ArticlePDF Available

Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases

Authors:

Abstract and Figures

The passage of leukocytes through basement membranes involves proteolytic degradation of extracellular matrix (ECM) components executed by focalized proteolysis. We have investigated whether the migration of leukocytes through 3-dimensional collagenous tissue scaffolds requires similar ECM breakdown. Human T blasts and SupT1 lymphoma cells expressed mRNA of MMP-9, MT1-MMP, MT4-MMP, cathepsin L, uPA, and uPAR as well as ADAM-9, -10, -11, -15, and -17. Upon long-term migration within 3-dimensional collagen matrices, however, no in situ collagenolysis was obtained by sensitive fluorescein isothiocyanate-collagen fragmentation analysis and confocal fluorescence/backscatter microscopy. Consistent with nonproteolytic migration, T-cell crawling and path generation were not impaired by protease inhibitor cocktail targeting MMPs, serine proteases, cysteine proteases, and cathepsins. Dynamic imaging of cell-ECM interactions showed T-cell migration as an amoeba-like process driven by adaptive morphology, crawling along collagen fibrils (contact guidance) and squeezing through pre-existing matrix gaps by vigorous shape change. The concept of nonproteolytic amoeboid migration was confirmed for multicomponent collagen lattices containing hyaluronan and chondroitin sulfate and for other migrating leukocytes including CD8+ T blasts, monocyte-derived dendritic cells, and U937 monocytic cells. Together, amoeboid shape change and contact guidance provide constitutive protease-independent mechanisms for leukocyte trafficking through interstitial tissues that are insensitive toward pharmacologic protease inhibitors.
Content may be subject to copyright.
doi:10.1182/blood-2002-12-3791
Prepublished online July 10, 2003;
2003 102: 3262-3269
Katarina Wolf, Regina Müller, Stefan Borgmann, Eva.-B. Bröcker and Peter Friedl
and other proteases
through fibrillar collagen is independent of matrix remodeling by MMPs
Amoeboid shape change and contact guidance: T-lymphocyte crawling
http://bloodjournal.hematologylibrary.org/content/102/9/3262.full.html
Updated information and services can be found at:
(5012 articles)Immunobiology
(2497 articles)Hemostasis, Thrombosis, and Vascular Biology
(1086 articles)Gene Expression
(790 articles)Cell Adhesion and Motility
Articles on similar topics can be found in the following Blood collections
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
Copyright 2011 by The American Society of Hematology; all rights reserved.
Washington DC 20036.
by the American Society of Hematology, 2021 L St, NW, Suite 900,
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
IMMUNOBIOLOGY
Amoeboid shape change and contact guidance: T-lymphocyte crawling through
fibrillar collagen is independent of matrix remodeling by MMPs and other proteases
Katarina Wolf, Regina Mu¨ller, Stefan Borgmann, Eva.-B. Bro¨cker, and Peter Friedl
The passage of leukocytes through base-
ment membranes involves proteolytic
degradation of extracellular matrix (ECM)
components executed by focalized prote-
olysis. We have investigated whether the
migration of leukocytes through 3-dimen-
sional collagenous tissue scaffolds re-
quires similar ECM breakdown. Human T
blasts and SupT1 lymphoma cells ex-
pressed mRNAof MMP-9, MT1-MMP, MT4-
MMP, cathepsin L, uPA, and uPAR as well
as ADAM-9, -10, -11, -15, and -17. Upon
long-term migration within 3-dimensional
collagen matrices, however, no in situ
collagenolysis was obtained by sensitive
fluoresceinisothiocyanate–collagen frag-
mentation analysis and confocal fluores-
cence/backscatter microscopy. Consis-
tent with nonproteolytic migration, T-cell
crawling and path generation were not
impaired by protease inhibitor cocktail
targeting MMPs, serine proteases, cys-
teine proteases, and cathepsins. Dy-
namic imaging of cell-ECM interactions
showed T-cell migration as an amoeba-
like process driven by adaptive morphol-
ogy, crawling along collagen fibrils (con-
tact guidance) and squeezing through
pre-existing matrix gaps by vigorous
shape change.The conceptof nonproteo-
lytic amoeboid migration was confirmed
for multicomponentcollagen latticescon-
taining hyaluronan and chondroitin sul-
fate and for other migrating leukocytes
including CD8
T blasts, monocyte-de-
rived dendritic cells, and U937 monocytic
cells. Together, amoeboid shape change
and contact guidance provide constitu-
tive protease-independent mechanisms for
leukocyte trafficking through interstitial tis-
sues that are insensitive toward pharma-
cologic protease inhibitors. (Blood. 2003;
102:3262-3269)
© 2003 by TheAmerican Society of Hematology
Introduction
The migration and recirculation of T lymphocytes through the
tissues is a multistep process that requires cell adhesion coupled
with cellular strategies to overcome physical tissue barriers.
1
The
principles of T-cell migration follow the paradigm of “amoeboid”
movement established for the single cell state of the lower eucaryote
Dictyostelium discoideum.
2,3
Amoeboid movement is a fast low-
affinity migration type driven by a roundish yet flexible cell
morphology, dynamic and polarized pseudopod protrusions and
retractions, which are independent of focal contact formation and
stress fibers.
2
Similar to Dictyostelium, migrating T lymphocytes
show an elliptoid polarized shape with a smooth yet dynamically
ruffling leading edge, and an additional trailing uropod.
4
This shape
generates fast (5-25 m/min) low-affinity gliding across surfaces
and through 3-dimensional (3D) collagenous scaffolds
4
driven by a
diffuse cortical actin cytoskeleton along the inner leaflet of the
plasma membrane devoid of focal contacts at substrate interactions
and stress fibers.
5,6
Amoeboid T-cell migration within extracellular
matrix (ECM) occurs independently of 1 integrin–mediated
adhesion both in vitro
6
and in vivo,
7
suggesting differences of the
molecular basis between the movement of lymphocytes and other
mobile cells, such as fibroblasts and tumor cells.
8,9
After transendothelial migration, T cells and other leukocytes
enter the fibrillar and reticular networks of the extracellular matrix
in peripheral and lymphatic tissues, predominantly composed of
fibrillar type I and III collagens.
10,11
In many cell types, adhesion
and cytoskeletal dynamics in collagenous tissues are coordinated
with proteolytic strategies to lower the physical matrix resistance.
Fibroblasts and tumor cells, as well as neutrophils, migrate across
ECM proteins such as gelatin and fibronectin while simultaneously
generating localized tracks of proteolytic substratedegradation,
12-15
prompting concepts on pericellular proteolysisas a cellular strategy
to overcome matrix barriers.
16,17
Pericellular matrix degradation is
mediated by secreted as well as cell-bound matrix proteases,
including matrix metalloproteinases (MMPs), serine proteases, and
cathepsins.
13,14,18
In tumor cells interacting with ECM, cell surface
MMPs and serine proteases become enriched at the surface of
leading pseudopods (“invadopodia”) to cooperate with integrins
and other adhesion receptors in a focalized manner.
12,17,18
Focaliza-
tion of ECM cleavage can occur by 2 principal mechanisms:
membrane-bound proteases such as MT-MMPs coclustering with
1or3 integrins at contacts to substrate,
18,19
or heterophilic
binding of soluble MMPs to cell surface receptors, such as MMP-2
binding to MT1-MMP or urokinase-type plasminogen activator
(uPA) to its receptor uPAR.
20,21
Whereas focal contacts recruit
proteases to substrate interactions in fibroblasts and tumor
cells,
18,19,22
it remains unresolved how pericellular proteolysis is
spatially and temporally achieved by short-lived and molecular
diffuse interactions to ECM substrate, as developed by rapidly
moving leukocytes that generate migration velocities that exceed
those of fibroblasts and tumor cells by 20- to 40-fold.
8
From the Department of Dermatology, University of Wu¨rzburg, Germany; and
the Department of Microbiology, University of Tu¨bingen, Germany.
Submitted December 17, 2002; accepted June 19, 2003 . Prepublished online
as Blood First Edition Paper, July 10, 2003; DOI 10.1182/blood-2002-12-3791.
Supported by the Deutsche Forschungsgemeinschaft (FR 5511/2-2 and 2-3).
K.W. was additionally supported by the foundation Evangelisches Studienwerk
e.V., Haus Villigst.
The online version of the article contains a data supplement.
Reprints: Peter Friedl, Department of Dermatology, University of Wu¨rzburg,
Josef-Schneider-Str 2, 97080 Wu¨rzburg, Germany; e-mail: peter.fr@mail.uni-
wuerzburg.de.
The publication costs of this article were defrayed in part by page charge
payment. Therefore, and solely to indicate this fact, this article is hereby
marked ‘‘advertisement’’in accordance with 18 U.S.C. section 1734.
© 2003 by TheAmerican Society of Hematology
3262 BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
In monocytes, dendritic cells, and neutrophils, proteases from
different classes are constitutively expressed, including MMPs,
sheddases (eg, a disintegrin and a metalloproteinase [ADAMs]),
cysteine, and aspartatic proteases (eg, cathepsins), as well as serine
proteases (eg, cathepsin G, urokinase-type plasminogen activator,
human leukocyte elastase [HLE]).
17,23,24
Resting peripheral T
lymphocytes express a more restricted spectrum of proteases and
only upon activation do T cells up-regulate or de novo express
MMP-2, MMP-9, cathepsins, members of the PA/plasmin-system,
and HLE.
25-27
The penetration of T cells and other leukocytes
through basement membrane equivalents (matrigel) in vitro is
facilitated by active MMP-2 and MMP-9,
26,28
however it remains
subject to debate in which tissue compartments and under which
conditions matrix proteases contribute to transendothelial migra-
tion in vitro and in vivo.
29-32
Although focalized proteolysis, as established for stromal cells,
is an important mechanism supporting cellmigration through tissue
scaffolds, alternative pathways for bypassing ECM barriers might
exist, depending on cell type and ECM environment. In the present
study, we used rapidly moving T cells and other leukocytes to study
the expression of MMPsand additional proteases and their function
in migration through 3D brillar collagen matrices. Although we
initially aimed at the mechanisms of focalized proteolysis, we
followed up on the unexpected nding of undiminished leukocyte
migration in the presence of protease inhibitors and show how
amoeboid migration provides a nonproteolytic mechanism to over-
come 3D collagenous tissue barriers.
