ArticlePDF AvailableLiterature Review

SPARC, a matricellular protein that functions in cellular differentiation and tissue response to injury

Authors:

Abstract and Figures

Expressed during many stages of development in a variety of organisms, the matricellular protein SPARC (secreted protein acidic and rich in cysteine, also known as osteonectin or BM-40) is restricted in adult vertebrates primarily to tissues that undergo consistent turnover or to sites of injury and disease (1). The capacity of SPARC to bind to several resident proteins of the ECM, to modulate growth factor efficacy, to affect the expression of matrix metalloproteinases, and to alter cell shape as a counteradhesive factor, supports the idea that SPARC acts to regulate cell interaction with the extracellular milieu during development and in response to injury (Figure (Figure1;1; see also ref. 1). SPARC is a member of a gene family whose members share structural similarities in one or more protein domains (1). In addition to the numerous studies in cultured cells, the function of SPARC in vivo has been examined primarily in three evolutionarily diverse organisms — Caenorhabditis elegans, Xenopus laevis, and mice. These systems have been used to study the effects of increased or inappropriate SPARC expression, as well as diminished activity resulting from the inactivation of SPARC mRNA, the blocking of protein activity, or mutation of the SPARC gene (Table (Table1).1). This Perspective will integrate results from studies in vitro with findings in vivo in an attempt to clarify the current information and to propose functions for SPARC in living tissues.
Content may be subject to copyright.
The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9 1049
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
Expressed during many stages of development in a vari-
ety of organisms, the matricellular protein SPARC
(secreted protein acidic and rich in cysteine, also known
as osteonectin or BM-40) is restricted in adult vertebrates
primarily to tissues that undergo consistent turnover or
to sites of injury and disease (1). The capacity of SPARC
to bind to several resident proteins of the ECM, to mod-
ulate growth factor efficacy, to affect the expression of
matrix metalloproteinases, and to alter cell shape as a
counteradhesive factor, supports the idea that SPARC
acts to regulate cell interaction with the extracellular
milieu during development and in response to injury
(Figure 1; see also ref. 1). SPARC is a member of a gene
family whose members share structural similarities in one
or more protein domains (1). In addition to the numer-
ous studies in cultured cells, the function of SPARC in
vivo has been examined primarily in three evolutionarily
diverse organisms — Caenorhabditis elegans, Xenopus laevis,
and mice. These systems have been used to study the
effects of increased or inappropriate SPARC expression,
as well as diminished activity resulting from the inactiva-
tion of SPARC mRNA, the blocking of protein activity, or
mutation of the SPARC gene (Table 1). This Perspective
will integrate results from studies in vitro with findings
in vivo in an attempt to clarify the current information
and to propose functions for SPARC in living tissues.
SPARC in ECM organization
Vertebrate SPARC binds to a number of different ECM
components including thrombospondin 1, vitronectin,
entactin/nidogen, fibrillar collagens (types I, II, III, and
V), and collagen type IV, the prevalent collagen in base-
ment membranes (1). Therefore, SPARC has the poten-
tial to contribute to the organization of matrix in con-
nective tissue as well as basement membranes.
Interestingly, SPARC is expressed abundantly in base-
ment membranes and in capsules that surround a vari-
ety of organs and tissues. In this regard, SPARC-null
mice display early cataractogenesis, a phenotype with
100% penetrance (2). Transmission electron microscopy
of lens epithelial cells in SPARC-null mice shows an
intrusion of cellular processes into the basement mem-
brane of the lens capsule, whereas wild-type lens epithe-
lial cells exhibit a precise border at the cell-matrix inter-
face (3). We have proposed that this phenotype reflects
aberrant cell behavior or differentiation resulting from
altered composition or structure of the basement mem-
brane formed in the absence of SPARC.
SPARC, a matricellular protein that functions
in cellular differentiation and tissue response to injury
Amy D. Bradshaw and E. Helene Sage
Department of Vascular Biology, The Hope Heart Institute, Seattle, Washington, USA
Address correspondence to: E. Helene Sage, The Hope Heart Institute, 1124 Columbia Street, Suite 720, Seattle, Washington 98104, USA.
Phone: (206) 903-2026; Fax: (206) 903-2044; E-mail: hsage@hopeheart.org.
Figure 1
Structure of SPARC protein. A ribbon diagram
derived from crystallographic data shows the
three modular domains of SPARC. Representa-
tive activities attributed to each domain are
shown beneath the designated amino acids. The
follistatin-like domain contains the peptide 2.1
(see ref. 1) shown in green and the (K)GHK
angiogenic peptide (amino acids 114–130)
shown in black. The E-C domain contains the
amino acids 255–274 (peptide 4.2) shown in yel-
low. Modified from Hohenester et al. (29) and
the Brookhaven Protein Database, accession no.
1BM0 from Ref. 1, with permission.
With respect to connective tissue, preliminary trans-
mission electron microscopy of dermal collagen fibers
also revealed differences between wild-type and SPARC-
null mice. Whereas collagen fibrils from wild-type skin
exhibit a variety of large and small diameters, as
observed previously in normal adult animals, SPARC-
null fibrils are smaller and more uniform in diameter
(A.D. Bradshaw et al., unpublished results). Differences
in collagen fibril size are consistent with our primary
observation that the skin of SPARC-null mice is more
easily stretched and weaker in tensile strength than
that of wild-type mice. Apparently the absence of
SPARC affects collagen fibrillogenesis, most likely dur-
ing development, although confirmation of this idea
awaits completion of experiments in which a develop-
mental time course of collagen fibril assembly will be
analyzed. Whether SPARC acts to affect fibrillogenesis
directly through its collagen-binding capacity or by
another mechanism is unknown. However, in connec-
tive tissues of mov-13 mice, which are deficient in col-
lagen I, SPARC is not distributed in specific matrices
that are known sites of SPARC deposition in wild-type
embryos (1). In developing Xenopus, SPARC appears to
be concentrated within the intersomitic furrows, a loca-
tion known to be rich in collagen fibers (4).
Further evidence for the importance of SPARC in con-
nective tissue is found in the curly tails of SPARC-null
mice, a characteristic reminiscent of thrombospondin
2–null mice (5). Thrombospondin 2 is another matri-
cellular protein with potential collagen-binding activi-
ty, as thrombospondin 2–null mice also display aberrant
collagen fibrils in the skin and an abnormally flexible
tail (5). In fact, a variety of ECM-associated components
have been implicated in collagen fibrillogenesis by virtue
of the phenotypic abnormalities observed in transgenic
mice with targeted deletions of the genes for decorin,
fibromodulin, lumican, and osteopontin, among others
(6–8). Since some of these proteins and proteoglycans
are known to affect collagen fibrillogenesis in vitro, phe-
notypic abnormalities in col-
lagen fibrils were not unex-
pected in these animals.
