ArticlePDF AvailableLiterature Review

*Mechanisms of integration of cells and extracellular matrices by integrins

Authors:

Abstract

While it is self-evident that all extracellular molecules are an integral part of a multicellular organism, it is paradoxical that they are often considered to be dissociated from cells. The reality is that a continuum of dynamic, bi-directional interactions links the intracellular environment through cell-surface receptors to multimolecular extracellular assemblies. These interactions not only control the behaviour of individual cells, but also determine tissue architecture. Adhesion receptor function is partly determined by an ability to tether the contractile cytoskeleton to the plasma membrane, but there is also evidence that integrin receptors modulate signalling events that are essential for cellular differentiation. A major challenge is now to integrate work at the atomic, molecular and cellular levels, and obtain holistic insights into the mechanisms controlling cell adhesion. In the present study, we review current knowledge of the molecular mechanisms employed by cells to integrate with the extracellular matrix. Two main topics are covered: the adaptation of integrin structure for bi-directional signalling and the integration of integrin signalling with other receptors.
822 Biochemical Society Transactions (2004) Volume 32, part 5
Mechanisms of integration of cells and
extracellular matrices by integrins
M.J. Humphries1, M.A. Travis, K. Clark and A.P. Mould
Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, 2.205 Stopford Building,
Oxford Road, Manchester M13 9PT, U.K.
Abstract
While it is self-evident that all extracellular molecules are an integral part of a multicellular organism, it
is paradoxical that they are often considered to be dissociated from cells. The reality is that a continuum
of dynamic, bi-directional interactions links the intracellular environment through cell-surface receptors
to multimolecular extracellular assemblies. These interactions not only control the behaviour of individual
cells, but also determine tissue architecture. Adhesion receptor function is partly determined by an ability
to tether the contractile cytoskeleton to the plasma membrane, but there is also evidence that integrin
receptors modulate signalling events that are essential for cellular differentiation. A major challenge is now
to integrate work at the atomic, molecular and cellular levels, and obtain holistic insights into the mech-
anisms controlling cell adhesion. In the present study, we review current knowledge of the molecular
mechanisms employed by cells to integrate with the extracellular matrix. Two main topics are covered: the
adaptation of integrin structure for bi-directional signalling and the integration of integrin signalling with
other receptors.
Integrin structure, priming and activation
In mammals, 18 α-and8β-integrin genes encode poly-
peptides that combine to form 24 α,βheterodimeric receptors
[1]. Both subunits are non-covalently associated, type I
transmembrane proteins with large extracellular and mostly
short cytoplasmic domains. On the basis of their primary
structure, integrins fall into two subfamilies determined by
the presence or absence of a von Willebrand factor A-domain
in the αsubunit. Beginning with the X-ray crystal structure of
the isolated αM integrin A-domain in 1995 [2], recent years
have seen major advances in our understanding of integrin
tertiary structure. A step-change came with the solution of
the first structure of a full-length extracellular dimer (for
αVβ3) in 2001 [3], followed closely by the structure of
a ligand mimetic–receptor complex, formed by soaking an
RGD peptide into pre-existing αVβ3crystals[4].Theoverall
shape of the crystallized conformer of αVβ3 resembles a
large ‘head’ on two ‘legs’ (Figure 1). In contrast with models
derived from electron microscopic and biophysical analyses
[5], both legs are bent in the crystal structure. The head of the
integrin, which is the minimal fragment that contains ligand-
binding activity, comprises a β-propeller from the αsubunit,
an A-domain from the βsubunit (the βA-domain) and an
immunoglobulin ‘hybrid’ domain below the βA-domain.
The α-subunit leg of the integrin comprises three β-sandwich
domains, termed ‘thigh’, ‘calf1’ and ‘calf2’. The β-subunit
leg contains a plexin–semaphorin–integrin domain, which
Key words: activation, adhesion, conformation, cytoskeleton, integrin, signalling.
Abbreviations used: ECM, extracellular matrix; FA, focal adhesion; FB, fibrillar adhesion; FC, focal
complex; FN, fibronectin; LIBS, ligand-induced-binding sites; mAb, monoclonal antibody.
1To whom correspondence should be addressed (email martin.humphries@man.ac.uk).
is disordered in the structure, four EGF-like repeats and a
cystatin-like fold. The bend in the integrin is between thigh
and calf1 (in αV), and at the conjunction of the hybrid domain,
two EGF repeats and the PSI domain (in β3). Since 2001, there
have been no further integrin crystal structures reported, and,
therefore, the conformational diversity of the family remains
unclear.