Materials and methods
Antibodies, cells, and cell culture
For ow cytometry, rabbit IgG-polyclonal antiMT1-MMPAb (Chemicon,
Hofheim, Germany) and isotypic rabbit IgG (Sigma, Taufenkirchen,
Germany) were used.
Human peripheral blood mononuclear cells (PBMCs) were obtained
from peripheral blood from different healthy donors (by density centrifuga-
tion on a 1.077 g/mL coll gradient (Lymphoprep; Axis-Shield, Oslo,
Norway), washed, and stimulated for 3 days with 1.25 g/mLConcavalinA
(Oncogene, Schwalbach, Germany) and 100 U/mL IL-2 (Strathman Bio-
tech, Hamburg, Germany) in RPMI medium. CD4
and CD8
T cells from
stimulated PBMC cultures were obtained by positive immunomagnetic
selection.
33,34
Purity of isolated CD4
and CD8
populations was 95%-
99%, as determined by ow cytometry.
34
Human monocytederived
dendritic cells (DCs) were generated from peripheral blood of different
donors by cultivation in the presence of 5% autologous serum, granulocyte-
macrophage colony-stimulating factor (GM-CSF), and interleukin-4 (IL-4)
for 10 days and terminal maturation using tumor necrosis factor (TNF),
IL-1, IL-6, and prostaglandin E
2
(PGE
2
) after 7 days.
33
The human lymphoma CD4
T-cell line SupT1
26
was obtained from the
Deutsche Sammlung fu¨r Mikroorganismen und Zellkulturen (DSMZ,
Braunschweig, Germany). SupT1 cells and U937 monocytic cells were
cultured as suspension in RPMI medium (PAN, Heidenheim, Germany).
HT-1080/MT1 brosarcoma cells overtransfected with MT1-MMP
21
were
used as collagenolytic positive controls (kindly provided by Dr A. J.
Strongin, The Burnham Institute, La Jolla, CA). If not stated otherwise, all
primary cells and cell lines were cultured in medium containing 50 U/mL
penicillin and 50 g/mL streptomycin (PAN, Verwiers, Belgium) and 10%
heat-inactivated fetal calf serum (FCS) (Biowhittaker, Verwiers, Belgium)
at 37°C in humidied 5% CO
2
atmosphere. HT-1080/MT1 cells were
cultured in DMEM (PAN) containing 0.2 mg/mLG418 (Oncogene).
Collagen matrix migration assay, cell tracking, and cell viability
3D collagen matrix cultures (collagen concentration: 1.67 mg/mL) of
spontaneously migrating cells were monitored by time-lapse videomicros-
copy.
34
The collagen (Vitrogen, Cohesion, Palo Alto, CA) was resistant to
trypsin and sensitive to degradation by MT1-MMP, conrming its native
state as shown by sodium dodecyl sulfatepolyacrylamide gel electrophore-
sis (SDS-PAGE) and silver staining (C. Overall, E. Tam, unpublished, May
2002). In some experiments, multicomponent lattices were generated by
copolymerizing dermal collagen (1.67 mg/mL), human umbilical high-
molecular-weight cord hyaluronan (1 mg/mL; Sigma) and chondroitin-4-
sulfate (20 mg/mL; Sigma), as described.
35
For all cell types spontaneous
migration was investigated except U937 cells, which were stimulated by
lysophosphatidic acid (0.5 M; Sigma).
Locomotor parameters were obtained by computer-assisted cell track-
ing and reconstruction of the xy coordinates of cell paths for a step interval
of 1 (lymphocytes; Sup T1 cells)
34
or 3 minutes (U937 cells; DCs). The
average velocity and percentage of locomoting cells in a population were
calculated from each step interval of randomly selected cells divided by the
number of cells.
34
Cell viability was routinely assessed after migration experiments. Cells
were released from the collagen lattice by collagenase digestion (Clos-
tridium histolyticum collagenase type I; Sigma), stained by propidium
iodide (Sigma), and analyzed by ow cytometry.
Reverse transcriptasepolymerase chain reaction
Total RNA from ConA-blasts and Sup-T1 cells grown in liquid culture was
isolated using the RNeasy kit (Qiagen, Hilden, Germany). One microgram
of total RNA was reversely transcribed (1st Strand cDNA Synthesis Kit;
Roche Diagnostics, Mannheim, Germany) to cDNA using AMV Reverse
Transcriptase (1000 U/mL), dNTPs (1 mM each), MgCl
2
(5 mM), random
primer p(dN)
6
(80 g/mL), gelatin (10 g/mL), RNase inhibitor (2500
U/mL), and reaction buffer (10 mM Tris [tris(hydroxymethyl)aminometh-
ane]; 50 mM KCl; pH8,3). Each 0.25 g cDNAwas amplied by PCR in a
reaction using bacterial recombinant Taq DNA polymerase (12.5 U/mL),
dNTPs (0.2 mM), MgCl
2
(1.5 mM), sense and antisense primer (0.2 M
each) (primer sequences and references can be viewed in Supplemental
Table S1 on the Blood website; see the Supplemental Materials link at the
top of the online article) and reaction buffer (10 mM Tris-HCl, pH 8.8; 50
mM KCl; 0.08% NP40; Life Technologies, Karlsruhe, Germany). Unique-
ness of primer region was conrmed using the National Center for
Biotechnology Information blastn program. PCR was performed for 30
cycles. Absence of DNA contamination was conrmed by subjecting total
cell lysates (0.25 g total RNA) to PCR in the absence of reverse
transcriptase. The resulting PCR products were analyzed by electrophoresis
in a 2% agarose gel and stained with ethidium bromide (1 g/mL).
Specicity was determined by the length of each PCR product.
Protease inhibitors
Broad-spectrum MMP inhibitor BB-2516 (marimastat) was kindly pro-
vided by British Biotech (British Biotech, Oxford, United Kingdom).
BB-2516 blocks most MMPsand to some extentADAMs, atnM or low M
ranges.
36
Further protease inhibitors were aprotinin, trans-epoxysucciny-L-
leucylamide-(4-guanidino)butane (E-64), pepstatinA(all from Sigma), and
leupeptin (Molecular Probes, Leiden, The Netherlands). Inhibitor cocktail
was used at the following concentrations:marimastat, E-64,and pepstatinA
at 20 M; aprotinin 0.7 M; leupeptin 2 M (for inhibitor specicity,
established inhibitory concentration range, and references, see Supplemen-
tal Table S2).
Detection of collagenolysis
The efciency of MMPinhibitor marimastat was controlled by zymography
using recombinant MT1-MMP (Invitek, Berlin, Germany), MMP-2, and
MMP-9 from supernatants conditioned by HT-1080/MT1 cells. Gelatin
zymograms were obtained overnight in the absence or presence of marimastat
(0.01-1 M) in enzyme buffer and developed by Coomassie blue staining.
Collagenolysis generated by live cells while migrating through the
native 3D collagen substrate was quantied by a novel uorescein
isothiocyanate (FITC)release assay. Two percent FITC-collagen type I
NONPROTEOLYTIC T-LYMPHOCYTE MIGRATION 3263BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
monomers from bovine skin (Molecular Probes) were copolymerized
with rat-tail collagen (Becton Dickinson, Heidelberg, Germany; nal
concentration 1.65 mg/mL)
18
and cells under phenol redfree conditions
(3 10
5
cells/l00 L medium/ well). After 40 hours culture in the absence
or presence of protease inhibitor cocktail, solid-phase collagen including
the cells was pelleted (15 000 g, 10 minutes, 4°C), and the supernatant
was analyzed for FITC contents by spectrouorometry. 100% values
represent total FITC recovery after collagenase digestion of cell-free
collagen lattices. Background uorescence was obtained for supernatants
from pelleted nondigested cell-free lattices. In contrast to lymphocytes and
monocytic cells, DCs generated high levels of unspecic FITC release that
was not sensitive to protease inhibitors nor associated with the structural
remodeling of collagen bers. Compared to cells isolated directly after
matrix polymerization, no increased cell-bound or endocytic FITC signal
was observed after 24 hours culture in FITC-collagen lattices in untreated
or protease inhibitortreated cells using ow cytometry.
FITC-collagen monomers were degraded by recombinant MT1-MMP,
MMP-2, as well as trypsin as shown by SDS-PAGE and silver staining (C.
Overall and E. Tam, unpublished, May 2002). Whereas FITC release
detected classical collagenase, gelatinase, and trypsinlike activity, it was
most sensitive to MMP-inhibitor BB-2516 (reduction by 80% in HT-1080
cells
18
), indicating MMPs as primary yet not sole enzyme family in
degrading the brillar collagen migration substrate. In contrast to detection
of collagen fragments by SDS-PAGE and silver staining, FITC-collagen
release was a more sensitive approach to detect collagen degradation in live
cell samples (K.W., unpublished observations,April 2003).
Focalized cell-associated collagen degradation in situ was visualized by
copolymerizing 5% quenched (q)FITC-collagen type I monomers from
bovine skin (Molecular Probes) with nonlabeled collagen. As assessed by
confocal microscopy, the uorescence intensity of qFITC in polymerized
collagen bers strongly increased upon focalized proteolysis generated by
proteolytic HT-1080 cells, which colocalized with the appearance of
collagen cleavage-site specic neo-epitope mAb COL2 3/4 C (K.W. and
P.F., unpublished,April 2003).
Confocal laser-scanning microscopy
3D confocal backscatter microscopy of xed samples was carried out on a
Leica TCS 4D system (Leica, Bensheim, Germany).
37
Dynamic sequences
from viable samples were obtained on the SP2 system (Leica) using a
temperature-controlled stage (37°C). Cells within the collagen lattice were
labeled with 1 M calcein-AM (Molecular Probes) and scanned at
20-second time intervals as 3D stacks of 4 to 8 z-sections. For dynamic
reconstruction, uorescence, reection and transmission signal were col-
lected simultaneously. Movies were obtained from 3-dimensionally recon-
structed image stacks over time. For digital image analysis, the National
Institutes of Health image program (software version 1.62) was used.