Others, such as SPARC and
thrombospondin 2, proved
to be more surprising. Clear-
ly the assembly and regula-
tion of collagen fibril size is
a complex process about
which a great deal remains
to be learned.
Similar to vertebrate
SPARC, C. elegans SPARC is
encoded by a single gene,
ost-1. Although there are
both structural and func-
tional differences between
vertebrate and nematode
SPARC, such as a reduced affinity for Ca2+, binding to
both collagen types I and IV is conserved (1). SPARC in
C. elegans is expressed primarily by body wall and sex
muscle cells, although SPARC protein is also associat-
ed with the basement membrane of the pharynx, a
tissue in which SPARC mRNA is not detected (9). A
similar disparity between sites of synthesis and depo-
sition has been noted in C. elegans for collagen IV, and
in mouse, for SPARC (1). Thus, nematode SPARC
appears to be transported extracellularly to basement
membranes at some distance from its sites of synthe-
sis, an observation suggesting a function for this pro-
tein in matrix organization or activity.
The capacity of SPARC to bind to a number of differ-
ent ECM proteins provides a basis for the association of
SPARC with both basement membranes and fibrous
connective tissue. During development, when many
ECMs are being laid down, higher levels of SPARC are
observed (1). Subsequently, the expression of SPARC is
restricted to sites of ECM turnover and is virtually unde-
tectable in normal cells within their established ECM.
In fact, Damjanovski et al. observed in Xenopus embryos
that expression of SPARC decreased precipitously upon
morphological differentiation of specific tissues (10).
Interestingly, SPARC is a substrate of transglutami-
nase, an enzyme that establishes covalent cross-links
between proteins (11). Although the function of tissue
transglutaminase in the stabilization of the ECM is not
completely understood, strong evidence for its impor-
tance in matrix assembly and cell interaction with ECM
is emerging (12). Possibly, expression of SPARC in
response to injury or tissue remodeling is necessary to
facilitate production of an ECM permissive for cell
migration, proliferation, and differentiation. SPARC
might substitute for other transglutaminase substrates
that provide structural support in the ECM: for exam-
ple, cross-linking of SPARC instead of fibronectin,
another substrate of transglutaminase, could result in
a more malleable matrix.
1050 The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
Table 1
Phenotypic abnormalities resulting from inactivation of SPARC in vivo
Organism Technique Phenotype Reference
C. elegans cRNA interference Embryonic lethality 9
Lack of gut granules
Nonfunctional gonads
X. laevis Antibody block Bent and shortened embryonic axes 28
Abnormal eye development
Mus musculus Transgenic-null Cataractogenesis 2 (and references therein)
Accelerated dermal wound healing A.D. Bradshaw et al., unpublished
Aberrant dermal collagen fibrils A.D. Bradshaw et al., unpublished
Curly tails
Osteopenia 17
Increased fat deposition A.D. Bradshaw and E.H. Sage,
unpublished
SPARC and growth factors
SPARC binds to and decreases the mitogenic potency
of both PDGF and VEGF, in part by its abrogation of
growth factor–receptor interaction (1, 2). In addition,
SPARC can counteract the proliferative capacity of
bFGF on smooth muscle cells, although physical inter-
action of SPARC with bFGF remains to be determined
(1, 2). Furthermore, addition of SPARC to myoblasts
promotes differentiation through inhibition of bFGF-
induced proliferation (K. Motamed et al., unpublished
results). That SPARC binds ECM components and
modulates the growth factor–induced proliferation
(and, in at least one case, differentiation) of at least
three separate mitogens adds another dimension to the
function of SPARC in the regulation of cell behavior.
In addition to PDGF, VEGF, and bFGF, SPARC can
also modulate the activity of TGF-β, as seen in recent
studies with SPARC-null mesangial cells. In culture,
these cells express decreased levels of TGF-βmRNA
and protein in comparison with wild-type mesangial
cells (1). Addition of recombinant SPARC to either
wild-type or SPARC-null cells increases TGF-βmRNA
and protein and is associated with an increase in colla-
gen I production. Interestingly, TGF-βcan interact
with the ECM through the binding of latent TGF-β
binding protein (LTBP-1), which is cross-linked to the
ECM in a transglutaminase-dependent manner and
facilitates activation of latent TGF-βin endothelial
cells (13). The lack of SPARC might lead to aberrant
assembly of ECM that in turn affects the localization
and subsequent activation of TGF-β.
SPARC as a counteradhesive protein
The capacity for SPARC to modulate growth factor
activity has far-reaching implications for many aspects
of cell behavior that include proliferation, migration,
and differentiation. Perhaps equally important, how-
ever, is the counteradhesive activity associated with
SPARC. For over a decade, SPARC has been appreciat-
ed as a modifier of cell shape (1). Addition of purified
SPARC to many cell types in culture induces cell round-
ing, a process thought to be distinct from the inhibi-
tion of cell cycle by SPARC. Cell rounding activity is
sensitive to tyrosine kinase inhibitors, whereas inhibi-
tion of proliferation depends in part upon a pertussis
toxin–sensitive pathway, at least in vascular smooth
muscle cells (1). Focal adhesions dissociate upon addi-
tion of purified SPARC to endothelial cell cultures,
although the mechanism by which SPARC influences
cell shape is not completely understood (14). Whether
SPARC acts upon cell surfaces through a specific recep-
tor or acts by blocking adhesive interactions is unclear.
Indeed these possibilities might each apply in different
situations and would thereby allow cells of various
types to respond to SPARC in a characteristic manner
(see Murphy-Ullrich, this Perspective series, ref. 15).
Cell shape, like growth factor activity, can influence
cell proliferation, migration, and differentiation. The
evidence that SPARC acts to modulate such process-
es in vivo is becoming substantially more credible,
based on experiments performed in C. elegans, Xenopus,
and transgenic mice that do not express SPARC. The
counteradhesive function of SPARC appears to be
conserved in the diverse organisms that express the
protein. Thus, overexpression of SPARC in C. elegans
leads to an uncoordinated phenotype that probably
reflects the compromised ability of the body wall
muscle cells to attach to the adjacent basement mem-
brane and to generate force (1). Huynh et al. (16) have
tested the effects on vertebrate development of a pep-
tide representing the COOH-terminal Ca2+-binding
domain of SPARC (Figure 1, E-C domain). This pep-
tide mimics the full-length protein in its ability to
provoke changes in cell shape (1). When injected into
developing Xenopus embryos, it caused the rounding
of migrating mesodermal cells and severe develop-
mental abnormalities — incomplete gastrulation and
a reduction in anterior structures (16). The severe
cataracts seen at early ages in SPARC-null mice might
also arise because of abnormalities in cell shape. As
shown in Figure 2a, differentiating lens epithelial cells
(shown in cross sections from the cortex of the lens
from 3-month-old animals) normally exhibit an
ordered structure of uniformly shaped cells. Age-
matched SPARC-null lens cells (Figure 2b) display
irregular shapes and alignment. Whether the changes
in cell shape that occur during the normal develop-
ment of the lens are due to the direct action of SPARC
or to a secondary effect occurring later in the differ-
entiation process is currently unknown.
