ArticlePDF AvailableLiterature Review

Dual contrasting roles of cysteine cathepsins in cancer progression: Apoptosis versus tumour invasion

Authors:

Abstract and Figures

Cysteine cathepsins have been known for a long time to play an important role in cancer progression and metastasis. Several studies have proposed the concept of anti-cathepsin therapy in cancer treatment. On the other hand, cysteine cathepsins have been recently found to play a role in tumour cell death through mediation of apoptosis. The purpose of this mini-review is therefore to provide an insight into the mechanisms by which cysteine cathepsins modulate apoptosis and/or participate in tumour invasion, and to evaluate the impact of these enzymes on both tumour progression and development of potential strategies for cancer treatment.
Content may be subject to copyright.
This article was published in an Elsevier journal. The attached copy
is furnished to the author for non-commercial research and
education use, including for instruction at the author’s institution,
sharing with colleagues and providing to institution administration.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy
Research paper
Dual contrasting roles of cysteine cathepsins in
cancer progression: Apoptosis versus tumour invasion
Olga Vasiljeva*, Boris Turk*
Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
Received 20 July 2007; accepted 12 October 2007
Available online 18 October 2007
Abstract
Cysteine cathepsins have been known for a long time to play an important role in cancer progression and metastasis. Several studies have
proposed the concept of anti-cathepsin therapy in cancer treatment. On the other hand, cysteine cathepsins have been recently found to play
a role in tumour cell death through mediation of apoptosis. The purpose of this mini-review is therefore to provide an insight into the mech-
anisms by which cysteine cathepsins modulate apoptosis and/or participate in tumour invasion, and to evaluate the impact of these enzymes
on both tumour progression and development of potential strategies for cancer treatment.
Ó2007 Elsevier Masson SAS. All rights reserved.
Keywords: Cysteine cathepsins; Cancer progression; Apoptosis; Tumour invasion; Cancer treatment
1. Introduction
In the last decade, it has become evident that multicellular
organisms are protected against redundant and potentially
harmful cells by a highly sophisticated program, termed apo-
ptosis. Apoptosis is a natural mechanism of cellular self de-
struction that is characterized by specific morphological
features, including cell shrinkage, membrane blebbing, chro-
matin condensation and nuclear fragmentation. Apoptosis is
a key process for the development and maintenance of cellular
homeostasis, and acts as part of a quality-control and repair
mechanism by elimination of unwanted, genetically damaged,
or senescent cells and as such is also critically important for
the development of multicellular organisms. In fact, defects
in apoptotic pathways contribute to a number of human
diseases, ranging from neurodegenerative disorders to malig-
nancy [1,2]. The molecular mechanisms that control and exe-
cute apoptotic cell death in cancer growth and resistance have
been coming into focus and are enabling a new era of drug de-
velopment for cancer treatment [3].
A family of cysteinyl aspartate-specific proteases, called
the caspases (Clan CD; C14 family), have a critical role in ap-
optosis [4]. Other proteases have also been found to participate
in apoptosis, among them being cysteine cathepsins (Clan CA;
C1 papain family) [5]. However, cysteine cathepsins are also
well known to play a tumour-promoting function in cancer
progression for example by actively participating in tumour in-
vasion processes [6]. The purpose of this mini-review is there-
fore to provide an insight into the mechanisms by which
cysteine cathepsins modulate apoptosis and/or participate in
tumour invasion, and to evaluate the impact of these enzymes
both for tumour progression and for potential strategies of can-
cer treatment.
2. Cysteine cathepsins
Eleven human cysteine cathepsins have been identified: ca-
thepsins B, C (J, dipeptidyl peptidase I), F, H, K (O, O2), L,
Abbreviations: BH3, Bcl-2 homology domain 3; Bid, BH3-interacting
death domain; ECM, extracellular matrix; MHC, major histocompatibility
complex; uPA, urokinase plasminogen activator; TNF, tumour necrosis factor;
TRAIL, TNF-related apoptosis inducing ligand.
* Corresponding authors. Tel.: þ386 1 477 3772; fax: þ386 1 477 3984.
E-mail addresses: olga.vasiljeva@ijs.si (O. Vasiljeva), boris.turk@ijs.si
(B. Turk).
0300-9084/$ - see front matter Ó2007 Elsevier Masson SAS. All rights reserved.
doi:10.1016/j.biochi.2007.10.004
A
vailable online at www.sciencedirect.com
Biochimie 90 (2008) 380e386
www.elsevier.com/locate/biochi
Author's personal copy
O, S, V (L2), W (lymphopain) and X (P, Y, Z) [7]. All have
been biochemically characterized except cathepsins O and
W[8,9]. Cysteine cathepsins share a common fold of the pa-
pain-like structure and are synthesized as inactive zymogens
that are activated by proteolytic removal of the N-terminal
propeptide [10]. Most of them are endopeptidases, with cathep-
sins H and B also being an aminopeptidase and a carboxydipep-
tidase, respectively [11,12]. The only true exopeptidases are
the aminodipeptidase cathepsin C and carboxymonopeptidase
cathepsin X [13]. Cysteine cathepsin activity is regulated by
several mechanisms that comprise regulation of synthesis, zy-
mogen processing, inhibition by endogenous inhibitors (e.g.
cystatins) and pH stability (reviewed in [8]).