In the RGD–αVβ3complexstructure,thepeptidelies
at the interface between the αVβ-propeller and the β3A-
domain, and the carboxyl group of the aspartate co-ordinates
a bivalent cation at a so-called MIDAS (metal-ion-dependent
adhesion site) [4]. A similar receptor–cation–ligand interac-
tion had been observed previously in a crystal structure of
acomplexbetweentheα2 A-domain and a triple-helical
collagen peptide [6]. While complementary conformations
of ligand acidic motifs and receptor cation-binding pockets
might determine the specificity of receptor-ligand binding,
there is also evidence that integrin ligands contain accessory
or ‘synergy’ sites for receptor binding. FN (fibronectin) is
the best-characterized ligand in this regard, and although it
has been suggested that receptor engagement of the synergy
site is transient [7], a combination of gain-of-function ana-
lyses with integrin αsubunit chimaeras, anti-functional
mAb (monoclonal antibody) epitope mapping, and structure
determination by small-angle X-ray scattering have led to
a detailed topological model for the FN–integrin complex
[8,9]. Solution of the structure of a macromolecular ligand–
integrin complex remains a priority for understanding
how integrins form stable interactions with ECM (extracel-
lular matrix) proteins.
The dynamic nature of integrin function, by which cells
use adhesion to sample their pericellular environment and
C
2004 Biochemical Society
Signalling Outwards and Inwards 823
Figure 1 Cartoon of the straightened form of an integrin which
lacks an αA-domain
The αsubunit is in red and the βsubunit in blue. The polypeptide
modules comprising both subunits and the major ligand-binding sites
are denoted. The sites of unbending are indicated by green arrows.
The conformational changes that take place during integrin priming and
activation are indicated by magenta arrows. These include movements
of α-helices within the βA-domain, a swing-out of the hybrid domain
away from the αsubunit, closing up of the interfaces between βA
and the hybrid domain, and the βA-domain and the β-propeller, and
leg/cytoplasmic domain separation.
respond by changing their position and differentiated state,
demands a highly responsive receptor structure (Figure 1).
Gross conformational changes in integrins have been detected
by a variety of biophysical and biochemical techniques
(reviewed in [10]), and much work has been performed on
activation-dependent binding of mAbs to integrins. A subset
of these mAbs recognize so-called LIBS (ligand-induced-
binding sites). It has been hypothesized that LIBS mAbs
stimulate integrin-ligand binding by displacing conform-
ational equilibria in favour of ligand-competent and ligand-
occupied forms of the integrin. Shape changes reported by
LIBS mAbs can also be triggered from the cytoplasmic
side of the plasma membrane [11], but the degree of simi-
larity between the changes induced from either side of the
plasma membrane is currently unclear. To separate these two
processes conceptually, we have proposed the term ‘priming’
to indicate the acquisition of ligand-binding ability by in-
tegrins and ‘activation’ to denote ligand-induced changes [12].
The availability of the crystal structure of αVβ3hasstimu-
lated structure–function analyses of integrin priming. The
relevance of the bend in the integrin dimer has received
much attention. Although it has been proposed that integrins
remain bent [13], several lines of evidence indicate that bent
integrins are inactive and extended integrins are primed. For
example, epitopes that become exposed on integrin priming,
and residues that restrain priming, are buried in the bent knee
of β2 integrins [14], and locked-bent integrins containing
engineered disulphide bonds have very low affinity for ligand
[15]. Furthermore, predominantly bent conformers of β3
integrins are observed by electron microscopy in the presence
of Ca2+, whereas extended conformers are found in Mn2+
[15].
It is now well established that leg separation underpins
both priming and activation of integrins. Thus some stimu-
latory mAb epitopes located in the leg regions are exposed
by activating cytoplasmic domain mutations [16], forced
association of the membrane-proximal regions of the α-and
β-subunit cytoplasmic domains with an engineered coiled-
coil constrains the integrin in an inactive state [16], unclasping
these constrained integrins leads to a spatial separation of the
legs [17], and fluorescence resonance energy transfer studies
show that integrin cytoplasmic domains are close to each
other in the resting state, but undergo spatial separation
during both priming and activation [18].
Using LIBS mAbs as activation state reporters, we have
shown that movement of both the α1andα7 helices of the
β1 A-domain contributes to priming [19,20]. Shape changes
in these elements had previously been observed in αA-domain
crystal structures. βA-domain conformational changes are
coupled with a swing-out of the underlying hybrid domain,
as activating mutations in the α7 helix cause increased
exposure of LIBS mAb epitopes obscured at a hybrid domain-
β-propeller contact [20]. Hybrid domain swing-out has also
been observed using electron microscopy [15] and small-angle
X-ray scattering [9]. The extent to which different ligands
stabilize different integrin conformers and in turn transduce
agonistic effects across the plasma membrane, and the
structural mechanisms that couple extension, leg separation
and βA-domain conformational change, are not clear, but
will underpin our future understanding of how integrins
work as dynamic receptors.
Integrin-receptor cross-talk and signalling
assemblies in migrating cells
Ligand engagement by integrins initiates signalling responses
that include transduction of mechanical force to the cyto-
skeleton and spatial compartmentalization of signalling
complexes. There is now evidence for alterations in the fluxes
of almost all known signalling pathways subsequent to integ-
rin engagement, suggesting that adhesion receptor function
is integrated with other receptor systems. However, the
mechanisms responsible for converting integrin ligation into
an efficacious signal are not known. A hierarchy of recruit-
ment of signalling and cytoskeletal molecules to integrins
has been demonstrated using immunocytochemical ap-
proaches [21], but as yet evidence for ligand/agonist-specific
differences in the composition and/or organization of this
hierarchy is lacking.