The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9 1051
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
Figure 2
SPARC-null lens epithelial cells display changes in
cell shape and alignment in comparison with
wild-type cells. Sections from similar areas of
wild-type and SPARC-null lens were stained with
hematoxylin and eosin. Cortical fibers from a 3-
month-old wild-type lens (a) are regular and pre-
cisely aligned, whereas those from a SPARC-null
lens (b) are irregular in shape and alignment.
Magnification, ×220.
A recent study of proteins that influence morphine-
induced locomotor sensitivity in the amygdala identi-
fied SPARC as important in this process. It is believed
that prolonged exposure to morphine leads to physio-
logical changes in the brain that might be dependent
upon synaptic rearrangement. Ikemoto et al. found
expression of SPARC in the amygdala, the region of the
brain known to control locomotor sensitivity, in rats
exposed to morphine (17). In fact, administration of
recombinant SPARC directly to the amygdala resulted
in locomotor sensitivity in the absence of morphine.
The authors also showed that SPARC induces shape
changes in neurons, as previously reported for several
other cell types (1). Perhaps the counteradhesive activi-
ty of SPARC facilitates neuronal rearrangement in vivo,
including synaptic plasticity, with its attendant physio-
logical and behavioral implications.
SPARC and differentiation
The body wall muscle dysfunction seen in nematodes
with aberrant expression of SPARC has been pursued by
cRNA interference to block the production of SPARC in
these organisms. The resulting phenotypes ranged from
embryonic lethality to surviving adults with significant
morphological abnormalities, including smaller adults,
a lack of gut granules, and no functional gonads (Table
1; ref. 9). Because the major site of SPARC mRNA tran-
scription in C. elegans occurs in the body wall muscle
cells, embryonic death has been attributed to defects in
muscle function, as no obvious differences in morphol-
ogy are evident at this stage. Smaller adults might result
from a nutritional defect caused by the lack of gut gran-
ules. Whether the absence of SPARC affects the ECM
upon which precursor cells migrate and receive appro-
priate signals for differentiation into gut granules, or
whether another developmental mechanism is dis-
turbed, remains to be determined. Clearly, functional
interruption of SPARC activity in both C. elegans and
Xenopus (Table 1) has severe consequences that most cer-
tainly involve differentiation, although the molecular
mechanisms are currently undefined.
Lens epithelial cells differentiate at specific points
along the lens periphery, during which cells lose contact
with the capsular basement membrane, elongate anteri-
orly and posteriorly, lose their organelles, and eventual-
ly form the highly ordered, transparent structure of the
lens. As SPARC is a component of the lens capsule, the
lack of SPARC might result in a significantly altered
ECM with respect to structure and/or growth factor dep-
osition (2). A hitherto unidentified function of SPARC
might also be critical for these highly specialized cells,
such as the regulation of intercellular adhesive proteins,
e.g., certain of the cadherins, and gap junction proteins,
which are critical for the differentiation of lens fibers.
In vertebrates, SPARC is a major noncollagenous com-
ponent of bone. Delaney et al. recently reported a severe
bone phenotype associated with SPARC-null mice that
was manifested as adult osteopenia (18). The absence of
SPARC led to substantial decreases in bone mass, the
severity of which increased with age. Interestingly, we
have observed significant increases in subcutaneous fat
in SPARC-null relative to wild-type mice, a difference
that also appears to increase with age. Other fat deposits
in SPARC-null mice are increased in size as well, in com-
parison with those of wild-type animals, although over-
all body weight is not substantially different. Both
osteoblasts and adipocytes originate from common pro-
genitor cells present in the bone marrow (19). The
decrease in bone mass and the increase in fat deposition
might reflect differentiation events that favor the for-
mation of adipocytes over that of osteoblasts. Indeed,
SPARC might act directly to influence cell fate in this
example; conversely, the absence of SPARC might affect
ECM assembly and/or localization of growth factors
that in turn could influence differentiation. Rodriguez
et al. recently reported an analysis of the differentiative
capacity of mesenchymal stem cells isolated from post-
menopausal women with and without osteoporosis (20).
Cells from osteoporotic patients produced less TGF-β
and collagen I and exhibited an increased incidence of
adipogenic differentiation in culture. Seemingly, the
deficiencies in TGF-βand hence the capacity to synthe-
size a collagen I–rich ECM favored adipose formation
over bone formation. Since a decrease in TGF-βand col-
lagen I is observed in SPARC-null mesangial cells versus
wild-type cells, a similar mechanism might be in place to
influence mesenchymal stem cell differentiation as well.
That SPARC-null mice develop to adulthood and
reproduce with a grossly normal skeleton and fat
deposits argues against a direct influence of SPARC in
decisions regarding cell fate. A more plausible hypoth-
esis is that subtle differences in ECM configuration
and/or growth factor activity in animals lacking
SPARC lead eventually to tissue failure, especially when
the animal is challenged with injury or disease.
SPARC and wound healing
Dermal wound healing involves many processes that
could be affected by the absence of SPARC: ECM
turnover and reassembly, cell migration, cell prolifera-
tion, and recruitment/differentiation of connective tis-
sue cells. Perhaps surprisingly, we have found that exci-
sional dermal wounds produced in SPARC-null mice
close more rapidly, relative to those in wild-type mice
(A.D. Bradshaw et al., unpublished results). The mech-
anisms by which SPARC could influence dermal clo-
sure and/or healing are many. One possibility is the
capacity of SPARC to influence the activity of TGF-β.
Recently, Ashcroft et al. reported that mice deficient in
Smad-3 (a downstream signaling molecule in the TGF-
βpathway) also display accelerated dermal wound heal-
ing, in comparison with wild-type mice (21). Although
TGF-βhas been shown to augment healing when
administered topically, the signaling pathways in vivo
1052 The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
are complex. Local and probably cell-specific inhibition
of TGF-βsignaling apparently enhances dermal heal-
ing. Thus, the absence of SPARC might decrease the
levels of active TGF-β, and consequently its availability
to certain cells, during dermal healing and would there-
by accelerate the process. In addition, the capacity of
SPARC to inhibit PDGF, bFGF, and VEGF, three fac-
tors that have been shown to improve healing, might
also contribute to the enhancement of wound closure
in the absence of SPARC.
The structure of the dermis in SPARC-null mice, in
particular the collagen fibrils, is clearly different from
that in wild-type mice. The decrease in size of the col-
lagen fibrils, perhaps with other alterations in the
ECM, might result in a more permissive milieu for cel-
lular infiltration and healing. It is likely that the
SPARC-null ECM would be more easily degraded by
matrix metalloproteinases. In addition, overall wound
contraction might be more easily achieved with a
SPARC-null matrix, given that it appears to be less
structured and hence possibly easier to contract than
that of wild-type mice.