For many years cysteine cathepsins were believed to be lo-
calized exclusively in lysosomes, however, it is clear now that
cathepsins including their splice variants are also found in
other cellular and extracellular compartments, including the
nucleus [14,15], extracellular milieu [16,17] and plasma mem-
brane [17,18], where they fulfil important functions. Besides
their main function as a lysosomal protein recycling machine,
the cathepsins have been shown to play roles in a variety of
physiological and pathological processes [9,10]. Cysteine ca-
thepsins are involved in major histocompatibility complex
(MHC) class II-mediated antigen presentation by generation
of the antigenic peptides and by processing the MHC class
II invariant chain, thereby participating in the maturation of
the MHC class II complex. Cathepsins also play a role in
bone remodelling, precursor protein activation (e.g. prohor-
mones), keratinocyte differentiation, etc. In addition, cysteine
cathepsins have also been demonstrated to participate in sev-
eral pathological conditions such as tumour progression and
metastasis, rheumatoid arthritis and osteoarthritis, and athero-
sclerosis (reviewed in [9]).
3. Cysteine cathepsins in apoptosis
Two alternative pathways can trigger apoptosis in mam-
mals: one is mediated by death receptors from the TNF family
on the cell surface (referred to as the ‘extrinsic pathway’) and
the other is mediated by mitochondria (referred to as the ‘in-
trinsic pathway’). The extrinsic pathway is initiated at the
cell surface through cytokine-induced death receptor-mediated
activation of caspase-8 (-10) followed by caspase-3 and -7
activation [19]. The intrinsic pathway is characterized by mi-
tochondrial dysfunction, resulting in release of cytochrome c,
followed by apoptosome formation and subsequent activation
of caspase-9 and, subsequently caspases-3 and -7. The latter
caspases, also called the executioner caspases, are responsible
for the morphological changes during apoptosis, including
membrane blebbing, cell shrinkage, and DNA fragmentation.
Both pathways are connected through Bid, a pro-apoptotic
Bcl-2 family member, which can be cleaved by caspase 8,
generating a truncated Bid fragment (tBid) [20] that induces
mitochondrial outer membrane permeabilization (MOMP)
via the multidomain pro-death molecules Bax or Bak [21].
In turn, Bax/Bak translocation to mitochondria induces the
release of cytochrome cinto the cytosol and subsequent acti-
vation of the executioner caspases.
Recently, cysteine cathepsins have attracted considerable
attention as potential mediators of apoptosis. It is well estab-
lished that massive lysosomal rupture would induce necrotic
autolysis of cells (cellular necrosis), a process mediated by
the lysosomal cathepsins and other ‘‘acidic’’ hydrolases. On
the other hand, more selective release of cysteine cathepsins
as a consequence of limited lysosome damage has been sug-
gested to lead to apoptosis, as also demonstrated in L-leucyl-
L-leucine methyl ester -induced apoptosis in HeLa cells [22].
Whereas lower concentrations of this lysosomotropic agent in-
duced apoptotic cell death, the use of higher concentrations re-
sulted in necrotic cell death with no caspase activation observed.
However, despite numerous reports on their involvement in ap-
optosis, the mechanism(s) are still not clear, except that they
need to be released into the cytosol in order to exert their proa-
poptotic activity. Several models for the release of lysosomal
cathepsins release into the cytosol have been proposed, mostly
based on the mode of lysosomal membrane permeabilization
[23,24]. Most of these stimuli directly target the lysosomal
membrane, either through lysosomal destabilization or
through lysosomal membrane permeabilization. These include
bile salts [25], sphingosine [26], reactive oxygen species [27],
polyclonal antithymocyte globulins [28],L-leucyl-L-leucine
methyl ester [22,29], microtubule stabilizing agents [30],
cold ischemia-warm reperfusion [31], or hexadecylphospho-
cholines [32]. Furthermore, it is now well established that ly-
sosomal permeabilization could occur in response to specific
signalling processes induced by death ligands such as tumour
necrosis factor a(TNF-a)[33] and TNF-related apoptosis in-
ducing ligand (TRAIL) [34,35].
Once released into the cytosol, cysteine cathepsins can in-
duce liberation of cytochrome cfrom the mitochondria, which
was suggested to proceed through the cleavage of pro-apopto-
tic factors, such as Bid [36]. It was shown that almost all cys-
teine cathepsin endopeptidases are capable of cleaving Bid in
the flexible loop to its truncated form [22]. Furthermore, ex-
perimental induction of early lysosomal destabilization by
treating cells with the lysosomotropic agent L-leucyl-L-leucine
methyl ester induced cleavage of Bid and cytochrome crelease
from mitochondria [22]. However, alternative pathways have
been proposed [37]. There is also evidence for a role of ca-
thepsins in the execution of cell death independent of caspase
activation, which results in an apoptosis-like morphology
[38,39].