Early studies employing receptor chimaeras demonstrated
that some integrin-specific functions were conferred by the
C
2004 Biochemical Society
824 Biochemical Society Transactions (2004) Volume 32, part 5
Figure 2 Indirect immunofluorescence analysis of human fibroblasts showing FAs and FBs
Cells were fixed and stained for β1integrin in a primed conformation (9EG7, green; left panels) and for α5integrin (using
the conformation-dependent mAb SNAKA51, red; middle panels). A merged image is shown on the right. While 9EG7 stains
both focal (white triangles) and FBs (yellow triangles), SNAKA51 localizes specifically to FBs, indicating a conformational
change in α5 during the transition from focal to FB.
cytoplasmic domain. On the basis of an NMR analysis of
a complex between recombinant forms of the αIIb and β3
cytoplasmic domains, it appears that both subunits are largely
unstructured, but are held together by a juxtamembrane
interaction of two α-helical regions [22]. A variety of ap-
proaches including affinity chromatography, equilibrium gel
filtration, IP blotting, synthetic peptide binding and two-
hybrid analysis have been used to identify molecules that
bind integrins (reviewed in [23]). Interacting proteins include
cytoskeletal components such as talin, α-actinin and filamin,
adapters such as paxillin, Rack-1, ICAP-1 and β3-endonexin
and kinases such as Src.
A central role in integrin signalling has been established
for talin. Genetic or translational inhibition of talin disrupts
the cytoskeleton and cell adhesion with a similar phenotype
to integrin deletions, whereas overexpression of an integrin-
binding fragment of talin induces priming triggered by mul-
tiple pathways [24] and causes structural perturbation of the
association between the integrin cytoplasmic domains [22].
Crystallization of a talin fragment–integrin β3 cytoplasmic
peptide chimaera has revealed a PTB domain-like mode of
binding that may be common to several integrin effectors
[25,26]. Since talin binds vinculin, actin and FAK, it has
the potential to co-ordinate the recruitment of many key
integrin-signalling molecules. Furthermore, talin binds and
activates PIPKIγ, which leads to increased PIP2production
and in turn activates several cytoskeletal proteins.
Integrins undergo cis interactions with a number of
different receptors, and thereby spatially regulate diverse
signalling responses (reviewed in [27]). Direct extracellular
associations with members of the TM4 family, the urokinase
receptor uPAR and CD47 have been established, as has
indirect co-clustering with several growth factor and cytokine
receptors. Cross-talk at the level of signalling occurs with
semaphorins, ephrins and syndecans. It is remarkable that
most ECM molecules possess both integrin- and syndecan-
binding sites, and a clear synergistic relationship exists
between these two families. Thus α5β1-dependent FA (focal
adhesion) formation on FN requires engagement of, and
signalling through, a syndecan co-receptor [28,29].
In spread fibroblasts adherent to FN in vitro, adhesion
signalling complexes are distributed focally rather than dif-
fusely, and are manifested as asymmetric patches, flecks and
stripes. These contact points are found all over the ventral
surface, and are usually associated with the contractile
polymers of the cytoskeleton. Detailed morphological and
functional analyses have defined three major forms of
adhesion contact: FC (focal complexes), FA and FB (fibril-
lar adhesions; Figure 2; reviewed in [30]). These contacts
reflect different stages of interaction of cells with the ECM,
and each is formed and disrupted in a cyclical manner as
cells translocate. Initially, FC form at the posterior edge of
ruffling membrane, where they anchor the short filopodial
struts and lamellipodial meshes of actomyosin that mediate
membrane protrusion. When protrusion ceases or the lamel-
lipodium retracts, FC transform into larger FA, which pro-
vide a more robust anchorage through transcellular acto-
myosin-containing stress fibres. In turn, FA evolve into FB,
which are the major sites of FN matrix deposition [31,32].
Adhesion contact-like structures have been observed in vivo
in smooth-muscle cell plaques and myotendinous junctions
and, recently, in embryonic three-dimensional ECM [33],
thereby validating the use of cell cultures for analysis.
The members of the Rho family of small GTPases are
recognized to play a central role in modulating the actin cyto-
skeleton (reviewed in [34]). Cdc42 and Rac1 promote FC
formation and membrane protrusions at the leading edge
through filopodia and lamellipodia, whereas RhoA mediates
C
2004 Biochemical Society
Signalling Outwards and Inwards 825
FA formation and causes retraction of the trailing edge. Co-
ordination between these pathways involves activation of
p190RhoGAP, which may permit membrane protrusion by
suppressing RhoA activity. Subsequently, the FC to FA
transition is dependent on RhoA and ROCK-mediated
myosin II contractility through phosphorylation of myosin
light chain. Integrin occupancy controls the activity of Rho
family GTPases and regulates their translocation to plasma
membrane microdomains [35,36], but the molecular links
between integrins and Rho GTPases are not understood.