Another contribution to enhanced wound closure
might be the increase in subdermal fat that we have
observed in SPARC-null mice. Many of the cells that
invade the wound bed to repair the injured tissue have
been shown to originate in the subdermal fat and mus-
cle layers (22). Perhaps an increase in the amount of fat
enhances the availability of cells that contribute to
wound closure and thus accelerates the healing
process. Moreover, Frank et al. reported recently that
leptin, a factor produced by adipose tissue, enhances
dermal wound healing in mice (23). An increase in the
subcutaneous fat layer in SPARC-null mice most like-
ly results in increased levels of leptin in the skin. Thus
accelerated closure might reflect an increase in leptin
levels at the wound site.
Cancer and angiogenesis
Similar to wound healing, substantial ECM turnover
and rearrangement also occur during tissue invasion by
tumor cells. High levels of SPARC are often associated
with metastatic tumors (1). In fact, SPARC has been
proposed as a diagnostic marker of invasive menin-
giomas (24). Inhibition of SPARC expression by anti-
sense RNA diminished both adhesive and invasive
capacities of human melanoma cells in vitro and in
vivo. Moreover, melanoma cells with suppressed
expression of SPARC were no longer able to generate
tumors when injected into athymic mice, whereas con-
trol melanoma cells showed 100% tumorigenicity (25).
Angiogenesis, the growth of new vessels from extant
vasculature, is a major factor in tumor growth and
metastasis (26). Since neovascularization includes
endothelial cell invasion and ECM remodeling, it was
not surprising to find that SPARC is expressed by
endothelial cells in culture and in tissues (1). Notably,
SPARC has been shown to be associated with growing
vessels in the chick chorioallantoic membrane, a high-
ly vascular structure that provides nutrients and gas
exchange for the growing embryo (1). Interestingly, the
authors reported the generation of proteolytic frag-
ments from SPARC that were localized to the tips of
growing vessels. The Cu2+-binding peptide GHK, previ-
ously characterized as an angiogenic compound in
plasma, as well as the more potent stimulator KGHK,
could be derived from SPARC (Figure 1; see also ref. 1).
To date, SPARC is the only extracellular protein found
to contain the sequence KGHK, which is conserved
throughout vertebrate SPARC (1). The existence of
regions in SPARC that can be released by proteolysis
and that display activities different from those of the
native, intact protein somewhat complicates our
understanding of the functions of SPARC, but at the
same time offers new possibilities for modulation of
cell-matrix interactions (27).
We have recently performed a sponge invasion assay
in SPARC-null mice to determine whether angiogene-
sis in the dermis is abnormal. Animals lacking SPARC
displayed a substantial increase in the amount of
fibrovascular invasion of subcutaneous polyvinyl alco-
hol sponges in comparison with that of wild-type mice
(A.D. Bradshaw et al., unpublished results). Whether an
increase in invasion and angiogenesis reflects a more
permissive milieu for cell migration and proliferation,
due to an altered ECM, or an increase in the availabili-
ty of angiogenic growth factors such as PDGF and
VEGF, remains to be determined. Also of interest is the
use of SPARC-null mice in selected tumor models to
determine whether metastatic potential or tumor
angiogenesis is affected by the absence of SPARC.
Concluding remarks and future directions
That SPARC is expressed by organisms as diverse as C.
elegans and humans is testimony to the fundamental
importance of this protein to multicellular life. Since
invertebrate SPARC does not bind hydroxyapatite, the
SPARC gene has apparently become specialized for an
additional function unique to vertebrates, i.e., bone
formation. The existence of other SPARC family
members, such as SC1, provides another layer of com-
plexity with regard to compensatory activities (1, 2).
SPARC might influence cell behavior through its
interactions with cell surfaces, growth factors, and
ECM. Elucidating the mechanisms by which SPARC
influences cell migration, proliferation, and differen-
tiation will contribute to our understanding of a vari-
ety of different biological processes including devel-
opment, wound healing, angiogenesis, and cancer.
Although significant progress has been made in the
characterization of SPARC, a substantial amount of
information remains to be unearthed, with results
which undoubtedly will prove to be as provocative as
they are gratifying.
The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9 1053
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
Acknowledgments
We would like to thank the members of the Sage labo-
ratory for helpful discussions and ideas. Qi Yan con-
tributed the photograph in Figure 2. This work was sup-
ported by NIH grants GM-40711 and HL-59475 to E.H.
Sage, and DK-07467 to A.D. Bradshaw.
1. Brekken, R.A., and Sage, E.H. 2000. SPARC, a matricellular protein: at
the crossroads of cell-matrix communication. Matrix Biol. 19:569–580.
2. Yan, Q., and Sage, E.H. 1999. SPARC, a matricellular glycoprotein with
important biological functions. J. Histochem. Cytochem. 47:1495–1505.
3. Norose, K., Lo, W.-K., Clark, J.I., Sage, E.H., and Howe, C.C. 2000. Lens-
es of SPARC-null mice exhibit an abnormal cell surface-basement mem-
brane interface. Exp. Eye Res. 71:295–307.
4. Ringuette, M., Drysdale, T., and Liu, F. 1992. Expression and distribu-
tion of SPARC in early Xenopus laevis embryos. Roux’s Archives of Develop-
mental Biology. 202:4–9.
5. Kyriakides, T.R., et al. 1998. Mice that lack thrombospondin 2 display
connective tissue abnormalities that are associated with disordered col-
lagen fibrillogenesis, an increased vascular density, and a bleeding
diathesis. J. Cell Biol. 140:419–430.
6. Danielson, K.G., et al. 1997. Targeted disruption of decorin leads to
abnormal collagen fibril morphology and skin fragility. J. Cell Biol.
136:729–743.
7. Ezura, Y., Chakravarti, S., Oldberg, A., Cheroneva, I., and Birk, D.E. 2000.
Differential expression of lumican and fibromodulin regulate collagen
fibrillogenesis in developing mouse tendons. J. Cell Biol. 151:779–788.
8. Liaw, L., et al. 1998. Altered wound healing in mice lacking a functional
osteopontin gene (spp1). J. Clin. Invest. 101:1468–1478.
9. Fitzgerald, M.C., and Schwarzbauer, J.E. 1998. Importance of the base-
ment membrane protein SPARC for viability and fertility in Caenorhab-
ditis elegans. Curr. Biol. 8:1285–1288.
10. Damjanovski, S., Malaval, L., and Ringuette, M.J. 1994. Transient expres-
sion of SPARC in the dorsal axis of early Xenopus embryos: correlation
with calcium-dependent adhesion and electrical coupling. Int. J. Dev. Biol.
38:439–446.