Results
In 3D collagen matrices, ConA-activated T cells spontaneously
developed an amoeboid migration type characterized by exible
elliptoid morphology. The rapid and frequent shape change and
oscillatory stop-and-go patterns resulted in a nonlinear path
structure (Figure 1). We have investigated which matrix-degrading
enzymes are expressed by T cells and how these proteases
contribute to migration and associated remodeling of the collagen
ber network.
Protease mRNAand surface expression
Primary human CD4
ConA-T blasts and, for conrmation, SupT1
lymphoma cells
26
were investigated for mRNA expression from
proteases of different classes, including MMPs, serine proteases,
and cathepsins (Figure 2 and Supplemental Table S1). Activated
CD4
T cells expressed MMP-9, MT1-MMP, MT4-MMP,
ADAM-9, -15, and -17, cathepsin L, urokinase-type plasminogen
activator (uPA) and its receptor uPAR as well as endogenous
protease inhibitors TIMP-2 and PAI-1 (Figure 2A). SupT1 cells
expressed MT1-MMP, ADAM-9, -10, -11, -17, uPA, and TIMP-2
(Figure 2A). MT1-MMP and cathepsin L were the only collag-
enases expressed by either cell type. ConA-stimulated CD4
T
blasts showed minor MT1-MMP surface expression (Figure 2B),
while MT1-MMP surface levels were identical to the isotypic
antibody in resting cells (not shown). As positive control, signi-
cant MT1-MMP surface staining was detected on HT-1080/MT1
cells (Figure 2B). However, even at low levels of surface protease
expression, T cells might be able to focalize protease activity to
Figure 1. Spontaneous locomotion of an activated CD4
T cell within 3D
collagen lattice. Changes in morphology and oscillatory path development. CD4
ConA blast migrating in a 3D collagen matrix in the absence of a chemotactic
gradient. The average velocity was 6 m/min. Time is indicated in hh:mm:ss (11
minutes total).
Figure 2. mRNA and cell surface expression of proteases and endogenous
protease inhibitors in CD4
T cells and SupT1 cells. (A) Total RNA from CD4
T
cells stimulated with ConA for 3 days and SupT1 lymphoma cells was subjected to
RT-PCR (30 cycles). Fragment lengths conformed to the expected size. No mRNA
was detected for MMP-8, -13, cathepsin B and K in either cell type (not shown).
Positive controls were performed from HT-1080/MT1 or HT-1080/neo cells (MMP-7
and -9). RTindicatesreverse transcriptase reaction. (B) FlowcytometryofMT1-MMP
surface expression (gray histogram) in ConA-T blasts and positive control cells
(HT-1080/MT1) as compared with isotypecontrol (white histogram).
3264 WOLF et al BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
substrate contacts and thereby generate local collagenolysis for
tissue penetration.
Lack of in situ collagenolysis
Initial dose-response studies on broad-spectrum MMP-inhibitor
BB-2516 (marimastat) showed complete inhibition of gelatin
degradation by MMP-2, MMP-9, and MT1-MMP at low M
concentration (Figure 3A). However, the topographic arrangement
and focalization of proteolysis executed by live cells within the
collagen substrate are not appropriately detected by zymography.
Therefore, in situ collagen degradation caused by T cells in the
process of migration was quantied as the FITC release from
FITC-labeled brillar collagen lattices. After 40 hours of culture,
ConA-stimulated CD4
T blasts did not cause FITC release into
the supernatant above background levels. In contrast, collageno-
lytic HT-1080/MT1 brosarcoma cells released high levels of
soluble FITC into the supernatant (Figure 3B). For inhibition of
proteases, a broad-spectrum inhibitor cocktail was used to simulta-
neously target MMPs, MT-MMPs,ADAMs, cathepsins, serine, and
cysteine proteases.
18
Near-total abrogation of collagenolysis by
inhibitor cocktail was conrmed for HT-1080/MT1 cells as positive
control, while no effect was obtained for CD4
cells (Figure 3B).
The lack of FITC release was not caused by a decrease in T-cell
viability, as detected by ow cytometry after cell isolation by
collagenase digestion after 40 hours (Figure 3C).
To exclude minor local collagen ber cleavage exerted by T
cells that might escape detection by FITC release, collagen lattices
containing quenched FITC-labeled collagen monomers were used
for confocal uorescence and backscatter microscopy. At physical
contacts of polarized CD4
blasts to collagen bers containing
quenched FITC, no focal in situ uorescence above background
uorescence was detected (Figure 3D, arrowheads), while signi-
cant focal uorescence was developed by HT-1080/MT1 cells at
interactions to these bers (Figure 3E, arrowheads). Thus, MT1-
MMP and other proteases, although expressed by CD4
, did not
degrade the collagenous migration substrate.
Persisting migration in the presence of protease inhibitors
The contribution of enzymatic protease activity to T-cell migration
in 3D collagen lattices was investigated in the absence or presence
of protease inhibitor cocktail (Figure 4). Addition of inhibitor
cocktail neither changed the amoeboid morphodynamics of CD4
T cells (Supplemental Video 1) nor the oscillatory structure of the
cell paths (Figure 4A). Also, the steady-state migration velocity
was approximately 6 m/min for both nontreated and inhibitor
cocktail-exposed cells (Figure 4B, left). The number of migrating
cells including their stop-and-go-pattern remained unchanged
(Figure 4B, right). Likewise, T-cell migration within multicompo-
nent matrices containing collagen, hyaluronan, and the cross-linker
Figure 3. Lack of in situ collagen degradation by T blasts. (A)
Dose-dependent inhibition of MMP-2, MMP-9, and recombinant
MT1-MMP activity by BB-2615 (marimastat) in gelatin zymogra-
phy. (B) Lack of FITC release from FITC-labeled collagen matri-
ces by T cells, but not by HT-1080/MT1 cells (positive control).
ConA-stimulated CD4
T cells or HT-1080/MT1 cells were cul
-
tured in 3D collagen lattices containing 2% FITC-collagen in the
absence or presence of protease inhibitor cocktail for 40 hours.
Data show the means SDs for 3 independent experiments.
(C) The percentage of viable CD4
cells remained unaffected
after 40 hours of culture in collagen. (D) CD4
T cell and (E)
HT-1080/MT1 cell incorporated in a 3D collagen lattice containing
5% quenched FITC-collagen. The false-color encoded confocal
FITC channel (color code inset: fluorescence channel for lowest
() and highest () intensity, respectively) of the 3D recon-
structed in focus sections (left) was superimposedontheshapeof
the cell body (right; red false color)obtainedfromthetransmission
image. In CD4
T cell, physical contact with collagen fibers
(arrowheads) did not result in increased cleavage-related fluores-
cence (blue false color; arrowheads), whereas HT-1080/MT1 cell
(positive control) generated focal FITC-fluorescence at fiber
binding sites (yellow false color; arrowheads). Fiber cleavage by
HT-1080 cells in situ was confirmed bystaining with cleavage-site
specific antibody (not shown). Imagesare representativefor 10 to
15 cells. Bars, 5 m.Black arrows, directionof migration.
Figure 4. Persisting migration of CD4
blasts in the presence of protease
inhibitors. Migration in 3D collagen (A-B) and multicomponent matrices (C) in the
absence or presence of protease inhibitor cocktail. (A) Digitized paths of 40 cells at
orthotopic position (2-hour tracking period; 1 representative of 3 independent
experiments). (B-C) Steady-state velocity and percentage of migrating CD4
T cells
in collagen (B) and collagen copolymerized with hyaluronan and chondroitin sulfate
(C). Data show the means of time-dependent population parameters SDs for 3
independent experiments (120 cells).
NONPROTEOLYTIC T-LYMPHOCYTE MIGRATION 3265BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
chondroitin sulfate did not depend on the function of matrix-
degrading enzymes (Figure 4C). Thus, activated CD4
T cells do
not engage the enzymatic activity of expressed collagenases for
their migration through 3D brillar collagen matrices.
Biophysics of nonproteolytic amoeboid T-cell migration
Because T-cell migration was neither accompanied by proteolytic
collagen ber degradation or sensitive to protease inhibitor cock-
tail, T-cell trafcking through brillar collagen should occur
through a nonproteolytic mechanism. The biophysics of T-cell
migration within the collagen network were reconstructed at high
resolution by 4D confocal backscatter and uorescence microscopy
(Figure 5). In both, control and inhibitor cocktailtreated cultures,
migrating Tcells exhibitedperiods of alignment along matrix bers
(Figure 5A-B, black arrowheads; Supplemental Video 2). Propul-
sive squeezing through regions of narrow space prompted the
formation of a narrow zone of the cell body, termed constriction
ring
38
(Figure 5Av-vi; Bii; white arrowheads). Whereas forward
movement resulted from alignment of the cell body in parallel to
more linear ber strands (Figure 5C, black arrowheads), directional
changes were frequent at regions of narrow space, forcing the cell
to circummigrate rather than degrade the obstacle (Supplemental
Video 3). Such migratory turns were caused by cell deection
along adjacent collagen bers (Figure 5D, arrowheads) and re-
sulted in angle changes along the length axis between leading edge
and trailing uropod (Figure 5D, inset; cyan arrow).As was apparent
from the movies that any change in cell shape, including cell
extension and ber pushing, cell contraction and ber pulling, as
well as outward dislocation of constraining bers upon cell
squeezing, resulted in temporary deformation of the collagen
network structure (dislocation up to 2 m; movies 2-3). These
contact-dependent and temporary changes did not, however, gener-
ate long-lasting structural changes in matrix structure after the cell
had detached.