At least 50 different proteins have been localized to FA
[37], but analyses of the differences between FC, FA and FB
are still in their infancy. FC and FA share many components,
but FC tend to lack zyxin and tensin [38], whereas FB contain
α5β1andtensin,butlackmostoftheotherFAcomponents,
including αVβ3 [32]. Thus while compositional changes do
take place as adhesion contacts mature, the signalling events
that regulate these transitions are not well understood. In
particular, how integrin activation with ligands in the ECM
modulates the assembly, maturation and turnover of different
adhesion contacts will be an important area of future study.
Preliminary evidence from our laboratory (Figure 2) suggests
that integrin conformation can vary with location in the cell,
implying that bi-directional communication is important for
co-ordinating cell–matrix integration.
This work was supported by grants from the Wellcome Trust.
References
1Hynes,R.O.(2002)Cell(Cambridge,Mass.)110,673687
2 Lee, J.O., Rieu, P., Arnaout, M.A. and Liddington, R. (1995)
Cell (Cambridge, Mass.) 80,631638
3 Xiong, J.P., Stehle, T., Diefenbach, B., Zhang, R., Dunker, R., Scott, D.L.,
Joachimiak, A., Goodman, S.L. and Arnaout, M.A. (2001) Science 294,
339–345
4Xiong,J.P.,Stehle,T.,Zhang,R.,Joachimiak,A.,Frech,M.,Goodman,S.L.
and Arnaout, M.A. (2002) Science 296,151155
5Carrell,N.A.,Fitzgerald,L.A.,Steiner,B.,Erickson,H.P.andPhillips,D.R.
(1985) J. Biol. Chem. 260,17431749
6Emsley,J.,Knight,C.G.,Farndale,R.W.,Barnes,M.J.andLiddington,R.C.
(2000) Cell (Cambridge, Mass.) 101,4756
7Takagi,J.,Strokovich,K.,Springer,T.A.andWalz,T.(2003)EMBOJ.22,
4607–4615
8 Mould, A.P., Askari, J.A. and Humphries, M.J. (2000) J. Biol. Chem. 275,
20324–20336
9 Mould, A.P., Symonds, E.J., Buckley, P.A., Grossmann, J.G., McEwan, P.A.,
Barton, S.J., Askari, J.A., Craig, S.E., Bella, J. and Humphries, M.J. (2003)
J. Biol. Chem. 278,3999339999
10 Humphries, M.J. (2000) Biochem. Soc. Trans. 28,311339
11 Mastrangelo, A.M., Homan, S.M., Humphries, M.J. and LaFlamme, S.E.
(1999) J. Cell Sci. 112,217229
12 Humphries, M.J., McEwan, P.A., Barton, S.J., Buckley, P.A., Bella, J. and
Mould, A.P. (2003) Trends Biochem. Sci. 28,313320
13 Xiong, J.P., Stehle, T., Goodman, S.L. and Arnaout, M.A. (2003)
Blood 102,11551159
14 Beglova, N., Blacklow, S.C., Takagi, J. and Springer, T.A. (2002)
Nat. Struct. Biol. 9,282287
15 Takagi, J., Petre, B.M., Walz, T. and Springer, T.A. (2002)
Cell (Cambridge, Mass.) 110,599611
16 Lu, C., Takagi, J. and Springer, T.A. (2001) J. Biol. Chem. 276,
14642–14648
17 Takagi, J., Erickson, H.P. and Springer, T.A. (2001) Nat. Struct. Biol. 8,
412–416
18 Kim, M., Carman, C.V. and Springer, T.A. (2003) Science 301,17201725
19 Mould, A.P., Askari, J.A., Barton, S., Kline, A.D., McEwan, P.A., Craig, S.E.
and Humphries, M.J. (2002) J. Biol. Chem. 277,1980019805
20 Mould, A.P., Barton, S.J., Askari, J.A., McEwan, P.A., Buckley, P.A., Craig,
S.E. and Humphries, M.J. (2003) J. Biol. Chem. 278,1702817035
21 Miyamoto, S., Teramoto, H., Gutkind, J.S. and Yamada, K.M. (1996)
J. Cell Biol. 135,16331642
22 Vinogradova, O., Velyvis, A., Velyviene, A., Hu, B., Haas, T., Plow, E. and
Qin, J. (2002) Cell (Cambridge, Mass.) 110,587597
23 Liu, S., Calderwood, D.A. and Ginsberg, M.H. (2000) J. Cell Sci. 113,
3563–3571
24 Tadokoro, S., Shattil, S.J., Eto, K., Tai, V., Liddington, R.C., de Pereda, J.M.,
Ginsberg, M.H. and Calderwood, D.A. (2003) Science 302,103106
25 Calderwood, D.A., Fujioka, Y., de Pereda, J.M., Garcia-Alvarez, B.,
Nakamoto, T., Margolis, B., McGlade, C.J., Liddington, R.C. and
Ginsberg, M.H. (2003) Proc. Natl. Acad. Sci. U.S.A. 100,22722277
26 Garcia-Alvarez, B., de Pereda, J.M., Calderwood, D.A., Ulmer, T.S.,
Critchley, D., Campbell, I.D., Ginsberg, M.H. and Liddington, R.C. (2003)