4. Cysteine cathepsins in cancer
Proteases are known to participate in different stages of
cancer progression. The first reports linking cathepsins to can-
cer originate from the 1930s [40] from studies on tumour
transplantation in rats. The second wave of interest for cathep-
sins in cancer occurred in the late 1960s [41,42], when raised
activity of cathepsins was detected in human tumour samples,
and has continued to the present. However, in spite of such
a relatively long history, the mechanisms and pathways for
381O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
Author's personal copy
the involvement of cysteine cathepsins in cancer are still under
investigation. Increased activity and/or expression of cysteine
cathepsins, mainly cathepsin B, has been detected in many hu-
man tumours, including breast [43e45], lung [46,47], brain
[48e50], gastrointestinal [51e53], prostate [54,55] cancer
and melanoma [56,57], especially in aggressive cancer cells
(for a review see [58]). Moreover, increased expression of cys-
teine cathepsins has been shown to have diagnostic and prog-
nostic value for patients with a variety of malignancies [59].
During malignant progression, peripheral redistribution of
lysosomes with subsequent translocation of cysteine cathep-
sins to the cell surface and/or their secretion into the extracel-
lular milieu appear to be general phenomena in tumour cells
[60e64]. Several mechanism for extracellular translocation
of cathepsins were proposed, among them acidification of
the tumour microenvironment, characteristic of many tumours,
was suggested to induce upregulation of extracellular cysteine
cathepsins, mainly cathepsin B [65,66]. Many studies support
the important role of cysteine cathepsins in the remodelling of
extracellular matrix (ECM) in the tumour microenvironment,
thus promoting tumour invasion and metastasis [67e69].
The proteolytic activity of pericellular cysteine cathepsins ap-
pears to be required for (i) direct proteolysis of ECM compo-
nents such as laminin, fibronectin, type IV collagen, (ii)
initiation of the proteolytic cascade by activating other prote-
ases such as pro-uPA (urokinase plasminogen activator) [70]
and (iii) inactivation of cell-adhesion proteins such as E-cad-
herin (reviewed in [6,71]). However, despite the major func-
tion of extracellularly translocated cysteine cathepsins in
ECM remodelling, the intracellular-vesicular proteolysis was
shown to be also important for digestion of ECM proteins
[72e74].
Cysteine cathepsins participate in hallmark tumour pro-
cesses such as angiogenesis, cell proliferation, apoptosis and
invasion [75]. Furthermore, the use of transgenic mouse cancer
models, in combination with deletion of specific cathepsin
genes, provided the possibility of dissecting their individual
roles in cancer development. This approach was used for pan-
creatic islet tumours (RIP1-Tag2) and for mammary cancer
(MMTV-PyMT) transgenic mouse models [63,71]. Both stud-
ies proved the important role of cysteine cathepsins for tumour
progression and invasion, and two important lessons can be
learned. First, individual cysteine cathepsin genes make dis-
tinctive contributions to tumourigenesis. It was revealed by
the investigation of four cysteine cathepsins null mutations
in the same RIP1-Tag2 mouse model that each enzyme has
an individual, important function in key processes such as tu-
mour cell proliferation, apoptosis, angiogenesis and tumour in-
vasion [71]. For instance, cathepsins B and L knockouts
exhibit reduce cell proliferation and tumour growth, cathepsins
B and S impaired tumour angiogenesis, and deletion of cathep-
sins B, L and S increases tumour cell death, while genetic ab-
lation of cathepsin C had no effect on any of those processes.
The second important message is that depletion of one prote-
ase might induce compensatory upregulation of other family
members. Vasiljeva et al. [63] have shown that genetic abla-
tion of cathepsin B in primary PyMT tumour cells induces
membrane translocation of another carboxypeptidase, cathep-
sin X, that restores the invasive capability of tumour cells and
annihilates the effect of the principal protease deletion on the
level of lung metastasis.
Taken together, these results support the potential therapeu-
tic value of cysteine cathepsins as anti-cancer drug targets and
suggest the use of selective inhibitors of cathepsins for tumour
treatment.
5. Cysteine cathepsins in cancer cell death
Apoptosis is a major defence mechanism against malignant
transformation. Defects in the processes controlling apoptosis
can extend the life span of a cell, thereby contributing to neo-
plastic cell expansion independent of cell division. Moreover,
failures in normal apoptotic pathways contribute to carcino-
genesis by creating a permissive environment for genetic insta-
bility and accumulation of gene mutations. Thus, elucidation
of the new cell death pathways will provide new insights
into tumour biology, revealing novel strategies for combating
cancer [3,76].
There is considerable evidence that cysteine cathepsins
play an important role in executing the apoptotic program in
several tumour cell lines induced by death ligands such as tu-
mour necrosis factor a(TNF-a)[38,77] or TNF-related apo-
ptosis inducing ligand (TRAIL) [34]. Cysteine cathepsins
have been shown to participate in TNF-ainduced apoptosis
of human cervical carcinoma ME-180, murine fibrosarcoma
WEHI-S [38], ovarian cancer OV-90, prostatic cancer PC-3,
hepatoma SMMC-7721 [77] cells. In the last few years, an ap-
optosis inducing agent TRAIL that was shown to specifically
target cancer cells without damaging normal cells and tissues
has received special interest in cancer therapy. The presence
of a cathepsin-mediated proteolytic event in the apoptotic
pathway triggered by TRAIL was recently discovered by sev-
eral groups. Initially the inhibition of TRAIL induced apopto-
sis was observed upon treatment of cells with cathepsin
inhibitors in oral squamous carcinoma cells [78]. Subse-
quently, the pathway for apoptosis induction through Bid ac-
tivation was confirmed for several other tumour cell lines
[34,35,79].