11. Aeschlimann, D., Kaupp, O., and Paulsson M. 1995. Transglutaminase-
catalyzed matrix cross-linking in differentiating cartilage: identification
of osteonectin as a major glutaminyl substrate. J. Cell Biol. 129:881–892.
12. Greenberg, C.S., Birchbichler, P.J., and Rice, R.H. 1991. Transglutami-
nases: multifunctional cross-linking enzymes that stabilize tissues.
FASEB J. 5:3071–3077.
13. Nunes, I., Gliezes, P.-M., Metz, C.N., and Rifkin, D.B. 1997. Latent trans-
forming growth factor-βbinding protein domains involved in activation
and transglutaminase-dependent cross-linking of latent transforming
growth factor-β. J. Cell Biol. 136:1151–1163.
14. Murphy-Ullrich, J.E., Lane, T.F., Pallero, M.A., and Sage, E.H. 1995.
SPARC mediates focal adhesion disassembly in endothelial cells through
a follistatin-like region and the calcium-binding EF-hand. J. Cell. Biochem.
57:341–350.
15. Murphy-Ullrich, J.E. 2001. The de-adhesive activity of matricellular pro-
teins: is intermediate cell adhesion an adaptive state? J. Clin. Invest.
107:785–790.
16. Huynh, M.-H., Sage, E.H., and Ringuette, M. 1999. A calcium-binding
motif in SPARC/osteonectin inhibits chordomesoderm cell migration
during Xenopus laevis gastrulation: evidence of counter-adhesive activity
in vivo. Dev. Growth Differ. 41:407–418.
17. Ikemoto, M., Takita, M., Imamura, T., and Inoue, K. 2000. Increased sen-
sitivity to the stimulant effects of morphine conferred by anti-adhesive
glycoprotein SPARC in amygdala. Nat. Med. 6:910–915.
18. Delaney, A.M., et al. 2000. Osteopenia and decreased bone formation in
osteonectin-deficient mice. J. Clin. Invest. 105:915–923.
19. Haynesworth, S.E., Goshima, J., Goldberg, V.M., and Caplan, A.I. 1992.
Characterization of cells with osteogenic potential from human marrow.
Bone. 13:81–88.
20. Rodriguez, J.P., Montecinos, L., Rios, S., Reyes, P., and Martinez, J. 2000.
Mesenchymal stem cells from osteoporotic patients produce a type I col-
lagen-deficient extracellular matrix favoring adipogenic differentiation.
J. Cell. Biochem. 79:557–565.
21. Ashcroft, G.S., et al. 1999. Mice lacking smad-3 show accelerated wound
healing and an impaired local inflammatory response. Nat. Cell Biol.
1:260–266.
22. Kurman, M., and Argyris, T.S. 1975. The proliferative response of epi-
dermis of hairless mice to full thickness wounds. Am. J. Pathol.
79:301–310.
23. Frank, S., Stallmeyer, B., Kampfer, H., Kolb, N., and Pfeilschifter, J. 2000.
Leptin enhances wound re-epithlelialization and constitutes a direct
function of leptin in skin repair. J. Clin. Invest. 106:501–509.
24. Rempel, S.A., Ge, S., and Gutierrez, J.A. 1999. SPARC: a potential diag-
nostic marker of invasive meningiomas. Clin. Cancer Res. 5:237–241.
25. Ledda, M.F., et al. 1997. Suppression of SPARC expression by antisense
RNA abrogates the tumorigenicity of human melanoma cells. Nat. Med.
3:171–176.
26. Folkman, J. 1995. Angiogenesis in cancer, vascular, rheumatoid and
other disease. Nat. Med. 1:27–31.
27. Sage, E.H. 1997. Pieces of eight: bioactive fragments of extracellular pro-
teins as regulators of angiogenesis. Trends Cell Biol. 7:182–186.
28. Purcell, L., Gruia-Gray, J., Scanga, S., and Ringuette, M. 1993. Develop-
mental anomalies of Xenopus embryos following microinjection of
SPARC antibodies. J. Exp. Zool. 265:153–164.
29. Hohenester, E., Maurer, P., and Timpl, R. 1997. Crystal structure of a pair
of follistatin-like and EF-hand calcium-binding domains in BM-40.
EMBO J. 16:3778–3786.
30. Protein Data Bank. http://www.rcsb.org/pdb/.
1054 The Journal of Clinical Investigation | May 2001 | Volume 107 | Number 9
PERSPECTIVE SERIES
E. Helene Sage, Series Editor
Matricellular proteins
... During tissue development and differentiation, SPARC expression is initially intense but it declines in most organs after maturation. Eventually, SPARC is restrictly expressed in post-developmental tissues with high ECM turnover such as bone and gut mucosa [3]. However, SPARC expression was upregulated during angiogenesis [3], inflammation [4], wound-healing [4] and tumor development [5]. ...
... Eventually, SPARC is restrictly expressed in post-developmental tissues with high ECM turnover such as bone and gut mucosa [3]. However, SPARC expression was upregulated during angiogenesis [3], inflammation [4], wound-healing [4] and tumor development [5]. ...
Article
Full-text available
Background Secreted protein acidic and rich in cysteine (SPARC) has been shown to modulate aggressive behavior in several benign and malignant tumors. Little is known about SPARC expression in odontogenic keratocyst (OKC), an odontogenic cyst with an aggressive nature. To the best of our knowledge, only one study has been investigated the expression of this protein in OKCs. This study aimed to characterize SPARC expression in OKCs. Additionally, to determine whether SPARC is associated with aggressive behavior in OKCs, SPARC expression in OKCs was compared with radicular cysts (RCs), dentigerous cysts (DCs) and calcifying odontogenic cysts (COCs). These odontogenic cysts showed no or less aggressive behavior. Methods SPARC expression was evaluated in 38 OKCs, 39 RCs, 35 DCs and 14 COCs using immunohistochemistry. The percentages of positive cells and the intensities of immunostaining in the epithelial lining and the cystic wall were evaluated and scored. Results Generally, OKCs showed similar staining patterns to RCs, DCs and COCs. In the epithelial lining, SPARC was not detected, except for ghost cells in all COCs. In the cystic wall, the majority of positive cells were fibroblasts. Compared between 4 groups of odontogenic cysts, SPARC expression in OKCs was significantly higher than those of RCs (P < 0.001), DCs (P < 0.001) and COCs (P = 0.001). Conclusions A significant increase of SPARC expression in OKCs compared with RCs, DCs and COCs suggests that SPARC may play a role in the aggressive behavior of OKCs.
... Rac family small GTPase 1 (RAC1), targeted by miR-146a-5p [163,164] and miR-574-3p [165], activates p38-MAPK signalling [166], increases ECM production in TM cells and elevates SPARC and proinflammatory IL-6 in TM cells [167,168]. SPARC binds to ECM proteins and regulates the expression of matrix metalloproteinases (MMPs) [169]. SPARC expression is up-regulated by TGFβ2 in TM cells and is inhibited by the miR-29 family in the TM [106,170]. ...