Mol. Cell 11,4958
27 Giancotti, F.G. and Tarone, G. (2003) Annu. Rev. Cell Dev. Biol. 19,
173–206
28 Mostafavi-Pour, Z., Askari, J.A., Parkinson, S.J., Parker, P.J., Ng, T.T. and
Humphries, M.J. (2003) J. Cell Biol. 161,155167
29 Woods, A., Couchman, J.R., Johansson, S. and Hook, M. (1986) EMBO J. 5,
665–670
30 Geiger, B., Bershadsky, A., Pankov, R. and Yamada, K.M. (2001) Nat. Rev.
Mol. Cell Biol. 2,793805
31 Pankov, R., Cukierman, E., Katz, B.Z., Matsumoto, K., Lin, D.C., Lin, S.,
Hahn, C. and Yamada, K.M. (2000) J. Cell Biol. 148,10751090
32 Zamir, E., Katz, M., Posen, Y., Erez, N., Yamada, K.M., Katz, B.Z., Lin, S.,
Lin, D.C., Bershadsky, A., Kam, Z. et al. (2000) Nat. Cell Biol. 2,191196
33 Cukierman, E., Pankov, R., Stevens, D.R. and Yamada, K.M. (2001)
Science 294,17081712
34 Etienne-Manneville, S. and Hall, A. (2002) Nature (London) 420,
629–635
35 Del Pozo, M.A., Alderson, N.B., Kiosses, W.B., Chiang, H.H., Anderson,
R.G. and Schwartz, M.A. (2004) Science 303,839842
36 Del Pozo, M.A., Kiosses, W.B., Alderson, N.B., Meller, N., Hahn, K.M. and
Schwartz, M.A. (2002) Nat. Cell Biol. 4,232239
37 Zamir, E. and Geiger, B. (2001) J. Cell Sci. 114,35833590
38 Zaidel-Bar, R., Ballestrem, C., Kam, Z. and Geiger, B. (2003) J. Cell Sci.
116,46054613
Received 9 July 2004
C
2004 Biochemical Society
... With the aim of evaluating MSC osteogenic commitment, we used a combined system for simultaneous stimulation of NaBC1 and α 5 β 1 and α v β 3 integrins 28 . Boron (B) in the form of Sodium Tetraborate Decahydrate (borax) was used dissolved in the culture media for NaBC1 stimulation, and fibronectin (FN) for FNbinding integrin activation. ...
... We evaluated the combined effect of stimulating NaBC1 together with FN-binding integrins after 3 h of culture using serum-depleted culture media to ensure that initial cell-material interaction was exclusively FN-mediated, and so targeting α 5 β 1 and α v β 3 integrins 28 . Cells were seeded at low density (5000 cells cm −2 ) to favor cell-material interaction and minimize cell-cell contacts in order to quantify focal adhesions (FA). ...
Article
Full-text available
The intrinsic properties of mesenchymal stem cells (MSCs) make them ideal candidates for tissue engineering applications. Efforts have been made to control MSC behavior by using material systems to engineer synthetic extracellular matrices and/or include soluble factors in the media. This work proposes a simple approach based on ion transporter stimulation to determine stem cell fate that avoids the use of growth factors. Addition of borax alone, transported by the NaBC1-transporter, enhanced MSC adhesion and contractility, promoted osteogenesis and inhibited adipogenesis. Stimulated-NaBC1 promoted osteogenesis via the BMP canonical pathway (comprising Smad1/YAP nucleus translocation and osteopontin expression) through a mechanism that involves simultaneous NaBC1/BMPR1A and NaBC1/α5β1/αvβ3 co-localization. We describe an original function for NaBC1 transporter, besides controlling borate homeostasis, capable of stimulating growth factor receptors and fibronectin-binding integrins. Our results open up new biomaterial engineering approaches for biomedical applications by a cost-effective strategy that avoids the use of soluble growth factors.
... This is important during development for CMs' differentiation and maturation. Later on, adhesion complexes provide the signals that sustain tissue formation and turnover of ECM proteins (Peters et al., 1994, Humphries et al., 2004. Mimicking those process seems important for myocardial regeneration, which otherwise seems to be very limited in the cardiac tissue, as reviewed in (Leong et al., 2017). ...
... Integrins are transmembrane cellsurface glycoproteins of type I that are heterodimers made up of non-covalently linked a and b subunits (13). These subunits are constructed from different domains, with each subunit containing a large ectodomain that is important for ligand binding, a single transmembrane domain, and a cytoplasmic domain (cytoplasmic tail) that, in most cases, ranges in size from 20 to 70 amino acid residues (2,14,15). ...