The lysosomal pathways of apoptosis are suggested to be
more complex, containing additional components such as
anti-apoptotic enzyme sphingosine kinase 1 that was shown
to be inactivated by cytosol released cathepsin B [80]. In ad-
dition, the human homologue of SETA binding protein 1
[81], bikunin and TSRC1 [77] were shown to interact with ca-
thepsin B in vitro and in vivo and to participate in the lyso-
somal enzymes-triggered pathway of TNF-a- induced
apoptosis.
Despite the well characterized mechanism for the pro-apo-
ptotic function of cysteine cathepsins through cleavage of Bid
and activation of the mitochondrial pathway of caspase-depen-
dent apoptosis, there have been reports suggesting dominant
execution functions of cysteine cathepsins in apoptosis, even
in the presence of pan-caspase inhibitors [38]. However,
some contradictory findings have been published, confirming
382 O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
Author's personal copy
the anti-apoptotic function of cysteine cathepsins, mostly con-
centrating on cathepsin L [71,82e84].
Thus, summarizing the results derived from a variety of re-
ports, it has become evident that cysteine cathepsins are in-
volved in different pathways to apoptosis in a manner
dependent on cell type and cell stress involved. This conclu-
sion is supported by results obtained recently by analysis of
apoptosis in cathepsin B deficient mouse cells. Whereas prote-
ase depleted hepatocytes were resistant to TNF-a- induced ap-
optosis both in vitro and in vivo [33,85], analysis of mammary
tumour cells from cathepsin B knock-out mice did not reveal
any differences either in spontaneous apoptosis in vivo or in
TNF-ainduced apoptosis in vitro (Vasiljeva et al., submitted).
Further, a significant increase in spontaneous apoptosis was
detected in cathepsin B depleted pancreatic tumour cells in
vivo [71]. In all cases, results were compared with those
from the respective cathepsin B wild-type cells. Nevertheless,
induction of lysosomal permeabilization would always result
in release of cathepsins into the cytoplasm where they can trig-
ger apoptotic cell death via various pathways [24] and thus
may prove effective in cancer treatment.
The use of chloroquine, a lysosomotropic antimalaria drug,
might serve as a successful example of such a pharmacological
approach as it has been shown to inhibit cell growth and in-
duce cell death in lung cancer cells [86], Myc-induced model
of lymphoma [87] and was used in clinical trials as adjuvant
therapy for patients with glioblastoma multiforme [88]. An-
other example is the use of lysosomotropic agent L-leucyl-L-
leucine ester, which was shown to be extremely effective in
killing various immune and myeloid tumour cells of bone-
marrow origin and to efficiently protect against graft-vs.-host
disease in a mouse model of bone marrow transplantation
Cytochrome c
TNF-α
TRAIL
Lysosome
Cathepsins
Caspase 8
APOPTOSIS
Mitochondria
Caspase 9
Caspase 3, 7
Bid
Bax/Bak
Caveolae
Collagen
TUMOR INVASION
Intracellular stimuli
ECM degradation
E-cadherin
MMP
Plasmin
uPA
Fig. 1. The multiple roles of cysteine cathepsins in tumour invasion and apoptosis. Depending on their localization, cysteine cathepsins can be involved in two
apparently opposing mechanisms of cancer progression: tumour invasion and apoptosis. Cysteine cathepsins that are secreted or associated with the plasma mem-
brane (e.g. caveolae) participate in tumour invasion through proteolytic cascade activation, ECM degradation and inactivation of cell-adhesion proteins (e.g. E-
cadherin). In addition, intracellular proteolysis by cysteine cathepsins contributes to degradation of endocytosed collagen in lysosomes. On the other hand, cytosol
translocated cysteine cathepsins trigger apoptosis via Bid cleavage followed by cytochrome crelease from mitochondria. This cascade activates downstream ex-
ecutioner caspases resulting in tumour cell death.
383O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
Author's personal copy
[89]. The compound is currently in Phase II clinical trials for
allogeneic hematopoietic stem cell transplantation (http://
clinicaltrials.gov).
6. Conclusion and future directions
We have put forward a scheme (Fig. 1), which resolves the
apparent incompatibility of the dual role that cysteine cathep-
sins play in cancer progression: first by promoting tumour in-
vasion through ECM degradation, etc., and second by
participating in apoptotic pathways. Although these two path-
ways have apparently opposing functions, there is a clear
physical separation of the processes, enabling the possibility
of their selective targeting. Whereas the proteolysis required
for tumour invasion occurs mostly in the extracellular space,
the apoptosis pathway is clearly intracellular and dependent
on translocation of cathepsins into the cytosol. Therefore,
this differential distribution appears to be the key feature
that separates the two functions of cysteine cathepsins de-
scribed and provides an interesting and unique example how
the same enzymes can have such contradictory functions.