Preprint
Full-text available
Primary open-angle glaucoma (POAG) is a progressive optic neuropathy with a complex, multifactorial aetiology. Raised intraocular pressure (IOP) is the most important clinically modifiable risk factor for POAG. All current pharmacological agents target aqueous hu-mour dynamics to lower IOP. Newer therapeutic agents are required as some patients with POAG show a limited therapeutic response or develop ocular and systemic side ef-fects to topical medication. Elevated IOP in POAG results from cellular and molecular changes in the trabecular meshwork driven by increased levels of transforming growth factor β (TGFβ) in the anterior segment of the eye. Understanding how TGF affects both the structural and functional changes in the outflow pathway and IOP is required to de-velop new glaucoma therapies that target the molecular pathology in the trabecular meshwork. In this study we evaluated the effects of TGF-1 and -2 treatment on miRNA expression in cultured human primary trabecular meshwork cells. Our findings are pre-sented in terms of specific miRNAs (miRNA-centric), but given miRNAs work in net-works to control cellular pathways and processes, a pathway-centric view of miRNA ac-tion is also reported. Evaluating TGF-responsive miRNA expression in trabecular meshwork cells will further our understanding of the important pathways and changes involved in the pathogenesis of glaucoma and could lead to the development of miRNAs as new therapeutic modalities in glaucoma.
... On the other hand, the accelerated degeneration of NP during aging in Smo knockout mice may be related to prolonged excessive mechanical stress caused by severer AF defects. Indeed, a causal relationship between AF defects and NP degeneration has been previously observed in several loss-of-function studies [4,9,39]. In addition to AF defects, defects in other tissues, such as CEP, vertebral bone mass or vertebral growth plate, may also contribute to the NP phenotypes in Smo Agcl mice. ...
Article
Full-text available
Intervertebral disc degeneration is closely related to abnormal phenotypic changes in disc cells. However, the mechanism by which disc cell phenotypes are maintained remains poorly understood. Here, Hedgehog-responsive cells were found to be specifically localized in the inner annulus fibrosus and cartilaginous endplate of postnatal discs, likely activated by Indian Hedgehog. Global inhibition of Hedgehog signaling using a pharmacological inhibitor or Agc1-CreERT2-mediated deletion of Smo in disc cells of juvenile mice led to spontaneous degenerative changes in annulus fibrosus and cartilaginous endplate accompanied by aberrant disc cell differentiation in adult mice. In contrast, Krt19-CreER-mediated deletion of Smo specifically in nucleus pulposus cells led to healthy discs and normal disc cell phenotypes. Similarly, age-related degeneration of nucleus pulposus was accelerated by genetic inactivation of Hedgehog signaling in all disc cells, but not in nucleus pulposus cells. Furthermore, inactivation of Gli2 in disc cells resulted in partial loss of the vertebral growth plate but otherwise healthy discs, whereas deletion of Gli3 in disc cells largely corrected disc defects caused by Smo ablation in mice. Taken together, our findings not only revealed for the first time a direct role of Hedgehog-Gli3 signaling in maintaining homeostasis and cell phenotypes of annuls fibrosus and cartilaginous endplate, but also identified disc-intrinsic Hedgehog signaling as a novel non-cell-autonomous mechanism to regulate nucleus pulposus cell phenotype and protect mice from age-dependent nucleus pulposus degeneration. Thus, targeting Hedgehog signaling may represent a potential therapeutic strategy for the prevention and treatment of intervertebral disc degeneration.
... SPARC is a novel myokine 18 secreted from the myocytes of muscle tissue. The SPARC molecule is known to have regenerative effects such as the regeneration of muscle tissue after physical or pathological damage through repair, cellular differentiation, and remodeling mechanisms 16 . In teleosts, SPARC is expressed in skeletal muscle during regeneration and development, as well as in satellite cells, suggesting that SPARC has a vital role in the skeletal muscle compartment 19 . ...
Article
Full-text available
SPARC is an extracellular Ca2+-binding, secreted glycoprotein that plays a dynamic role in the growth and development of organisms. This study aimed to describe the isolation, characterization, and expression analysis of HdhSPARC in Pacific abalone (Haliotis discus hannai) to infer its potential functional role. The isolated HdhSPARC was 1633 bp long, encoding a polypeptide of 284 amino acid residues. Structurally, the SPARC protein in abalone is comprised of three biological domains. However, the structure of this protein varied between vertebrates and invertebrates, as suggested by their distinct clustering patterns in phylogenetic analysis. In early development, HdhSPARC was variably expressed, and higher expression was found in veliger larvae. Moreover, HdhSPARC was highly expressed in juvenile abalone with rapid growth compared to their slower-growing counterparts. Among the testicular development stages, the growth stage exhibited higher HdhSPARC expression. HdhSPARC was also upregulated during muscle remodeling and shell biomineralization, as well as in response to different stressors such as heat shock, LPS, and H2O2 exposure. However, this gene was downregulated in Cd-exposed abalone. The present study first comprehensively characterized the HdhSPARC gene, and its spatio-temporal expressions were analyzed along with its responses to various stressors.
... and fibrosis. [10][11][12] In the lung, SPARC has been studied to be linked to cancer pathogenesis, pulmonary fibrosis, pulmonary vascular remodeling and chronic airway diseases. 11,[13][14][15] Veith et al. 14 revealed that SPARC was one of consistently downregulated genes by whole genome DNA microarray analysis in a reverse-remodeling model of pulmonary hypertension, and SPARC knockdown in adult mice by adeno-associated virus dramatically improved hemodynamic and pulmonary vascular remodeling. ...
Article
Full-text available
Purpose Airway remodeling is a critical feature of asthma. Secreted protein acidic and rich in cysteine (SPARC), which plays a cardinal role in regulating cell-matrix interactions, has been implicated in various fibrotic diseases. However, the effect of SPARC in asthma remains unknown. Methods We studied the expression of SPARC in human bronchial epithelial cells and serum of asthmatics as well as in the lung tissues of chronic asthma mice. The role of SPARC was examined by using a Lentivirus-mediated SPARC knockdown method in the ovalbumin (OVA)-induced asthma mice. The biological processes regulated by SPARC were identified using RNA sequencing. The function of SPARC in the remodeling process induced by transforming growth factor β1 (TGFβ1) was conducted by using SPARC small interfering RNA (siRNA) or recombinant human SPARC protein in 16HBE cells. Results We observed that SPARC was up-regulated in human bronchial epithelia of asthmatics and the asthmatic mice. The levels of serum SPARC in asthmatics were also elevated and negatively correlated with the forced expiratory volume in one second (FEV1) to forced vital capacity ratio (FVC) (r = −0.485, P < 0.01) and FEV1 (%predicted) (r = −0.425, P = 0.001). In the chronic asthmatic mice, Lentivirus-mediated SPARC knockdown significantly decreased airway remodeling and airway hyper-responsiveness. According to gene set enrichment analysis, negatively enriched pathways found in the OVA + short hairpin-SPARC group included ECM organization and collagen formation. In the lung function studies, knockdown of SPARC by siRNA reduced the expression of remodeling-associated biomarkers, cell migration, and contraction by blocking the TGFβ1/Smad2 pathway. Addition of human recombinant SPARC protein promoted the TGFβ1-induced remodeling process, cell migration, and contraction in 16HBE cells via the TGFβ1/Smad2 pathway. Conclusions Our studies provided evidence for the involvement of SPARC in the airway remodeling of asthma via the TGFβ1/Smad2 pathway.