Article
Full-text available
Integrins are a large group of cell-surface proteins that are classified as transmembrane proteins. Integrins are classified into different types based on sequence variations, leading to structural and functional diversity. They are broadly distributed in animals and have a wide range of biological functions such as cell-to-cell communication, intracellular cytoskeleton organization, cellular signaling, immune responses, etc. Integrins are among the most abundant cell surface proteins in insects, exhibiting their indispensability in insect physiology. Because of their critical biological involvement in physiological processes, they appear to be a novel target for designing effective pest control strategies. In the current literature review, we first discuss the discovery and expression responses of integrins against various types of pathogens. Secondly, we examine the specific biological roles of integrins in controlling microbial pathogens, such as phagocytosis, encapsulation, nodulation, immune signaling, and so on. Finally, we describe the possible uses of integrins to control agricultural insect pests.
... Each of these subunits is composed of large extracellularly located amino acid (AA) sequence, together with ∼740-780 AAs as part of the β kinds of subunits. There is also a singular domain of the structure located across the membrane consisting of nearly 20 AAs together with a short structure part located in the cytoplasm having a chain length of 40-50 AAs ( Figure 2) [8,9]. However, an exceptionally high chain length of 1,000 AAs is found for the β4 subunits [10]. ...
Article
Full-text available
The integrins belong to the cell-surface polypeptide family and are the mediating partners among the cells, and extracellular matrix (ECM). They are also involved in the biological processes of cell migration, wound healing, blood clotting, immunological response generation, tissue morphogenesis, leucocyte reticulations, and angiogenesis and are therefore very relevant in stem cell technology and are useful as biomarkers, diagnostic probes, and drug-target ligands. The ανβ3 ( alpha-nu-beta 3) integrin antagonists are an excellent target example for designing and developing newer drug candidates, drug leads and templates for various diseases, and physiological malfunctioning, including cancers. The current review examines the ανβ3 integrin structural features involved in the drug design and its antagonistic ligands and highlights the development of anti-ανβ3 integrin-antagonists as nano-architectural design-based nanomedicine, especially for cancer chemotherapy. The perspectival review discusses the ανβ3 integrin structure, mode of action, involved pathways, and the concepts utilized in nanomedicine design, and ligands related to integrins. It also covers the latest thyrointegrin approaches toward the development of anti-angiogenesis agents and entails the anti-angiogenesis approach to cancer growth inhibition through targeting by the anti-integrin ligands and related chemical entities. The current perspective on the nano-architectural design approach for the known anti-integrin compounds is also outlined.
... The signal transduction and these interactions are mainly facilitated by integrins, a large family of heterodimeric transmembrane cell surface receptors that are capable to induce signaling upon ligand binding (Fig.3A) [214]. Integrins interact with the ECM and cytoskeleton, in collaboration with other receptors such as growth factor receptors [215][216][217]. The heparin-induced up-regulation of HSPG may occur through its interaction with ECM molecules, such as fibronectin, which sequentially is recognized by integrins via the primary sequence motif Arg-Gly-Asp (RGD) [214,218]. ...
Article
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
... Fn matrix assembly requires the polymerization of multiple molecules together into fibrils [55]. The self-assembly of Fn fibrils is a cell mediated process that involves Fn recognition by cell surface receptors such as integrins, and the elongation of its structure to expose cyptic binding sites necessary for fibrillogenesis [3,56,57]. The Fn cryptic binding sites can be exposed by tension, heat denaturation, or cleavage of specific domains (e.g., FnIII) [58,59]. ...
Article
Full-text available
We previously reported that corneal fibroblasts within 3D fibrin matrices secrete, bind, and organize fibronectin into tracks that facilitate cell spreading and migration. Other cells use these fibronectin tracks as conduits, which leads to the development of an interconnected cell/fibronectin network. In this study, we investigate how cell-induced reorganization of fibrin correlates with fibronectin track formation in response to two growth factors present during wound healing: PDGF BB, which stimulates cell spreading and migration; and TGFβ1, which stimulates cellular contraction and myofibroblast transformation. Both PDGF BB and TGFβ1 stimulated global fibrin matrix contraction (p < 0.005); however, the cell and matrix patterning were different. We found that, during PDGF BB-induced cell spreading, fibronectin was organized simultaneously with the generation of tractional forces at the leading edge of pseudopodia. Over time this led to the formation of an interconnected network consisting of cells, fibronectin and compacted fibrin tracks. Following culture in TGFβ1, cells were less motile, produced significant local fibrin reorganization, and formed fewer cellular connections as compared to PDGF BB (p < 0.005). Although bands of compacted fibrin tracks developed in between neighboring cells, fibronectin labeling was not generally present along these tracks, and the correlation between fibrin and fibronectin labeling was significantly less than that observed in PDGF BB (p < 0.001). Taken together, our results show that cell-induced extracellular matrix (ECM) reorganization can occur independently from fibronectin patterning. Nonetheless, both events seem to be coordinated, as corneal fibroblasts in PDGF BB secrete and organize fibronectin as they preferentially spread along compacted fibrin tracks between cells, producing an interconnected network in which cells, fibronectin and compacted fibrin tracks are highly correlated. This mechanism of patterning could contribute to the formation of organized cellular networks that have been observed following corneal injury and refractive surgery.