To analyze the dynamic nature of cellular interactions with
collagen bers quantitatively, the cell boundary of a migrating T
cell (depicted in Figure 1) was obtained as 2D outline every 24
seconds (Figure 6A, dark blue) and superimposed onto the 3D
reconstruction of the transmigrated collagen matrix (Figure 6A,
green). Colocalization of outline and collagen bers resulted in
bright pixels (Figure 6A, cyan; arrowheads) at different position
along the migration track (Figure 6A, red solid line). The position
of collagen bers that were touched upon migration shows that the
volume of the cell follows precisely the predened collagen
scaffold (Figure 6B, white ber zones). The location and frequency
of cell-ber guidance along the extracted cell outlines and their
regions in parallel contact with bers (Figure 6C, black segments)
were quantied along 4 virtual tracks placed into the cell shape
from step to step in order to represent the geometry of both
maximum cell width (Figure 6D, a and d) and more narrow regions
near the uropod (Figure 6D, b and c). Along the migration path,
each of these tracks showed alternating contacts with collagen
bers ranging from 1 to 10 m in length (Figure 6E, top).Although
each interaction segment was of short-lived nature, together they
generated a multidimensional physical scaffold for cell alignment
to at least one, frequently 2 or more, bers simultaneously (Figure
6E, bottom). By means of elongation and, presumably, by volume
(which was not considered in this analysis yet apparent from the
movies), polarized T cells simultaneously aligned at several
positions to different bers resulting in near-always guiding contact
by outside cues (see also movies 1-3). Because such contact
guidance determined both forward movement as well as migratory
turns (Figure 6C, asterisks), no bundling, structural degradation, or
remodeling of matrix bers were required for fast T-cell crawling
within collagenous scaffolds. These ndings contrast with the
migration-associated proteolytic degradation andremodeling of the
same collagen substrateby broblasts and tumor cells (Friedl et al,
8
Wolf et al,
18
Maaser et al,
35
and movies therein).
Nonproteolytic amoeboid migration in other leukocytes
Similar to T cells, a spectrum of matrix-degrading proteases is
expressed in other leukocytes, such as SupT1 cells
26
(Figure 2B),
DCs,
23
and monocytic cells.
24
Similar to T cells, however, only
background release of FITC was obtained from FITC-collagen for
these cells upon migration over an extended time period of 40
hours (Figure 7A), and no reduction of FITC release was achieved
by protease inhibitor cocktail (Figure 7A). The protease-
independence of migration in these cells was shown as an
undiminished velocity as well as number of moving cells in the
presence of protease inhibitor cocktail (Figure 7B; Supplemental
Video 4). Although the size and polarized morphology differ
between lymphocytes, monocytes, and DCs, each cell type fol-
lowed the principles of amoeboid movement, as indicated by
rapid shape change (movie 4), the formation of constriction rings
(Figure 7C and Supplemental Video 5), and concomitant cytoplas-
mic streaming through regions of narrow space (Supplemental
Video 5).
Figure 5.AmoeboidT-cell migration, physicalcell-berinteraction,andcontact
guidance within 3D collagen matrix in the absence or presence of protease
inhibitor cocktail. (A) Untreated and (B) protease inhibitor cocktailteated calcein-
stained T cells. Crawling, alignment along ber strands (black arrowheads) and
formation of constriction rings (white arrowheads). Image sequences (i-vi) represent
as a time series 8 (A) and 3 (B) minutes of observation time. (C) T-cell alignment in
parallel to matrix bers (white arrowheads) upon forward migration (black arrow).
(D) Change in migration direction. Angle turns along the cellular length axis (cyan
arrow, small inset) and physical connement of the cell body and uropod along
guiding collagen bers (white arrowheads), reecting the direction change. Bars,
5 m.
3266 WOLF et al BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
Discussion
In contrast to broblasts
39
and tumor cells,
9,18
the migration of T
cells and other leukocytes within 3D collagen matrices is indepen-
dent of proteolytic ECM remodeling and therefore insensitive to
protease inhibitor treatment. Our data provide a mechanism on how
amoeboid crawling supports nondestructive leukocyte trafcking
through interstitial tissues.
8
Lack of migration-associated collagenolysis inmigrating T cells
In this study, T lymphocytes were used as prototype cells for
amoeboid movement in collagen. Although several matrix-
degrading proteases are expressed by activated CD4
T cells and
SupT1 lymphoma cells, including MT1-MMP, MT4-MMP, cathep-
sin L,ADAM-9, and -15
26,40
and although cathepsin Land MT1-MMP
are potent collagenases toward native type I collagen,
41,42
no degrada
-
tion of the brillar collagen substrate was caused by them upon
migration. We used a combination of physical and biochemical
approaches to detect collagenolysis in situ and inhibitor efciency:
(1) gelatin zymography, (2) FITC-collagen release from FITC-
labeled collagen matrices, (3) focal in-situ uorescence at T-cell
interactions to collagen bers containing quenched FITC mol-
ecules, and (4) structural ber remodeling using high-resolution
confocal backscatter microscopy from live cell samples. Specic
FITC release from FITC-collagen brils was at low levels for
CD4
and CD8
blasts, SupT1 lymphoma cells, and U937
monocytic cells and largely insensitive to the multicomponent
protease inhibitor cocktail. These values were 20- to 100-fold
lower than seen in HT-1080/MT1 brosarcoma cells used as
positive controls.
18,21
As exception, DCs generated signicant
FITC release by mechanisms that were independent of the protease
classes targeted by our inhibitor cocktail, thereby currently preclud-
ing a meaningful use of the FITC release assay for DCs. Besides
collagenases, other enzymes generated by DCs, such as amino
hydrolases,
43
could dissolve the amid bond between FITC and
protein carrier without cleaving the collagenous backbone.
Migrating T cells neither bundled, extensively pulled, nor
reorganizedcollagen bers nor generated focalcleavageof quenched
FITC-collagen or proteolytic matrix defects, in contrast to collagen-
olytic HT-1080 cells.
18
The protease inhibitor cocktail reduced
collagenolysis by 95%-98% and abrogated ber remodeling and
proteolytic path generation in tumor cells.
18
In leukocytes, how
-
ever, the protease inhibitor cocktail did not alter any of the
migration parameters assessed by sensitive cell tracking, including
percentage of migrating cells, their velocity or stop-and-go-pattern,
nor their path proles and the underlying interaction dynamics to
collagen bers. These ndings suggest that leukocytes do not need
to structurally modify the brillar collagen network for their
migration, thereby extending previous indirect evidence on T cells
penetrating radioactive-labeled collagen matrices
4
and neutrophils
migrating within amnionic membrane.
44
We cannot completely
exclude minor focal cleavage of collagen by biochemical means.
However, collagen backscatter imaging shows that the physical
ber structure and position remain intact during and after T-cell
migration, whereas migrating tumor cells generate 3D matrix
defects using the same collagen substrate.
18,37
Together, these data
argue against a biophysically signicant change in matrix architec-
ture by leukocytesalongtheir migrationtracks and further strengthen
concepts on differences of molecular migration strategies between
leukocytes and stromal cells.
8,9
Figure 6. Reconstruction of contact guidance of migrating T cell by collagen
bers. (A) The outlineof the cell depictedin Figure 1was obtained every 24seconds
and superimposed on the 3D reconstructed backscatter signal of the collagen bers.
Cell boundary (blue), 3D collagen matrix (green; 10 m in depth), and colocalization
of cell boundary and individual collagen bers (cyan, arrowheads). The mean path
and pseudopodal extensions are shown by the solid red line. Migration occurred
towardthetop right corner (black arrow). (B) False color representation of thephysics
of the transmigrated path (white segments), calculated from pixel colocalization
between T-cell boundary and collagen bers. (C) Extracted cell boundary (gray line)
and segments colocalized with collagen bers (black sections). Asterisks indicate
turnsguidedbyoutsidebercues.(D)Denitionof4virtualtracksalongthemigration
path toapproximate 4major morphological regions of a polarized Tcell; for instance,
lateral portions of the anterior head (a, d) and posterior cell parts near the uropod (b,
c). (E) Calculation of pixel series that represent black segments in (C) for each track
along the migration path (E, top graph), resulting in the cumulative number of bers
that were simultaneously aligned along the forward moving cell edge (E, bottom
graph). These reconstruction data are representative for more than 5 independent
cells reconstructed by dynamic confocal backscatter imaging (compare movies 1-3).
Bars, 5 m.
NONPROTEOLYTIC T-LYMPHOCYTE MIGRATION 3267BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
Amoeboid shape change: a nonproteolytic, biophysical
migration mechanism bypassing matrix barriers
Previous dynamicreconstruction on nonlabeled neutrophils,T cells,and
dendritic cells show leukocyte movement in collagen or amnionic
membrane as a contact guidance-dependent process that appears to
follow pre-existing matrix scaffolds.
4,6,44-46
We haveused improved
dynamic real-time reconstruction of uorescently labeled cells
together with high-resolution imaging of the 3D matrix architec-
ture. The typical random paths and turns developed by T cells in 3D
collagen lattices result from stringent alignment of the cell body in
parallel to brils and ber bundles that border random gaps and
trails pre-existing in the lattice. While following brillar guidance
tracks, the volume of the cell body reects the preformed space of
trails of least resistance within the matrix. These reconstruction
data support previous work that suggests physical contact guidance
mechanisms to direct leukocyte migration in scaffolds indepen-
dently of proteolytic ECM breakdown.
6,44,45
In regions of narrow
space, depending on the pore size, 2 different mechanisms sustain
nonproteolytic movement. First, squeezing through narrow gaps by
adapting the cell shape and the formation of a constriction ring,
38
followed by cytoplasmic streaming through this immobile external
connement zone. Alternatively, if matrix barriers are too dense
relative to the deformation ability of the cell (below 1-2 min
diameter; compare movie 3), a change in migration direction provide
movement around the obstacle. Hence, nonpenetrable regions are
circummigrated at near-undiminished velocity.
These hallmarks of amoeboid biomechanics are clearly retained
in migrating T cells, lymphoma cells, and monocytes. In DCs, the
ubiquitous dendrites and an even more striking morphologic
exibility allow for sometimes grotesque deformation of the cell
body (movie 4). Although the principal elliptoid amoeboid shape
appears concealed in DCs, other amoeboid characteristics, such as
shape change in response to outside cues, cell constriction and
movement independent of proteases and 1 integrins remain
retained throughout migration.