Article
Satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute and chronic muscle injuries. The balance between stem cell self‐renewal and differentiation determines the kinetics and efficiency of skeletal muscle regeneration. This study assessed the function of Islr in SC asymmetric division. The deletion of Islr reduced muscle regeneration in adult mice by decreasing the SC pool. Islr is pivotal for SC proliferation, and its deletion promoted the asymmetric division of SCs. A mechanistic search revealed that Islr bound to and degraded secreted protein acidic and rich in cysteine (SPARC), which activated p‐ERK1/2 signaling required for asymmetric division. These findings demonstrate that Islr is a key regulator of SC division through the SPARC/p‐ERK1/2 signaling pathway. These data provide a basis for treating myopathy.
Article
Full-text available
Low pH in the gut is associated with severe inflammation, fibrosis, and colorectal cancer (CRC) and is a hallmark of active inflammatory bowel disease (IBD). Subsequently, pH-sensing mechanisms are of interest for the understanding of IBD pathophysiology. Tissue hypoxia and acidosis—two contributing factors to disease pathophysiology—are linked to IBD, and understanding their interplay is highly relevant for the development of new therapeutic options. One member of the proton-sensing G protein-coupled receptor (GPCR) family, GPR65 (T-cell death-associated gene 8, TDAG8), was identified as a susceptibility gene for IBD in a large genome-wide association study. In response to acidic extracellular pH, GPR65 induces an anti-inflammatory response, whereas the two other proton-sensing receptors, GPR4 and GPR68 (ovarian cancer G protein-coupled receptor 1, OGR1), mediate pro-inflammatory responses. Here, we review the current knowledge on the role of these proton-sensing receptors in IBD and IBD-associated fibrosis and cancer, as well as colitis-associated cancer (CAC). We also describe emerging small molecule modulators of these receptors as therapeutic opportunities for the treatment of IBD.
Preprint
Full-text available
Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the differentiation of cardiac fibroblasts (cFbs) into myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α−SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro . Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere alignment, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
Article
Full-text available
The expression of tissue transglutaminase in skeletal tissues is strictly regulated and correlates with chondrocyte differentiation and cartilage calcification in endochondral bone formation and in maturation of tracheal cartilage (Aeschlimann, D., A. Wetterwald, H. Fleisch, and M. Paulsson. 1993. J. Cell Biol. 120:1461-1470). We now demonstrate the transglutaminase reaction product, the gamma-glutamyl-epsilon-lysine cross-link, in the matrix of hypertrophic cartilage using a novel cross-link specific antibody. Incorporation of the synthetic transglutaminase substrate monodansylcadaverine (amine donor) in cultured tracheal explants reveals enzyme activity in the pericellular matrix of hypertrophic chondrocytes in the central, calcifying areas of the horseshoe-shaped cartilages. One predominant glutaminyl substrate (amine acceptor) in the chondrocyte matrix is osteonectin as revealed by incorporation of the dansyl label in culture. Indeed, nonreducible osteonectin-containing complexes of approximately 65, 90, and 175 kD can be extracted from mature tracheal cartilage. In vitro cross-linking of osteonectin by tissue transglutaminase gives similar products of approximately 90 and 175 kD, indicating that the complexes in cartilage represent osteonectin oligomers. The demonstration of extracellular transglutaminase activity in differentiating cartilage, i.e., cross-linking of osteonectin in situ, shows that tissue transglutaminase-catalyzed cross-linking is a physiological mechanism for cartilage matrix stabilization.
Article
Full-text available
Thrombospondin (TSP) 2, and its close rela- tive TSP1, are extracellular proteins whose functions are complex, poorly understood, and controversial. In an attempt to determine the function of TSP2, we dis- rupted the Thbs2 gene by homologous recombination in embryonic stem cells, and generated TSP2-null mice by blastocyst injection and appropriate breeding of mu- tant animals. Thbs2 2 / 2 mice were produced with the expected Mendelian frequency, appeared overtly nor- mal, and were fertile. However, on closer examination, these mice displayed a wide variety of abnormalities. Collagen fiber patterns in skin were disordered, and ab- normally large fibrils with irregular contours were ob- served by electron microscopy in both skin and tendon. As a functional correlate of these findings, the skin was fragile and had reduced tensile strength, and the tail was unusually flexible. Mutant skin fibroblasts were de- fective in attachment to a substratum. An increase in total density and in cortical thickness of long bones was documented by histology and quantitative computer to- mography. Mutant mice also manifested an abnormal bleeding time, and histologic surveys of mouse tissues, stained with an antibody to von Willebrand factor, showed a significant increase in blood vessels. The basis for the unusual phenotype of the TSP2-null mouse could derive from the structural role that TSP2 might play in collagen fibrillogenesis in skin and tendon. However, it seems likely that some of the diverse mani- festations of this genetic disorder result from the ability of TSP2 to modulate the cell surface properties of mes- enchymal cells, and thus, to affect cell functions such as adhesion and migration.
Article
Full-text available
The generation of animals lacking SMAD proteins, which transduce signals from transforming growth factor- (TGF-), has made it possible to explore the contribution of the SMAD proteins to TGF- activity in vivo. Here we report that, in contrast to predictions made on the basis of the ability of exogenous TGF- to improve wound healing, Smad3-null (Smad3ex8/ex8) mice paradoxically show accelerated cutaneous wound healing compared with wild-type mice, characterized by an increased rate of re-epithelialization and significantly reduced local infiltration of monocytes. Smad3ex8/ex8 keratinocytes show altered patterns of growth and migration, and Smad3ex8/ex8 monocytes exhibit a selectively blunted chemotactic response to TGF-. These data are, to our knowledge, the first to implicate Smad3 in specific pathways of tissue repair and in the modulation of keratinocyte and monocyte function in vivo.