Article
Analysis of the participation of short peptides GER and FGER containing common tripeptide fragment in the regulation of adhesive response of CHO-K1 cells was conducted. Both peptides stimulated cell adhesion both to untreated plastic and to gelatin-coated plastic, but did not change cell attachment to poly-L-lysine-coated plastic. Tripeptide GER had larger stimulation effect on cell adhesion to untreated plastic. Peptide FGER increased the rate of cell attachment to gelatin in a wider range of concentrations as compared to adhesion to untreated plastic. Variativity of cell spreading to different substrates under peptide action was demonstrated. On untreated plastic both investigated peptides practically in equal extent stimulated cell spreading. On gelatin peptide FGER kept the stimulation effect on cell spreading, but peptide GER partly inhibited cell spreading as compared to cell spreading on untreated plastic. It was established that insertion of additional N-terminal hydrophobic amino acid residue Phe to tripeptide fragment GER changes the regulatory activity of peptide at the cell adhesion model depending on the stage of cell connection with substrate and/or on substrate properties. The structural-functional activity of investigated short peptides on the instance of different structural components of adhesive structures is discussed.
Article
Use of stem cells for treating degenerative diseases like osteoarthritis has been gaining popularity. Glucosamine Sulphate is a nutritional supplement used for relieving inflammation during Osteoarthritis. Aloe vera juice is a widely consumed drink used in traditional practices to help reduce inflammation. In this experiment, the effect of Glucosamine Sulphate and Aloe vera on 2D and 3D proliferation of MSC cells was studied. Initial study to determine a suitable matrix for growing MSC cells was performed by growing the cells on 5mg/ml and 10mg/ml collagen and fibronectin coated wells for seven days. Cells adhered the least on fibronectin 10ug/ml and cells proliferated the highest on fibronectin at 10ug/ml, thus establishing fibronectin 10ug/ml as the matrix for the study. The effect of Glucosamine Sulphate on 2D proliferation of MSC cells growing on 10ug/ml fibronectin matrix was studied by exposing the cells to 0mg/ml, 0.1mg/ml, 0.5mg/ml and 1 mg/ml of Glucosamine Sulphate for seven days. The cells adhered the least on 0.5mg/ml and proliferated the highest on 0.5mg/ml as well (p<0.05). The effect of Aloe vera on MSC cells grown on fibronectin at 10ug/ml was studied by using concentrations of 0%, 2%, 5% and 10% Aloe vera juice for seven days. It was found that cell proliferation radically decreased in all conditions after Day 4 and the least proliferation was observed at 10%. 3D proliferation of MSC cells in a fibrin construct was studied with 0.1 mg/ml of Glucosamine Sulphate and 2% and 5% Aloe vera juice. It was observed that 5% Aloe vera juice allowed highest proliferation of cells, followed by 0.1 mg/ml Glucosamine Sulphate and 2% Aloe vera. This indicated that further analysis with 2% and 5% Aloe vera along with 0.1mg/ml as threshold could help identify an optimum dose range for uptake of Glucosamine Sulphate with Aloe vera juice to enhance relief from inflammation during osteoarthritis
Article
Full-text available
Here we use time-lapse microscopy to analyse cell–matrix adhesions in cells expressing one of two different cytoskeletal proteins, paxillin or tensin, tagged with green fluorescent protein (GFP). Use of GFP–paxillin to analyse focal contacts and GFP–tensin to study fibrillar adhesions reveals that both types of major adhesion are highly dynamic. Small focal contacts often translocate, by extending centripetally and contracting peripherally, at a mean rate of 19 micrometres per hour. Fibrillar adhesions arise from the medial ends of stationary focal contacts, contain 51 integrin and tensin but not other focal-contact components, and associate with fibronectin fibrils. Fibrillar adhesions translocate centripetally at a mean rate of 18 micrometres per hour in an actomyosin-dependent manner. We propose a dynamic model for the regulation of cell–matrix adhesions and for transitions between focal contacts and fibrillar adhesions, with the ability of the matrix to deform functioning as a mechanical switch.
Article
Full-text available
Integrin-mediated cell adhesions provide dynamic, bidirectional links between the extracellular matrix and the cytoskeleton. Besides having central roles in cell migration and morphogenesis, focal adhesions and related structures convey information across the cell membrane, to regulate extracellular-matrix assembly, cell proliferation, differentiation, and death. This review describes integrin functions, mechanosensors, molecular switches and signal-transduction pathways activated and integrated by adhesion, with a unifying theme being the importance of local physical forces.
Article
Currently >50 proteins have been reported to be associated with focal contacts and related ECM adhesions. Most of these contain multiple domains through which they can interact with different molecular partners, potentially forming a dense and heterogeneous protein network at the cytoplasmic faces of the adhesion site. The molecular and structural diversity of this ‘submembrane plaque’ is regulated by a wide variety of mechanisms, including competition between different partner proteins for the same binding sites, interactions triggered or suppressed by tyrosine phosphorylation, and conformational changes in component proteins, which can affect their reactivity. Indeed, integrin-mediated adhesions can undergo dynamic changes in structure and molecular properties from dot-like focal complexes to stress-fiber-associated focal contacts, which can further ‘mature’ to form fibronectin-bound fibrillar adhesions. These changes are driven by mechanical force generated by the actin- and myosin-containing contractile machinery of the cells, or by external forces applied to the cells, and regulated by matrix rigidity.