46
Together, the data suggest that,
similar to neutrophils,
2
amoeboid features are retained in migrating
T cells, monocytes, and DCs, likewise.
Nonproteolytic leukocyte migration in vivo
Whereas direct in vivo measurements on protease function in
leukocyte migration are currently lacking, circumstantial evidence
suggests the contribution of protease-independent processes to
T-cell trafcking through the body. First, no obvious changes in
peripheral T-cell counts and redistribution are apparent in vivo during
MMP and other protease inhibitorbased therapy.
31,32,47
Second, resting
T cells are capable of entering lymphatic tissues through basement
membranes underlying blood vessels and recirculating, although they
produce little or no proteases. Third, resting as well as activated T cells
generate high migration velocities up to 30 m/min in vivo.
48
As in
3D collagen, the resulting contact times toward individual collagen
bers in vivo are in the range of only 5 to 60 seconds (similar to in
vitro kinetics; compare Figure 6E), thereby greatly exceeding
interaction dynamics achieved by stromal cells that execute
pericellular proteolysis (contact time in the range of minutes to
hours).
8
Lastly, a lack of matrix degradation by migrating T cells is
obvious from in vivo imaging in the intact lymph node.
48
Besides leukocyte shape change and contact guidance, addi-
tional (patho)physiologic mechanisms are likely to support nonpro-
teolytic lymphocyte trafcking through 3D tissues. In vivo, regions
of loose brillar collagen networks support oedematous swelling
and reversible widening of matrix gaps in response to vasodilata-
tion and inammation. Local edema is likely to generate biome-
chanically suitable pathways for rapid leukocyte trafcking inde-
pendent of proteolytic matrix degradation. In the dermis, such
loose connective tissue zones, which are closely mimicked by
collagen matrices, include perivascular spaces located along blood
vessels, adjacent to basement membranes, and in the dermal
papillae.
10,11,18
These trafcking highwaysare preferentially used
by passenger leukocytes upon acute and chronic inammation,
such as in eczema.
10
Although the basic migration program in T
cells and other leukocytes does not require proteases for removing
matrix barriers, the next steps leading toward inammatory connec-
tive tissue turnover and destruction remain to be investigated.
These likely require additional microenvironmentalchanges,includ-
ing the up-regulation and function of proteases in bystanding
broblasts, endothelial cells, and/or other leukocytes.
Implications
Amoeboid shape change and guidance along pre-existing matrix
structures provide supramolecular strategies for protease-indepen-
dent movement through different brillar tissue matrices. Because
rapidly moving leukocytes and, in particular, T cells establish only
short-lived substrate contacts coupled to diffusely distributed
integrins and actin cytoskeleton, it is reasonable to assume that
their particular membrane architecture may not support sufcient
receptor focalization required for contact-mediated proteolysis
seen in slower moving cell types, such as broblasts and tumor
Figure 7. Protease-independent migrationof other leukocytes. (A) ReleaseofFITC from FITC-containing 3Dcollagenmatrixby migrating SupT1 lymphomacells,CD8
T
blasts, U937 monocytic cells, and monocyte-derived human dendritic cells. Cells were cultured in FITC-collagen for 40 hours in the absence or presence of protease inhibitor
cocktail. Data represent the means SD from 3 independent experiments. High nonspecic uorescence of unclear origin was obtained for supernatants from dendritic cells
only. This uorescence was neither sensitive to protease inhibitors nor did it reect ber degradation, as previously shown by confocal analysis.
46
(B) Mean velocity and
percentage of locomotingcells in theabsence or presenceof inhibitor cocktail. Data show the mean activities over time (1hour) and standarddeviations from 3independent
experiments (120 cells) obtained from cell tracking. (C) Amoeboid movement of U937 cell resulting in the formation of a constriction ring (arrow) that remains unchanged in
position. Bar,10 m.
3268 WOLF et al BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
cells.
18
Amoeboid leukocyte movement, hence, represents a special
-
ized migration type that is resistant to pharmacotherapeutic target-
ing of MMPs and other proteases, thereby explaining why clinical
trials have not been complicated by leukocyte recirculation and
immune defects. These characteristics may provide a mechanism
for persistent tissue integrity despite abundant constitutive lympho-
cyte inux and recirculation.
Acknowledgments
We acknowledge Margit Ott for invaluable technical assistance,
Bong-Seok Song for assistance with confocal microscopy, and Jo¨rg
Geiger for expert help in uorometric analysis. Eric Tam and Chris
Overall are acknowledged for collagen cleavage analysis.
References
1. von Andrian UH, Mackay CR. T-cell function and
migration: two sides of the same coin. N Engl
J Med. 2000;343:1020-1034.
2. Devreotes PN, Zigmond SH. Chemotaxis in eu-
karyotic cells: a focus on leukocytes and Dictyo-
stelium.Annu Rev Cell Biol. 1988;4:649-686.
3. Friedl P, Borgmann S, Brocker EB. Leukocyte
crawling through extracellular matrix and the Dic-
tyostelium paradigm of movementlessons from
a socialamoeba. JLeukoc Biol.2001;70:491-509.
4. Schor SL,Allen TD,Winn B.Lymphocyte migration
into three-dimensionalcollagen matrices:a quantita-
tive study. JCell Biol.1983;96:1089-1096.
5. Entschladen F, Niggemann B, Za¨nker KS, Friedl
P. Differential requirement of protein tyrosine ki-
nases and protein kinase C in the regulation of T
cell locomotion in three-dimensional collagen ma-
trices. J Immunol. 1997;159:3203-3210.
6. Friedl P, Entschladen F, Conrad C, Niggemann B,
Zanker KS. CD4
T lymphocytes migrating in
three-dimensional collagen lattices lack focal ad-
hesions and utilize beta1 integrin-independent
strategies for polarization, interaction with colla-
gen bers and locomotion. Eur J Immunol. 1998;
28:2331-2343.
7. Brakebusch C, Fillatreau S, PotocnikAJ, et al.
Beta1 integrin is not essential for hematopoiesis
but is necessary for the T celldependent IgM
antibody response. Immunity. 2002;16:465-477.
8. Friedl P, Zanker KS, Bro¨cker EB. Cell migration
strategies in 3-D extracellular matrix: differences
in morphology, cell matrix interactions, and inte-
grin function. Microsc ResTech. 1998;43:369-378.
9. Friedl P, Wolf K. Tumour cell invasion and migra-
tion: diversity and escape mechanisms. Nature
Rev Cancer. 2003;3:362-374.
10. FriedlP, Brocker EB.T cell migrationin three-dimen-
sional extracellularmatrix: guidanceby polarityand
sensations. DevImmunol. 2000;7:249-266.
11. Stoitzner P, Pfaller K, Stossel H, Romani N.A
close-up view of migrating Langerhans cells in
the skin. J Invest Dermatol. 2002;118:117-125.
12. Nakahara H, Howard L, Thompson EW, et al.
Transmembrane/cytoplasmic domain-mediated
membrane type 1-matrix metalloprotease docking
to invadopodia is required for cell invasion. Proc
NatlAcad Sci U SA. 1997;94:7959-7964.
13. dOrtho MP, Stanton H, Butler M,Atkinson SJ,
Murphy G, Hembry RM. MT1-MMP on the cell
surface causes focal degradation of gelatin lms.
FEBS Lett. 1998;421:159-164.
14. Campbell EJ, Campbell MA, Owen CA. Bioactive
proteinase 3 on the cell surface of human neutro-
phils: quantication, catalyticactivity, and susceptibil-
ity toinhibition. JImmunol. 2000;165:3366-3374.
15. Kindzelskii AL, Zhou MJ, Haugland RP, Boxer LA,
Petty HR. Oscillatory pericellular proteolysis and
oxidant deposition during neutrophil locomotion.
Biophys J. 1998;74:90-97.
16. Murphy G, Gavrilovic J. Proteolysis and cell mi-
gration: creating a path? Curr Opin Cell Biol.
1999;11:614-621.
17. Owen CA, CampbellEJ.Thecell biologyof leukocyte-
mediated proteolysis. JLeukocBiol. 1999;65:137-
150.
18. Wolf K, Mazo I, LeungH, Engelke K,et al.Compen-
sation mechanismin tumorcell migration:mesen-
chymal-amoeboid transitionafter blockingof pericel-
lular proteolysis.J CellBiol. 2003;160:267-277.
19. Belkin AM, Akimov SS, Zaritskaya LS, Ratnikov
BI, Deryugina EI, StronginAY. Matrix-dependent
proteolysis of surface transglutaminase by mem-
brane-type metalloproteinase regulates cancer
cell adhesion and locomotion. J Biol Chem. 2001;
276:18415-18422.
20. Stamenkovic I. Matrix metalloproteinases in tu-
mor invasion and metastasis. Semin Cancer Biol.
2000;10:415-433.
21. Deryugina EI, Bourdon MA, Reisfeld RA, Strongin
A. Remodeling of collagen matrix by human tu-
mor cells requires activation and cell surface as-
sociation of matrix metalloproteinase-2. Cancer
Res. 1998;58:3743-3750.
22. Wei Y, Lukashev M, Simon DI, et al. Regulation of
integrin function by the urokinase receptor. Sci-
ence. 1996;273:1551-1555.
23. Ratzinger G, Stoitzner P, Ebner S, et al. Matrix
metalloproteinases 9 and 2 are necessary for the
migration of Langerhans cells and dermal den-
dritic cells from human and murine skin. J Immu-
nol. 2002;168:4361-4371.
24. Machein U, Conca W. Expression of several ma-
trix metalloproteinase genes in human monocytic
cells.Adv Exp Med Biol. 1997;421:247-251.
25. Bristow CL, Lyford LK, Stevens DP, Flood PM.
Elastase is a constituent product of T cells. Bio-
chem Biophys Res Commun. 1991;181:232-239.
26. Leppert D, Hauser SL, Kishiyama JL,An S, Zeng
L, Goetzl EJ. Stimulation of matrix-metallopro-
teinase-dependent migration of Tcells by eico-
sanoids. FASEB J. 1995;1473-1481.