Given our current understanding of cathepsin activity in
tumour progression, it is evident that cysteine proteases could
be a rational target for cancer therapy. However, the pharmaceu-
tical approaches must differ, depending on the particular process
one would aim at. Consequently, while the use of a lysosomo-
tropic agents inducing lysosomal membrane permeabilization
could promote tumour cell death, the use of non cell-permeable
inhibitors of cathepsins would decrease the invasivepotential of
tumour cells in vivo. This underlines the need for further re-
search on the molecular mechanisms of the processes that cyste-
ine cathepsins are involved in, and on the development of
specific cathepsin inhibitors and lysosomotropic agents to gen-
erate new cancer therapies.
Acknowledgements
We apologize to the many contributors to this field whose
work we were unable to cite owing to space restrictions. We
would like to thank Dr Veronika Stoka for insightful discus-
sions and Prof. Roger H. Pain for critical reading of the man-
uscript. This work was supported by grants from the Ministry
of Higher Education, Science and Technology of the Republic
of Slovenia and from Human Frontiers Science Program
(RGP0024/2006-C).
References
[1] C.B. Thompson, Apoptosis in the pathogenesis and treatment of disease,
Science 267 (1995) 1456e1462.
[2] G. Kroemer, J.C. Reed, Mitochondrial control of cell death, Nat. Med. 6
(2000) 513e519.
[3] J.C. Reed, Apoptosis-targeted therapies for cancer, Cancer Cell 3 (2003)
17e22.
[4] P. Fuentes-Prior, G.S. Salvesen, The protein structures that shape caspase
activity, specificity, activation and inhibition, Biochem. J. 384 (2004)
201e232.
[5] B. Turk, V. Stoka, J. Rozman-Pungercar, T. Cirman, G. Droga-Mazovec,
K. Oresic, V. Turk, Apoptotic pathways: involvement of lysosomal
proteases, Biol. Chem. 383 (2002) 1035e1044.
[6] M.M. Mohamed, B.F. Sloane, Cysteine cathepsins: multifunctional en-
zymes in cancer, Nat. Rev. Cancer 6 (2006) 764e775.
[7] A. Rossi, Q. Deveraux, B. Turk, A. Sali, Comprehensive search for cyste-
ine cathepsins in the human genome, Biol. Chem. 385 (2004) 363e372.
[8] B. Turk, D. Turk, V. Turk, Lysosomal cysteine proteases: more than scav-
engers, Biochim. Biophys. Acta 1477 (2000) 98e111.
[9] O. Vasiljeva, T. Reinheckel, C. Peters, D. Turk, V. Turk, B. Turk, Emerg-
ing roles of cysteine cathepsins in disease and their potential as drug tar-
gets, Curr. Pharm. Des. 13 (2007) 385e401.
[10] V. Turk, B. Turk, G. Guncar, D. Turk, J. Kos, Lysosomal cathepsins:
structure, role in antigen processing and presentation, and cancer, Adv.
Enzyme Regul. 42 (2002) 285e303.
[11] O. Vasiljeva, M. Dolinar, V. Turk, B. Turk, Recombinant human cathep-
sin H lacking the mini chain is an endopeptidase, Biochemistry 42 (2003)
13522e13528.
[12] D. Musil, D. Zucic, D. Turk, R.A. Engh, I. Mayr, R. Huber, T. Popovic,
V. Turk, T. Towatari, N. Katunuma, et al., The refined 2.15 A X-ray crys-
tal structure of human liver cathepsin B: the structural basis for its spec-
ificity, EMBO J. 10 (1991) 2321e2330.
[13] D. Turk, B. Turk, V. Turk, Papain-like lysosomal cysteine proteases and
their inhibitors: drug discovery targets? Biochem. Soc. Symp. (2003)
15e30.
[14] B. Goulet, A. Baruch, N.S. Moon, M. Poirier, L.L. Sansregret,
A. Erickson, M. Bogyo, A. Nepveu, A cathepsin L isoform that is devoid
of a signal peptide localizes to the nucleus in S phase and processes the
CDP/Cux transcription factor, Mol. Cell 14 (2004) 207e219.
[15] B. Goulet, L. Sansregret, L. Leduy, M. Bogyo, E. Weber, S.S. Chauhan,
A. Nepveu, Increased expression and activity of nuclear cathepsin L in
cancer cells suggests a novel mechanism of cell transformation, Mol.
Cancer Res. 5 (2007) 899e907.
[16] S. Roshy, B.F. Sloane, K. Moin, Pericellular cathepsin B and malignant
progression, Cancer Metastasis Rev. 22 (2003) 271e286.
[17] D.T. Jane, L. Morvay, L. Dasilva, D. Cavallo-Medved, B.F. Sloane,
M.J. Dufresne, Cathepsin B localizes to plasma membrane caveolae of
differentiating myoblasts and is secreted in an active form at physiolog-
ical pH, Biol. Chem. 387 (2006) 223e234.
[18] A.M. Lechner, I. Assfalg-Machleidt, S. Zahler, M. Stoeckelhuber,
W. Machleidt, M. Jochum, D.K. Nagler, RGD-dependent binding of pro-
cathepsin X to integrin alphavbeta3 mediates cell-adhesive properties, J.
