing

    fibroblast. American Journal of Physiology-Renal Physiology,

    291, F902F912.

    47. Chen, J., Park, H. C., Addabbo, F., et al. (2008). Kidney-derived

    mesenchymal stem cells contribute to vasculogenesis, angiogen-

    esis and endothelial repair. Kidney International, 74, 879889.

    48. Patschan, D., Michurina, T., Shi, H. K., et al. (2007). Normal

    distribution and medullary-to-cortical shift of Nestinexpressing

    cells acute renal ischemia. Kidney International, 71, 744754.

    49. Huang, Y., Johnston, P., Zhang, B., et al. (2009). Kidney-derived

    stromal cells modulate dendritic and T cell responses. Journal of

    the American Society of Nephrology, 20, 831841.

    50. Park, F., Mattson, D. L., Roberts, L. A., & JR, C. A. W. (1997).

    Evidence for the presence of smooth muscle -actin within

    pericytes of the renal medulla. Am J Physiol Regulatory

    Integrative Comp Physiol, 273, 17421748.

    51. Michos, O. (2009). Kidney development: from ureteric bud formation

    to branching morphogenesis.Current Opin Genet Dev, 19, 484490.

    52. Guillaume, R., Bressan, M., & Herzlinger, D. (2009). Paraxialmesoderm contributes stromal cells to the developing kidney.

    Developmental Biology, 329, 169175.

    53. Anglani, F., Forino, M., Del Prete, D., Tosetto, E., Torregrossa, R.,

    & DAngelo, A. (2004). In search of adult renal stem cells.

    Journal of Cellular and Molecular Medicine, 8, 474487.

    54. Ekblon, P., & Weller, A. (1991). Ontogeny of tubular interstitial

    cells. Kidney International, 39, 394400.

    55. Al-Awati, Q., & Oliver, J. A. (2002). Stem cells in the kidney.

    Kidney International, 61, 387397.

    56. Dressler, R. D. (2009). Advances in early kidney specification,

    development and patterning. Development, 136, 38633874.

    57. Cullen-McEwen, L. A., Caruana, G., & Bertran, J. F. (2005). The

    where, what and why of the developing renal stroma. Experimen-

    tal Nephrology, 99, e1e8.

    58. Vaughan, M. R., & Quaggin, S. E. (2008). How do mesangial and

    endothelial cells form glomerular tuft. Journal of the American

    Society of Nephrology, 19, 2433.

    59. Tonlorenzi, R., Dellavalle, A., Schnapp, E., Cossu, G., &

    Sampaolesi, M. (2007). Isolation and characterization of meso-

    angioblasts from mouse, dog and human tissue. Curr Proc Stem

    Biol, chapter 2:unit 2B.1.

    60. Brunelli, S., Tagliafico, E., De Angelis, F. G., et al. (2004). Msx2

    and necdin combined activities are required for smooth muscle

    differentiation in mesoangioblast stem cells.Circulation Research,

    94, 15711578.

    Stem Cell Rev and Rep (2010) 6:650657 657