27. Gundersen D, Tran-Thang C, Sordat B, Mourali F,
Ruegg C. Plasmin-induced proteolysis of tenas-
cin-C: modulation by Tlymphocyte-derived uroki-
nase-type plasminogen activator and effect on T
lymphocyte adhesion, activation, and cell cluster-
ing. J Immunol. 1997;158:1051-1060.
28. Leppert D, Waubant E, Galardy R, Bunnett NW,
Hauser SL.T cell gelatinases mediate basement
membrane transmigration in vitro. J Immunol.
1995;154:4379-4389.
29. Ikegami M, Umehara F, Ikegami N, Maekawa R,
Osame M. Selective matrix metalloproteinase
inhibitor, N-biphenyl sulfonyl phenylalanine hy-
droxamic acid, inhibits the migration of CD4
T
lymphocytes in patients with HTLV-I-associated
myelopathy. J Neuroimmunol. 2002;127:134-138.
30. Allport JR, LimYC, Shipley JM, et al. Neutrophils
from MMP-9- or neutrophil elastase-decient
mice show no defect in transendothelial migration
under ow in vitro.J LeukocBiol. 2002;71:821-828.
31. Betsuyaku T, Shipley JM, Liu Z, Senior RM. Neu-
trophil emigration in the lungs, peritoneum, and
skin does not require gelatinase B.Am J Respir
Cell Mol Biol. 1999;20:1303-1309.
32. Mackarel AJ, Cottell DC, Russell KJ, FitzGerald
MX, OConnor CM. Migration of neutrophils
across human pulmonary endothelial cells is not
blocked by matrix metalloproteinase or serine
protease inhibitors.Am J Respir Cell Mol Biol.
1999;20:1209-1219.
33. Gunzer M, Scha¨fer A, Borgmann S, et al.Antigen
presentation in three-dimensional extracellular
matrix: interactions of Tcells with dendritic cells
are dynamic, short lived, and sequential. Immu-
nity. 2000;13:323-332.
34. Friedl P, Noble PB, Za¨nker KS.T Lymphocyte
locomotion in a three-dimensional collagen ma-
trix: expression and function of cell adhesion mol-
ecules. J Immunol. 1995;154:4973-4985.
35. Maaser K, Wolf K, Klein CE, et al. Functional hier-
archy of simultaneously expressed adhesion re-
ceptors: integrin alpha2beta1 but not CD44 medi-
ates MV3 melanoma cell migration and matrix
reorganization within three-dimensional hyaluro-
nan-containing collagen matrices. Mol Biol Cell
1999;10:3067-3079.
36. Roghani M, Becherer JD, Moss ML, et al. Metal-
loprotease-disintegrin MDC9: intracellular matu-
ration and catalytic activity. J Biol Chem. 1999;
274:3531-3540.
37. Friedl P, Maaser K, Klein CE, Niggemann B,
Krohne G, Zanker KS. Migration of highly aggres-
sive MV3 melanoma cells in 3-dimensional colla-
gen lattices results in local matrix reorganization
and shedding of alpha2 and beta1 integrins and
CD44. Cancer Res. 1997;57:2061-2070.
38. Lewis WH. On the locomotion of the polymorpho-
nuclear neutrophiles of the rat in autoplasma cul-
tures. Bull Johns Hopkins Hosp. 1934;4,273-279.
39. Langholz O, Rockel D, Mauch C, et al. Collagen
and collagenase gene expression in three-dimen-
sional collagen lattices are differentially regulated
by alpha 1 beta 1 and alpha 2 beta 1 integrins.
J Cell Biol. 1995;131:1903-1915.
40. Esparza J, Vilardell C, Calvo J, et al. Fibronectin
upregulates gelatinase B (MMP-9) and induces
coordinated expression of gelatinaseA(MMP-2)
and its activator MT1-MMP (MMP-14) by human
T lymphocyte cell lines: a process repressed
through RAS/MAPkinase signaling pathways.
Blood. 1999;94:2754-2766.
41. Kakegawa H, NikawaT, Tagami K, et al. Partici-
pation of cathepsin Lon bone resorption. FEBS
Lett. 1993;321:247-250.
42. Ohuchi E, Imai K,Fujii Y, Sato H, SeikiM, OkadaY.
Membrane type1 matrixmetalloproteinase digests
interstitial collagensand otherextracellular matrix
macromolecules. JBiol Chem.1997;272:2446-2451.
43. Matias I, Pochard P, Orlando P, Salzet M, Pestel
J, Di Marzo V. Presence and regulation of the en-
docannabinoid system in human dendritic cells.
Eur J Biochem. 2002;269:3771-3778.
44. Mandeville JT, Lawson MA, Maxeld FR. Dy-
namic imaging of neutrophil migration in three
dimensions: mechanical interactions between
cells and matrix. J Leukoc Biol. 1997;61:188-200.
45. Wilkinson PC, Lackie JM. The inuence of con-
tact guidance on chemotaxis of human neutrophil
leukocytes. Exp Cell Res. 1983;145:255-264.
46. Gunzer M, Ka¨mpgen E, Bro¨cker E-B, Za¨nker KS,
Friedl P. Migration of dendritic cells in 3D-colla-
gen lattices: visualisation of dynamic interactions
with the substratum and the distribution of sur-
face structures via anovel confocalreectionimag-
ing technique.AdvExp MedBiol. 1997;417:97-103.
47. Coussens LM, Fingleton B, Matrisian LM. Matrix
metalloproteinase inhibitors and cancer: trials
and tribulations. Science. 2002;295:2387-2392.
48. Miller MJ, Wei SH, ParkerI, CahalanMD.Two-photon
imaging of lymphocytemotilityand antigenresponse
in intact lymphnode.Science. 2002;296:1869-1873.
NONPROTEOLYTIC T-LYMPHOCYTE MIGRATION 3269BLOOD, 1 NOVEMBER 2003
VOLUME 102, NUMBER 9
For personal use only. by guest on May 30, 2013. bloodjournal.hematologylibrary.orgFrom
... Interestingly, we noticed an abrupt decrease in mtROS amount between these two doses of oligomycin, correlating with the reduction in 3D motility (Fig. 4C) and suggesting that also mtROS might positively sustain 3D motility. ROS have pleiotropic roles in the regulation of cell migration [33][34][35][36] , particularly the amoeboid-like motility typical of T cells within 3D environment 37,38 . To better clarify the role of ATP and mtROS in CD8 + T cell 3D migration, we performed additional experiments. ...
... Interestingly, these cells also showed an improved 3D motility in collagen gel(Fig. 5Dand SupplementaryMovies 36,37), an effect dependent on the increased mitochondrial activity, since both oligomycin (Fig. 5Eand Supplementary Movies 38-41) and hypoxia (Supplementary ...
Article
Full-text available
The ability of CD8⁺ T cells to infiltrate solid tumors and reach cancer cells is associated with improved patient survival and responses to immunotherapy. Thus, identifying the factors controlling T cell migration in tumors is critical, so that strategies to intervene on these targets can be developed. Although interstitial motility is a highly energy-demanding process, the metabolic requirements of CD8⁺ T cells migrating in a 3D environment remain unclear. Here, we demonstrate that the tricarboxylic acid (TCA) cycle is the main metabolic pathway sustaining human CD8⁺ T cell motility in 3D collagen gels and tumor slices while glycolysis plays a more minor role. Using pharmacological and genetic approaches, we report that CD8⁺ T cell migration depends on the mitochondrial oxidation of glucose and glutamine, but not fatty acids, and both ATP and ROS produced by mitochondria are required for T cells to migrate. Pharmacological interventions to increase mitochondrial activity improve CD8⁺ T cell intratumoral migration and CAR T cell recruitment into tumor islets leading to better control of tumor growth in human xenograft models. Our study highlights the rationale of targeting mitochondrial metabolism to enhance the migration and antitumor efficacy of CAR T cells in treating solid tumors.
... However, it is unclear how three-dimensional (3D) cell shape dynamics drives 3D cell locomotion, facilitating an effective cell navigation through the complex, sterically tortuous environments. For example, it is not completely understood why T cells do not require specific biochemical or structural modifications to the surrounding collagen matrix for amoeboid motility and how they rely on the active changing of cell shape and steric (i.e., nonadhesive) interactions with a collagen matrix to circumnavigate, i.e., steer around the collagen fibers, while finding available lumens for passage (2,3). Moreover, it is also not clear why the observed efficiency of amoeboid motility is proportional to the contractility of cortical actomyosin and inversely proportional to the cell-microenvironment adhesive interactions across all sterically interactive environments, such as nanotopographic surfaces, 3D collagen matrices, and cancer tumor tissue samples (4)(5)(6). ...
... Functionally, the nonstretchable cortical rings may serve to resist further stretching forces provided by myosin-rich contractile spheroid cell segments, e.g., by limiting "unsafe" levels of cortex-expanding hydrostatic pressure against surrounding steric obstacles. Thus, while mesenchymal cells use anchored myosin-rich stress fibers to enable the mechanosensing of the adhesive microenvironments, the nonadhesive amoeboid cells may use the anchorless myosindepleted cortical rings for safe circumnavigation, i.e., obstacle avoidance, during the nonadhesive motility in sterically complex environments (1,3,7,47,48). ...
Article
Full-text available
The all-terrain motility of lymphocytes in tissues and tissue-like gels is best described as amoeboid motility. For amoeboid motility, lymphocytes do not require specific biochemical or structural modifications to the surrounding extracellular matrix. Instead, they rely on changing shape and steric interactions with the microenvironment. However, the exact mechanism of amoeboid motility remains elusive. Here, we report that septins participate in amoeboid motility of T cells, enabling the formation of F-actin and α-actinin–rich cortical rings at the sites of cell cortex–indenting collisions with the extracellular matrix. Cortical rings compartmentalize cells into chains of spherical segments that are spatially conformed to the available lumens, forming transient “hourglass”-shaped steric locks onto the surrounding collagen fibers. The steric lock facilitates pressure-driven peristaltic propulsion of cytosolic content by individually contracting cell segments. Our results suggest that septins provide microenvironment-guided partitioning of actomyosin contractility and steric pivots required for amoeboid motility of T cells in tissue-like microenvironments.