Biol. Chem. 281 (2006) 39588e39597.
[19] M.O. Hengartner, The biochemistry of apoptosis, Nature 407 (2000)
770e776.
[20] H. Li, H. Zhu, C.J. Xu, J. Yuan, Cleavage of BID by caspase 8 mediates
the mitochondrial damage in the Fas pathway of apoptosis, Cell 94
(1998) 491e501.
[21] S. Desagher, A. Osen-Sand, A. Nichols, R. Eskes, S. Montessuit,
S. Lauper, K. Maundrell, B. Antonsson, J.C. Martinou, Bid-induced con-
formational change of Bax is responsible for mitochondrial cytochrome c
release during apoptosis, J. Cell Biol. 144 (1999) 891e901.
[22] T. Cirman, K. Oresic, G.D. Mazovec, V. Turk, J.C. Reed, R.M. Myers,
G.S. Salvesen, B. Turk, Selective disruption of lysosomes in HeLa cells
triggers apoptosis mediated by cleavage of Bid by multiple papain-like
lysosomal cathepsins, J. Biol. Chem. 279 (2004) 3578e3587.
[23] C. Tardy, P. Codogno, H. Autefage, T. Levade, N. Andrieu-Abadie, Ly-
sosomes and lysosomal proteins in cancer cell death (new players of
an old struggle), Biochim. Biophys. Acta 1765 (2006) 101e125.
[24] V. Stoka, V. Turk, B. Turk, Lysosomal cysteine cathepsins: signaling
pathways in apoptosis, Biol. Chem. 388 (2007) 555e560.
[25] L.R. Roberts, H. Kurosawa, S.F. Bronk, P.J. Fesmier, L.B. Agellon,
W.Y. Leung, F. Mao, G.J. Gores, Cathepsin B contributes to bile salt-
induced apoptosis of rat hepatocytes, Gastroenterology 113 (1997)
1714e1726.
[26] K. Kagedal, M. Zhao, I. Svensson, U.T. Brunk, Sphingosine-induced
apoptosis is dependent on lysosomal proteases, Biochem. J. 359 (2001)
335e343.
384 O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
Author's personal copy
[27] M. Zhao, F. Antunes, J.W. Eaton, U.T. Brunk, Lysosomal enzymes pro-
mote mitochondrial oxidant production, cytochrome crelease and apo-
ptosis, Eur. J. Biochem. 270 (2003) 3778e3786.
[28] M.C. Michallet, F. Saltel, X. Preville, M. Flacher, J.P. Revillard,
L. Genestier, Cathepsin-B-dependent apoptosis triggered by antithymo-
cyte globulins: a novel mechanism of T-cell depletion, Blood 102
(2003) 3719e3726.
[29] D.L. Thiele, P.E. Lipsky, Mechanism of L-leucyl-L-leucine methyl ester-
mediated killing of cytotoxic lymphocytes: dependence on a lysosomal
thiol protease, dipeptidyl peptidase I, that is enriched in these cells,
Proc. Natl. Acad. Sci. U.S.A. 87 (1990) 83e87.
[30] L.E. Broker, C. Huisman, S.W. Span, J.A. Rodriguez, F.A. Kruyt,
G. Giaccone, Cathepsin B mediates caspase-independent cell death in-
duced by microtubule stabilizing agents in non-small cell lung cancer
cells, Cancer Res. 64 (2004) 27e30.
[31] E.S. Baskin-Bey, A. Canbay, S.F. Bronk, N. Werneburg, M.E. Guicciardi,
S.L. Nyberg, G.J. Gores, Cathepsin B inactivation attenuates hepatocyte
apoptosis and liver damage in steatotic livers after cold ischemia-warm
reperfusion injury, Am. J. Physiol. Gastrointest. Liver Physiol. 288
(2005) G396eG402.
[32] C. Paris, J. Bertoglio, J. Breard, Lysosomal and mitochondrial pathways
in miltefosine-induced apoptosis in U937 cells, Apoptosis 12 (2007)
1257e1267.
[33] M.E. Guicciardi, J. Deussing, H. Miyoshi, S.F. Bronk, P.A. Svingen,
C. Peters, S.H. Kaufmann, G.J. Gores, Cathepsin B contributes to
TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial
release of cytochrome c, J. Clin. Invest. 106 (2000) 1127e1137.
[34] N.S. Nagaraj, N. Vigneswaran, W. Zacharias, Cathepsin B mediates
TRAIL-induced apoptosis in oral cancer cells, J. Cancer Res. Clin. On-
col. 132 (2006) 171e183.
[35] T.O. Garnett, M. Filippova, P.J. Duerksen-Hughes, Bid is cleaved up-
stream of caspase-8 activation during TRAIL-mediated apoptosis in hu-
man osteosarcoma cells, Apoptosis 12 (2007) 1299e1315.
[36] V. Stoka, B. Turk, S.L. Schendel, T.H. Kim, T. Cirman, S.J. Snipas,
L.M. Ellerby, D. Bredesen, H. Freeze, M. Abrahamson, D. Bromme,
S. Krajewski, J.C. Reed, X.M. Yin, V. Turk, G.S. Salvesen, Lysosomal
protease pathways to apoptosis. Cleavage of bid, not pro-caspases, is
the most likely route, J. Biol. Chem. 276 (2001) 3149e3157.