Article
Full-text available
Osteopontin (OPN) is an arginine-glycine-aspartate (RGD)- containing glycoprotein encoded by the gene secreted phosphoprotein 1 (spp1). spp1 is expressed during embryogenesis, wound healing, and tumorigenesis; however, its in vivo functions are not well understood. Therefore, OPN null mutant mice were generated by targeted mutagenesis in embryonic stem cells. In OPN mutant mice, embryogenesis occurred normally, and mice were fertile. Since OPN shares receptors with vitronectin (VN), we tested for compensation by creating mice lacking both OPN and VN. The double mutants were also viable, suggesting that other RGD-containing ligands replace the embryonic loss of both proteins. We tested the healing of OPN mutants after skin incisions, where spp1 was upregulated as early as 6 h after wounding. Although the tensile properties of the wounds were unchanged, ultrastructural analysis showed a significantly decreased level of debridement, greater disorganization of matrix, and an alteration of collagen fibrillogenesis leading to small diameter collagen fibrils in the OPN mutant mice. These data indicate a role for OPN in tissue remodeling in vivo, and suggest physiological functions during matrix reorganization after injury.
Article
Full-text available
The healing of full thickness surgical wounds from 0 to 14 days has been studied in hairless mice. Within 2 days after wounding the surrounding epidermis is thickened and primarily composed of enlarged basophilic cells. The remnants of the hair follicles attached to the epidermis are converted into cords of enlarged basophilic cells. Epidermal thickening is maximum at 7 days. Associated with the epidermal thickening surrounding the wound edge is an increase in the total number of cells in the stratum granulosum and in the stratum spinosum. The number of basal cells does not change. Mitotic activity and mitotic rate increase within 1 day after wounding. Both reach their peaks by approximately 5 to 7 days and then begin to return to control levels. Mitotic duration does not change.
Article
Specific stages of angiogenesis are regulated by extracellular proteins. This review discusses the endogenous proteolysis of eight of these proteins and the release of polypeptide fragments that have biological activities different from those of the native, parent protein. The generation of natural cleavage products could provide a precise mechanism for the regulation of angiogenesis. Although further experimental confirmation of this mechanism is needed, this leitmotif offers an attractive explanation, in part, for the complex role of proteolysis in vascular morphogenesis.
Article
Secreted protein, acidic, rich in cysteine (SPARC) is a Ca2+-binding, counter-adhesive, extracellular glycoprotein associated with major morphogenic events and tissue remodeling in vertebrates. In Xenopus laevis embryos, SPARC is expressed first by dorsal mesoderm cells at the end of gastrulation and undergoes complex, rapid changes in its pattern of expression during early organogenesis. Another study has reported that precocious expression of SPARC by injection of native protein into the blastocoele cavity of pregastrula embryos leads to a concentration-dependent reduction in anterior development. Thus, normal development requires that the timing, spatial distribution, and/or levels of SPARC be regulated precisely. In a previous study, we demonstrated that injection of a synthetic peptide corresponding to the C-terminal, Ca2+-binding, EF-hand domain of SPARC (peptide 4.2) mimicked the effects of native SPARC. In the present investigation, peptide 4.2 was used to examine the cellular and molecular bases of the phenotypes generated by the aberrant presence of SPARC. Exposure of late blastula embryos to LiCl also generated a concentration-dependent reduction in anterior development; therefore, injections of LiCl were carried out in parallel to highlight the unique effects of peptide 4.2 on early development. At concentrations that caused a similar loss in anterior development (60–100 ng peptide 4.2 or 0.25–0.4 μg LiCl), LiCl had a greater inhibitory effect on the initial rate of chordomesoderm cell involution, in comparison with peptide 4.2. However, as gastrulation progressed, peptide 4.2 had a greater inhibitory effect on prospective head mesoderm migration than that seen in the presence of LiCl. Moreover, peptide 4.2 and LiCl had distinct influences on the expression pattern of dorso-anterior markers at the neural and tail-bud stages of development. Scanning electron microscopy showed that peptide 4.2 inhibited spreading of migrating cells at the leading edge of the involuting chordomesoderm. While still in close proximity to the blastocoele roof, many of the cells appeared rounded and lacked lamellipodia and filopodia extended in the direction of migration. In contrast, LiCl had no effect on the spreading or shape of involuting cells. These data are the first evidence of a counter-adhesive activity for peptide 4.2 in vivo, an activity demonstrated for both native SPARC and peptide 4.2 in vitro.
Article
The publisher regrets that the above article was published with several typographical errors. The corrected version appears on the following pages. SPARC is a multifunctional glycoprotein that belongs to the matricellular group of proteins. It modulates cellular interaction with the extracellular matrix (ECM) by its binding to structural matrix proteins, such as collagen and vitronectin, and by its abrogation of focal adhesions, features contributing to a counteradhesive effect on cells. SPARC inhibits cellular proliferation by an arrest of cells in the G1 phase of the cell cycle. It also regulates the activity of growth factors, such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF)-2, and vascular endothelial growth factor (VEGF). The expression of SPARC in adult animals is limited largely to remodeling tissue, such as bone, gut mucosa, and healing wounds, and it is prominent in tumors and in disorders associated with fibrosis. The crystal structure of two of the three domains of the protein has revealed a novel follistatin-like module and an extracellular calcium-binding (EC) module containing two EF-hand motifs. The follistatin-like module and the EC module are shared by at least four other proteins that comprise a family of SPARC-related genes. Targeted disruption of the SPARC locus in mice has shown that SPARC is important for lens transparency, as SPARC-null mice develop cataracts shortly after birth. SPARC is a prototypical matricellular protein that functions to regulate cell–matrix interactions and thereby influences many important physiological and pathological processes.
Article
Studies using animal tissue suggest that bone marrow contains cells with the potential to differentiate into cartilage and bone. We report the extension of these studies to include human marrow. Bone marrow from male and female donors of various ages was obtained either from the femoral head or as aspirates from the iliac crest, and introduced into culture. Culture-adherent cells were expanded, subcultured, and then tested for bone and cartilage differentiation potential utilizing two different in vivo assays in nude mice. One assay involved subcutaneous implantation of porous calcium phosphate ceramics loaded with cultured, marrow-derived, mesenchymal cells; the other involved peritoneal implantation of diffusion chambers, also inoculated with cultured, marrow-derived, mesenchymal cells. Histological evaluation showed bone formation in ceramics implanted with cultured, marrow-derived, mesenchymal cells originating from both the femoral head and the iliac crest. Immunocytochemical analysis indicates that the bone is derived from the implanted human cells and not from the cells of the rodent host. No cartilage was observed in any of these ceramic grafts. In contrast, aliquots from the same preparations of cultured, marrow-derived, mesenchymal cells failed to form bone or cartilage in diffusion chambers. These data suggest that human marrow contains cells with osteogenic potential, which can be enriched and expanded in culture. Our findings also suggest that subcutaneous implantation of these cells in porous calcium phosphate ceramics may be a more sensitive in vivo assay than diffusion chambers for measuring their osteogenic lineage potential.