... Additionally, it serves as the initial line of defence against aggressors like infections, chemicals, and physical contact. Due to the function of the epidermis, which blocks the entry or exit of water-or water-soluble substances, and the hypodermis, which blocks heat loss due to the poor thermal conductivity of fat, it also has the capacity to prevent water loss and preserve temperature [354][355][356][357]. ...
Article
Full-text available
Background The study of plant-based medications, or phytomedicine, involves a wide spectrum of biological activi- ties. Due to the existence of secondary metabolites, herbal medicine has been used and practiced throughout history for the treatment of both acute and chronic conditions. Over the past century or so, numerous novel compounds with medicinal potential have been derived from plants. In the age of growing super infections and the emergence of resistant strains, natural medicines are inspiring optimism. Main body of the abstract The review discusses the role of herbal medicine as antibacterial agents and their use in wound care and management of wounds and the critical role of secondary metabolites of herbal plants in fight- ing bacterial infections. Some medicinal plants such as St. John’s wort (SJW) (Hypericum perforatum), Rosemary (Rosmarinus officinalis), Ginger (Zingiber officinale), and nopal cactus (Opuntia ficusindica (L.)) also possess wide range of biological activities and can give a synergistic effect if combined with antibiotics. In addition, natural biopolymers play an important role in the management of wounds as well as the physiological processes of the skin (hemostasis, inflammation, proliferation, and remodelling). Method A narrative review of papers relevant to the use of phytomedicine in treating infections was conducted by using electronic databases PubMed, CrossREF, and Google Scholar. Short conclusion Phytomedicine is one of the top options for the treatment of chronic illnesses for millions of peo- ple around the world. To learn about the bioactive components of medicinal plants, their medical benefits, and their synergistic or additive effects to enhance the action of medications, substantial new studies are still needed.
... The migration of T-cells is pivotal for scanning antigens presented by antigen-presenting cells and engaging in interactions with other cells crucial to the immune response. Thus, T-cell migration, characterized by nonproteolytic amoeboid movement, involves adaptive morphology, transient contact with surrogate ECM components, and navigation through matrix pores and plays an essential role in Tcell responses [52]. Increased density in breast cancer samples has been demonstrated to impede T-cell infiltration [11]. ...
Preprint
Full-text available
T-cells navigate through various mechanical environments within the body, adapting their behavior in response to these cues. An altered extracellular matrix (ECM) characterized by increased density and enhanced fibril alignment, as observed in cancer tissues, can significantly impact essential T-cell functions critical for immune responses. In this study, we used 3D collagen matrices with controlled density and fibril alignment to investigate T-cell migration, activation, and proliferation. Our results revealed that dense and aligned collagen matrices suppress T-cell activation through enhanced YAP signaling. By inhibiting YAP signaling, we demonstrated that T-cell activation within these challenging microenvironments improved, suggesting potential strategies to enhance the efficacy of immunotherapy by modulating T-cell responses in dense and aligned ECMs. Overall, our study deepens our understanding of T-cell mechanobiology within 3D relevant cellular microenvironments and provides insights into countering ECM-induced T-cell immunosuppression in diseases such as cancer.
... While no significant difference in the orientation of fibers around cancer cell clusters in the two coculture systems was observed (Fig. S10), the density of collagen fibers consistently decreased as a function of the distance towards cancer cell clusters ( Fig. S11 and Movie S3-S6). Therefore, we argue that as CTLs approached cancer cells, they encountered higher collagen density, leading a higher probability to change their orientation by following the fibers or penetrating through the fiber gaps, as suggested by others (26). ...
Preprint
One of the primary challenges in current cancer immunotherapy is the insufficient infiltration of cytotoxic T cells into solid tumors. Despite ongoing investigations, the mechanisms restricting T cell infiltration in immune-cold tumors remains elusive, hindered by the intricate tumor microenvironment. Here, we co-cultured mouse cancer cell lines with cancer-specific cytotoxic T cells to study the influence of cancer-T cell interactions on T cell motility, a crucial factor for effective tumor infiltration. By quantifying T cell motility patterns, we found that cancer-specific T cells exhibited extended contact time with cancer-cell clusters and higher directional persistence than non-specific T cells. Computational modelling suggested that T cells with stronger persistence could facilitate efficient searching for cancer clusters. Transcriptomic profiling revealed T cells recognizing cancer cells orchestrate accumulation on cancer cell clusters by activating adhesion proteins on both cancer cells and T cells, thereby fostering prolonged interaction on cancer cells. Furthermore, we observed that there were two distinct subpopulations of cancer cells after co-culturing with cancer-specific T cells: one expressing elevated levels of T-cell attractants and antigen-presentation molecules, while the other expressing immunosuppressive molecules and undergoing epithelial-to-mesenchymal transition. These dynamic insights into the complex interplay of cancer-T cell interactions and their impact on T cell motility hold implications for refining more efficacious cancer immunotherapy strategies.
... Many studies leverage these tools to decipher the migration mechanisms of immune cells. In the 3D context, CD8+ T cell amoeboid migration operates through a contractility driven mechanism via RhoA activation that is unique to the 2D environment [129][130][131]. Additionally, the T cell transfection efficiency can be modified on microfluidic systems through mechanoporation via stretching to engineer T cells with greater motility, antigen recognition, and antigen elimination [132,133]. ...
Article
Full-text available
GBM is the most aggressive and common form of primary brain cancer with a dismal prognosis. Current GBM treatments have not improved patient survival, due to the propensity for tumor cell adaptation and immune evasion, leading to a persistent progression of the disease. In recent years, the tumor microenvironment (TME) has been identified as a critical regulator of these pro-tumorigenic changes, providing a complex array of biomolecular and biophysical signals that facilitate evasion strategies by modulating tumor cells, stromal cells, and immune populations. Efforts to unravel these complex TME interactions are necessary to improve GBM therapy. Immunotherapy is a promising treatment strategy that utilizes a patient’s own immune system for tumor eradication and has exhibited exciting results in many cancer types; however, the highly immunosuppressive interactions between the immune cell populations and the GBM TME continue to present challenges. In order to elucidate these interactions, novel bioengineering models are being employed to decipher the mechanisms of immunologically “cold” GBMs. Additionally, these data are being leveraged to develop cell engineering strategies to bolster immunotherapy efficacy. This review presents an in-depth analysis of the biophysical interactions of the GBM TME and immune cell populations as well as the systems used to elucidate the underlying immunosuppressive mechanisms for improving current therapies.
... Activated T cells are known to be highly migratory on ICAM1 and Fibronectin (FN) coated surfaces, even in the absence of chemokines. Furthermore, we and others have shown that collagen I fiber structure can modulate T cell motility in 3D hydrogels [5,34]. To better delineate the underlying mechanism, we next examined activated human CD4þ and CD8þ T cells on the coated surfaces. ...
Article
The tumor extracellular matrix (ECM) is a barrier to anti-tumor immunity in solid tumors by disrupting T cell-tumor cell interaction underlying the need for elucidating mechanisms by which specific ECM proteins impact T cell motility and activity within the desmoplastic stroma of solid tumors. Here, we show that Collagen VI (Col VI) deposition correlates with stromal T cell density in human prostate cancer specimens. Furthermore, motility of CD4+ T cells is completely ablated on purified Col VI surfaces when compared with Fibronectin and Collagen I. Importantly, T cells adhered to Col VI surfaces displayed reduced cell spreading and fibrillar actin, indicating a reduction in traction force generation accompanied by a decrease in integrin β1 clustering. We found that CD4+ T cells largely lack expression of integrin α1 in the prostate tumor microenvironment and that blockade of α1β1 integrin heterodimers inhibited CD8+ T cell motility on prostate fibroblast-derived matrix, while re-expression of ITGA1 improved motility. Taken together, we show that the Col VI-rich microenvironment in prostate cancer reduces the motility of CD4+ T cells lacking integrin α1, leading to their accumulation in the stroma, thus putatively inhibiting anti-tumor T cell responses.
... Immune cells typically use an amoeboid migration mode with limited or no proteolytic activity and limited or no specific (e.g. integrin-mediated) adhesion to the extracellular matrix (ECM) [3][4][5] . Therefore, these cells are not expected to generate substantial traction forces, implying that they do not form mature focal adhesion contacts and thus are not able to significantly pull on and rearrange ECM fibers 4,6 . ...
Preprint
Full-text available
To reach targets outside the bloodstream, immune cells can extravasate and migrate through connective tissue. However, in contrast to migrating mesenchymal cells, the importance of matrix adhesion and traction force generation for immune cell migration is not well understood. We use time-lapse confocal reflection microscopy to obtain simultaneous measurements of migration velocity, directional persistence, and cell contractility. While we confirm that immune cells use a non-contractile amoeboid migration mode by default, we also find that NK92 cells as well as ex-vivo expanded NK cells exert substantial acto-myosin driven contractile forces on the extracellular matrix during short contractile phases reaching up to 100nN. Even non-activated primary B, T, NK cells, neutrophils, and monocytes exhibit this burst-like contractile behavior, and NK activation with cytokines increases both the magnitude and frequency of contractile bursts. Importantly, we show that cell speed and directional persistence of NK cells increase during and after these contractile phases, implying that the cells actively use traction forces to overcome steric hindrance and avoid getting stuck in narrow pores of the ECM. Accordingly, reducing cell adhesion to the ECM reduces the fraction of motile cells and their directional persistence, while the remaining motile cells mostly maintain their cell speed. We conclude that steric hindrance can induce a switch in the migration mode of immune cells, from a non-adhesive amoeboid migration mode to a highly contractile migration mode that closely resembles the gliding motion of motile mesenchymal cells.