[37] M.E. Guicciardi, S.F. Bronk, N.W. Werneburg, X.M. Yin, G.J. Gores, Bid
is upstream of lysosome-mediated caspase 2 activation in tumor necrosis
factor alpha-induced hepatocyte apoptosis, Gastroenterology 129 (2005)
269e284.
[38] L. Foghsgaard, D. Wissing, D. Mauch, U. Lademann, L. Bastholm,
M. Boes, F. Elling, M. Leist, M. Jaattela, Cathepsin B acts as a dominant
execution protease in tumor cell apoptosis induced by tumor necrosis fac-
tor, J. Cell Biol. 153 (2001) 999e1010.
[39] J.H. Li, J.S. Pober, The cathepsin B death pathway contributes to TNF
plus IFN-gamma-mediated human endothelial injury, J. Immunol. 175
(2005) 1858e1866.
[40] A. Purr, Cathepsin in rats with transplantable cancer, Biochem. J. 28
(1934) 1907e1910.
[41] D.J. Etherington, W.H. Taylor, Investigation of the cathepsins of human
gastric carcinomata, Biochem. J. 115 (1969) 43Pe44P.
[42] T. Schersten, L. Wahlqvist, L.G. Johansson, Lysosomal enzyme activity
in liver tissue from patients with renal carcinoma, Cancer 23 (1969)
608e613.
[43] A.R. Poole, K.J. Tiltman, A.D. Recklies, T.A. Stoker, Differences in se-
cretion of the proteinase cathepsin B at the edges of human breast carci-
nomas and fibroadenomas, Nature 273 (1978) 545e547.
[44] T. Castiglioni, M.J. Merino, B. Elsner, T.T. Lah, B.F. Sloane,
M.R. Emmert-Buck, Immunohistochemical analysis of cathepsins D,
B, and L in human breast cancer, Hum. Pathol. 25 (1994) 857e862.
[45] A. Bervar, I. Zajc, N. Sever, N. Katunuma, B.F. Sloane, T.T. Lah, Inva-
siveness of transformed human breast epithelial cell lines is related to ca-
thepsin B and inhibited by cysteine proteinase inhibitors, Biol. Chem.
384 (2003) 447e455.
[46] B. Werle, H. Lotterle, U. Schanzenbacher, T.T. Lah, E. Kalman,
K. Kayser, H. Bulzebruck, J. Schirren, M. Krasovec, J. Kos, E. Spiess,
Immunochemical analysis of cathepsin B in lung tumours: an indepen-
dent prognostic factor for squamous cell carcinoma patients, Br. J. Can-
cer 81 (1999) 510e519.
[47] N. Fujise, A. Nanashim, Y. Taniguchi, S. Matsuo, K. Hatano,
Y. Matsumoto, Y. Tagawa, H. Ayabe, Prognostic impact of cathepsin B
and matrix metalloproteinase-9 in pulmonary adenocarcinomas by im-
munohistochemical study, Lung Cancer 27 (2000) 19e26.
[48] S.A. Rempel, M.L. Rosenblum, T. Mikkelsen, P.S. Yan, K.D. Ellis,
W.A. Golembieski, M. Sameni, J. Rozhin, G. Ziegler, B.F. Sloane, Ca-
thepsin B expression and localization in glioma progression and invasion,
Cancer Res. 54 (1994) 6027e6031.
[49] T. Strojnik, J. Kos, B. Zidanik, R. Golouh, T. Lah, Cathepsin B immuno-
histochemical staining in tumor and endothelial cells is a new prognostic
factor for survival in patients with brain tumors, Clin. Cancer Res. 5
(1999) 559e567.
[50] T. Flannery, S. McQuaid, C. McGoohan, R.S. McConnell, G. McGregor,
M. Mirakhur, P. Hamilton, J. Diamond, G. Cran, B. Walker, C. Scott,
L. Martin, D. Ellison, C. Patel, C. Nicholson, D. Mendelow,
D. McCormick, P.G. Johnston, Cathepsin S expression: an independent
prognostic factor in glioblastoma tumours ea pilot study, Int. J. Cancer
119 (2006) 854e860.
[51] M. Watanabe, T. Higashi, A. Watanabe, T. Osawa, Y. Sato, Y. Kimura,
S. Tominaga, N. Hashimoto, Y. Yoshida, S. Morimoto, et al., Cathepsin
B and L activities in gastric cancer tissue: correlation with histological
findings, Biochem. Med. Metab. Biol. 42 (1989) 21e29.
[52] Y. Liu, S. Xiao, Y. Shi, L. Wang, W. Ren, B.F. Sloane, Cathepsin B on
invasion and metastasis of gastric carcinoma, Chin, Med. J. (Engl.)
111 (1998) 784e788.
[53] M.P. Ebert, S. Kruger, M.L. Fogeron, S. Lamer, J. Chen, M. Pross,
H.U. Schulz, H. Lage, S. Heim, A. Roessner, P. Malfertheiner,
C. Rocken, Overexpression of cathepsin B in gastric cancer identified
by proteome analysis, Proteomics 5 (2005) 1693e1704.