Article
Integrins mediate cell adhesion, migration, and a variety of signal transduction events. These integrin actions can overlap or even synergize with those of growth factors. We examined for mechanisms of collaboration or synergy between integrins and growth factors involving MAP kinases, which regulate many cellular functions. In cooperation with integrins, the growth factors EGF, PDGF-BB, and basic FGF each produced a marked, transient activation of the ERK (extracellular signal-regulated kinase) class of MAP kinase, but only if the integrins were both aggregated and occupied by ligand. Transmembrane accumulation of total tyrosine-phosphorylated proteins, as well as nonsynergistic MAP kinase activation, could be induced by simple integrin aggregation, whereas enhanced transient accumulation of the EGF-receptor substrate eps8 required integrin aggregation and occupancy, as well as EGF treatment. Each type of growth factor receptor was itself induced to aggregate transiently by integrin ligand-coated beads in a process requiring both aggregation and occupancy of integrin receptors, but not the presence of growth factor ligand. Synergism was also observed between integrins and growth factors for triggering tyrosine phosphorylation of EGF, PDGF, and FGF receptors. This collaborative response also required both integrin aggregation and occupancy. These studies identify mechanisms in the signal transduction response to integrins and growth factors that require various combinations of integrin aggregation and ligands for integrin or growth factor receptors, providing opportunities for collaboration between these major regulatory systems.
Article
Activation of the ligand binding function of integrin heterodimers requires transmission of an “inside-out” signal from their small intracellular segments to their large extracellular domains. The structure of the cytoplasmic domain of a prototypic integrin αIIbβ3 has been solved by NMR and reveals multiple hydrophobic and electrostatic contacts within the membrane-proximal helices of its α and the β cytoplasmic tails. The interface interactions are disrupted by point mutations or the cytoskeletal protein talin that are known to activate the receptor. These results provide a structural mechanism by which a handshake between the α and the β cytoplasmic tails restrains the integrin in a resting state and unclasping of this interaction triggers the inside-out conformational signal that leads to receptor activation.
Article
We have determined the high resolution crystal structure of the A domain from the a chain of integrin CR3. The domain adopts a classic α/β “Rossmann” fold and contains an unusual Mg2+ coordination site at its surface. One of the coordinating ligands is the glutamate side chain from another A domain molecule. We suggest that this site represents a general metal ion-dependent adhesion site (MIDAS) for binding protein ligands. We further propose that the subunits of integrins contain a MIDAS motif within a modified A domain. Our crystal structure will allow reliable models to be built for other members of the A domain superfamily and should facilitate development of novel adhesion modulatory drugs.
Article
Integrins are adhesion molecules that convey signals both to and from the cytoplasm across the plasma membrane. In resting cells, integrins in a low affinity state can be activated by 'inside-out signaling', in which signals affecting integrin heterodimer cytoplasmic domains cause a conformational change in the integrin ligand-binding headpiece connected to the membrane by two long, approximately 16 nm stalks. Here we demonstrate a mechanism for conveying a conformational change over the long distance from the plasma membrane to the headpiece. We prepared soluble, alpha5beta1 integrin heterodimer extracellular fragments in which interactions between alpha- and beta-subunit cytoplasmic domains were replaced with an artificial clasp. Release of this C-terminal clasp by specific protease cleavage resulted in an approximately 14 nm separation of the stalks coupled to increased binding to fibronectin. This activation did not require any associated molecules or clustering and was observed with physiological concentrations of divalent cations. These findings suggest that the overall mechanism for integrin inside-out activation involves the spatial separation of the cytoplasmic and/or transmembrane domains.
Article
We have determined the crystal structure of a complex between the I domain of integrin α2β1 and a triple helical collagen peptide containing a critical GFOGER motif. Three loops on the upper surface of the I domain that coordinate a metal ion also engage the collagen, with a collagen glutamate completing the coordination sphere of the metal. Comparison with the unliganded I domain reveals a change in metal coordination linked to a reorganization of the upper surface that together create a complementary surface for binding collagen. Conformational changes propagate from the upper surface to the opposite pole of the domain, suggesting both a basis for affinity regulation and a pathway for signal transduction. The structural features observed here may represent a general mechanism for integrin–ligand recognition.
Article
Cells adhere to the extracellular matrix throughout most of their lifetime. This close, intimate contact with the matrix exerts an extraordinary control on the behavior of cells, determining whether they move or stay put, proliferate or remain quiescent, and even live or die. Attachment to the matrix not only enables cells to respond to soluble growth factors and cytokines but also determines the nature of the response. The integrins are a large family of receptors that attach cells to the matrix, organize their cytoskeleton, and cooperate with receptor protein tyrosine kinases to regulate cell fate. Research on integrin signaling is beginning to explain the complex and specific effects that the extracellular matrix exerts on cells.