Preprint
Full-text available
Many cell types, including transformed cells and immune cells, such as T cells, can produce protrusive, blister-like plasma membrane patches initially devoid of F-actin, called blebs. Despite recent progress in understanding the amoeboid-mesenchymal migration phenotype balance, it remains largely unknown how bleb-producing cells mechanically move through complex environments and what factors set their migration speed and directionality. Here, we have developed a hybrid stochastic-mean field biophysical model of bleb-based cell motility to study the potential for adhesion-free bleb-based migration. We find that simulated cells can only inefficiently migrate in the absence of adhesion-based forces, i.e., cell swimming, by producing high-to-low cortical contractility oscillations, where a high cortical contractility phase characterized by multiple bleb nucleation events is followed by an intracellular pressure buildup recovery phase at low cortical tensions, resulting in net cell motion. Our model suggests that bleb-producing cells can employ a hybrid bleb- and adhesion-based migration mechanism for optimum cell motility and identifies conditions for optimality. We find that blebs nucleate in subcellular regions of high cortical tension, low membrane-cortex linker density and high intracellular osmotic pressure. Lower extracellular matrix stiffnesses favor bleb growth, hence bleb-based cell swimming, which stands in contrast to classical mesenchymal/motor-clutch migration, where cell motility generally increases with increasing matrix stiffness. The developed model is expected to help generate design criteria for engineered immune therapies and provides a physical perspective of the potential migratory mechanisms underlying rapid single-cell migration, particularly in the context of bleb-based/amoeboid migration.
Article
Full-text available
Cell migration may depend on integrin-mediated adhesion to and deadhesion from extracellular matrix ligands. This concept, however, has not yet been confirmed for T lymphocytes migrating in three-dimensional extracellular matrices. We investigated receptor involvement in T cell migration combining a three-dimensional collagen matrix model with time-lapse videomicroscopy, computer-assisted cell tracking and confocal microscopy. In collagen lattices, the migration of CD4+ T cells (1) involved interactions with collagen fibers at the leading edge and uropod likewise, (2) occurred independently of the co-clustering of beta1, beta2, or beta3 integrins with F-actin, focal adhesion kinase, and phosphotyrosine at interactions with collagen fibers, (3) was counteracted by high-affinity beta1 integrin binding induced by antibody TS2/16; however, (4) the migration could not be blocked by a combination of adhesion-perturbing anti-beta1, -beta2, -beta3, and alpha v integrin antibodies. Integrin blocking neither affected cell polarization, interaction with fibers, beta1 integrin distribution, migration velocity, path structure, nor the number of locomoting cells in spontaneously migrating or concanavalin A-activated cells. Hence, T lymphocytes migrating in three-dimensional collagen matrices may utilize highly transient interactions with collagen fibers of low adhesivity, thereby differing from focal adhesion-dependent migration strategies employed by other cells.
Article
Full-text available
Membrane type 1 matrix metalloproteinase (MT1-MMP) is expressed on cancer cell membranes and activates the zymogen of MMP-2 (gelatinase A). We have recently isolated MT1-MMP complexed with tissue inhibitor of metalloproteinases 2 (TIMP-2) and demonstrated that MT1-MMP exhibits gelatinolytic activity by gelatin zymography (Imai, K., Ohuchi, E., Aoki, T., Nomura, H., Fujii, Y., Sato, H., Seiki, M., and Okada, Y. (1996) Cancer Res. 56, 2707-2710). In the present study, we have further purified to homogeneity a deletion mutant of MT1-MMP lacking the transmembrane domain (ΔMT1) and native MT1-MMP secreted from a human breast carcinoma cell line (MDA-MB-231 cells) and examined their substrate specificities. Both proteinases are active, without any treatment for activation, and digest type I (guinea pig), II (bovine), and III (human) collagens into characteristic 3/4 and 1/4 fragments. The cleavage sites of type I collagen are the Gly775-Ile776 bond for α1(I) chains and the Gly775-Leu776 and Gly781-Ile782 bonds for α2(I) chains. ΔMT1 hydrolyzes type I collagen 6.5- or 4-fold more preferentially than type II or III collagen, whereas MMP-1 (tissue collagenase) digests type III collagen more efficiently than the other two collagens. Quantitative analyses of the activity of ΔMT1 and MMP-1 indicate that ΔMT1 is 5-7.1-fold less efficient at cleaving type I collagen. On the other hand, gelatinolytic activity of ΔMT1 is 8-fold higher than that of MMP-1. ΔMT1 also digests cartilage proteoglycan, fibronectin, vitronectin and laminin-1 as well as α1-proteinase inhibitor and α2-macroglobulin. The activity of ΔMT1 on type I collagen is synergistically increased with co-incubation with MMP-2. These results indicate that MT1-MMP is an extracellular matrix-degrading enzyme sharing the substrate specificity with interstitial collagenases, and suggest that MT1-MMP plays a dual role in pathophysiological digestion of extracellular matrix through direct cleavage of the substrates and activation of proMMP-2.
Article
Full-text available
The invasion of human malignant melanoma cells into the extracellular matrix (ECM) involves the accumulation of proteases at sites of ECM degradation where activation of matrix metalloproteases (MMP) occurs. Here, we show that when membrane type 1 MMP (MT-MMP) was overexpressed in RPMI7951 human melanoma cells, the cells made contact with the ECM, activated soluble and ECM-bound MMP-2, and degraded and invaded the ECM. Further experiments demonstrated the importance of localization of the MT-MMP to invadopodia. Overexpression of MT-MMP without invadopodial localization caused activation of soluble MMP-2, but did not facilitate ECM degradation or cell invasiveness. Up-regulation of endogenous MT-MMP with concanavalin A caused activation of MMP-2. However, concanavalin A treatment prevented invadopodial localization of MT-MMP and ECM degradation. Neither a truncated MT-MMP mutant lacking transmembrane (TM) and cytoplasmic domains (Delta TMMT-MMP), nor a chimeric MT-MMP containing the interleukin 2 receptor alpha chain (IL-2R) TM and cytoplasmic domains (Delta TMMT-MMP/TMIL-2R) were localized to invadopodia or exhibited ECM degradation. Furthermore, a chimera of the TM/cytoplasmic domain of MT-MMP (TMMT-MMP) with tissue inhibitor of MMP 1 (TIMP-1/TMMT-MMP) directed the TIMP-1 molecule to invadopodia. Thus, the MT-MMP TM/cytoplasmic domain mediates the spatial organization of MT-MMP into invadopodia and subsequent degradation of the ECM.
Article
Cell movement within three-dimensional tissues is a cycling multistep process that requires the integration of complex biochemical and biophysical cell functions. Different cells solve this challenge differently, which leads to differences in migration strategies. Migration principles established for leukocytes share many characteristics with those described for ameba of the lower eukaryoteDictyostelium discoideum. The hallmarks of amoeboid movement include a simple polarized shape, dynamic pseudopod protrusion and retraction, flexible oscillatory shape changes, and rapid low-affinity crawling. Amoeboid crawling includes haptokinetic adhesion-dependent as well as biophysical migration mechanisms on or within many structurally and functionally different substrates. We describe central aspects of amoeboid movement in leukocytes and the implications for leukocyte crawling and positioning strategies within interstitial tissues.
Article
The reorganization of extracellular matrix (ECM) is an important function in many biological and pathophysiological processes. Culture of fibroblasts in a three-dimensional collagenous environment represents a suitable system to study the underlying mechanisms resulting from cell-ECM interaction, which leads to reprogramming of fibroblast biosynthetic capacity. The aim of this study was to identify receptors that transduce ECM signals into cellular events, resulting in reprogramming of connective tissue metabolism. Our data demonstrate that in human skin fibroblasts alpha 1 beta 1 and alpha 2 beta 1 integrins are the major receptors responsible for regulating ECM remodeling: alpha 1 beta 1 mediates the signals inducing downregulation of collagen gene expression, whereas the alpha 2 beta 1 integrin mediates induction of collagenase (MMP-1). Applying mAb directed against different integrin subunits resulted in triggering the heterodimeric receptors and enhancing the normal biochemical response to receptor ligation. Different signal transduction inhibitors were tested for their influence on gel contraction, expression of alpha 1(I) collagen and MMP-1 in fibroblasts within collagen gels. Ortho-vanadate and herbimycin A displayed no significant effect on any of these three processes. In contrast, genistein reduced lattice contraction, and completely inhibited induction of MMP-1, whereas type I collagen down-regulation was unaltered. Calphostin C inhibited only lattice contraction. Taken together, these data indicate a role of tyrosine-specific protein kinases in mediating gel contraction and induction of MMP-1, as well as an involvement of protein kinase C in the contraction process. The data presented here indicate that different signaling pathways exist leading to the three events discussed here, and that these pathways do not per se depend upon each other.
Article
Lymphocytes have been plated onto the surface of three-dimensional gels of native collagen fibers, and their distribution throughout the three-dimensional collagen matrix has been determined in a quantitative fashion at various times thereafter. Information regarding the total number of applied cells may be obtained by this means. Lymphocyte penetration into the collagen gel does not appear to involve the expression of collagenolytic activity, nor does it require the presence of serum. Analysis of the kinetics of lymphocyte penetration into the gel matrix indicates that lymphocytes are migrating in a "random-walk" fashion. Our objective has been to establish a model system for studying the cell-matrix and cell-cell interactions which influence the pattern of lymphocyte recirculation in vivo and the results presented here are discussed in this context.
Article
Lymphocyte motility is vital for trafficking within lymphoid organs and for initiating contact with antigen-presenting cells. Visualization of these processes has previously been limited to in vitro systems. We describe the use of two-photon laser microscopy to image the dynamic behavior of individual living lymphocytes deep within intact lymph nodes. In their native environment, T cells achieved peak velocities of more than 25 micrometers per minute, displaying a motility coefficient that is five to six times that of B cells. Antigenic challenge changed T cell trajectories from random walks to "swarms" and stable clusters. Real-time two-photon imaging reveals lymphocyte behaviors that are fundamental to the initiation of the immune response.
Article
Proteases produced by immune cells have been found to be important components of the immune response to antigen. A protease previously unrecognized as a specific T cell product has been identified which has the gene sequence, serologic crossreactivity, and enzymatic specificity of elastase. T cell elastase, found in combination with the natural elastase inhibitor α1-antitrypsin (α1-protease inhibitor, α1-PI), is produced by both CD4+ and CD8+ T lymphocytes, and is found both in a membrane-bound and in a soluble form in murine T cell lines.