[54] A.A. Sinha, D.F. Gleason, N.A. Staley, M.J. Wilson, M. Sameni,
B.F. Sloane, Cathepsin B in angiogenesis of human prostate: an immuno-
histochemical and immunoelectron microscopic analysis, Anat. Rec. 241
(1995) 353e362.
[55] D.K. Nagler, S. Kruger, A. Kellner, E. Ziomek, R. Menard, P. Buhtz,
M. Krams, A. Roessner, U. Kellner, Up-regulation of cathepsin X in
prostate cancer and prostatic intraepithelial neoplasia, Prostate 60
(2004) 109e119.
[56] E. Frohlich, B. Schlagenhauff, M. Mohrle, E. Weber, C. Klessen,
G. Rassner, Activity, expression, and transcription rate of the cathepsins
B, D, H, and L in cutaneous malignant melanoma, Cancer 91 (2001)
972e982.
[57] J. Kos, B. Stabuc, A. Schweiger, M. Krasovec, N. Cimerman, N. Kopitar-
Jerala, I. Vrhovec, Cathepsins B, H, and L and their inhibitors stefin A
and cystatin C in sera of melanoma patients, Clin. Cancer Res. 3
(1997) 1815e1822.
[58] C. Jedeszko, B.F. Sloane, Cysteine cathepsins in human cancer, Biol.
Chem. 385 (2004) 1017e1027.
[59] J. Kos, T.T. Lah, Cysteine proteinases and their endogenous inhibitors:
target proteins for prognosis, diagnosis and therapy in cancer (review),
Oncol. Rep. 5 (1998) 1349e1361.
[60] L.L. Demchik, M. Sameni, K. Nelson, T. Mikkelsen, B.F. Sloane, Ca-
thepsin B and glioma invasion, Int. J. Dev. Neurosci. 17 (1999) 483e494.
[61] H. Buth, B. Wolters, B. Hartwig, R. Meier-Bornheim, H. Veith,
M. Hansen, C.P. Sommerhoff, N. Schaschke, W. Machleidt,
N.E. Fusenig, P. Boukamp, K. Brix, HaCaT keratinocytes secrete lyso-
somal cysteine proteinases during migration, Eur. J. Cell Biol. 83
(2004) 781e795.
[62] I. Podgorski, B.E. Linebaugh, M. Sameni, C. Jedeszko, S. Bhagat,
M.L. Cher, B.F. Sloane, Bone microenvironment modulates expression
and activity of cathepsin B in prostate cancer, Neoplasia 7 (2005) 207e223.
[63] O. Vasiljeva, A. Papazoglou, A. Kruger, H. Brodoefel, M. Korovin,
J. Deussing, N. Augustin, B.S. Nielsen, K. Almholt, M. Bogyo,
C. Peters, T. Reinheckel, Tumor cell-derived and macrophage-derived ca-
thepsin B promotes progression and lung metastasis of mammary cancer,
Cancer Res. 66 (2006) 5242e5250.
385O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
Author's personal copy
[64] A. Klose, P. Zigrino, R. Dennhofer, C. Mauch, N. Hunzelmann, Identifi-
cation and discrimination of extracellularly active cathepsins B and L in
high-invasive melanoma cells, Anal. Biochem. 353 (2006) 57e62.
[65] J. Rozhin, M. Sameni, G. Ziegler, B.F. Sloane, Pericellular pH affects
distribution and secretion of cathepsin B in malignant cells, Cancer
Res. 54 (1994) 6517e6525.
[66] E.K. Rofstad, B. Mathiesen, K. Kindem, K. Galappathi, Acidic extracel-
lular pH promotes experimental metastasis of human melanoma cells in
athymic nude mice, Cancer Res. 66 (2006) 6699e6707.
[67] M.R. Buck, D.G. Karustis, N.A. Day, K.V. Honn, B.F. Sloane, Degrada-
tion of extracellular-matrix proteins by human cathepsin B from normal
and tumour tissues, Biochem. J. 282 (Pt 1) (1992) 273e278.
[68] J. Mai, M. Sameni, T. Mikkelsen, B.F. Sloane, Degradation of extracel-
lular matrix protein tenascin-C by cathepsin B: an interaction involved in
the progression of gliomas, Biol. Chem. 383 (2002) 1407e1413.
[69] S. Mohanam, S.L. Jasti, S.R. Kondraganti, N. Chandrasekar, S.S. Lakka,
Y. Kin, G.N. Fuller, A.W. Yung, A.P. Kyritsis, D.H. Dinh, W.C. Olivero,
M. Gujrati, F. Ali-Osman, J.S. Rao, Down-regulation of cathepsin B ex-
pression impairs the invasive and tumorigenic potential of human glio-
blastoma cells, Oncogene 20 (2001) 3665e3673.
[70] H. Kobayashi, N. Moniwa, M. Sugimura, H. Shinohara, H. Ohi,
T. Terao, Effects of membrane-associated cathepsin B on the activation
of receptor-bound prourokinase and subsequent invasion of reconsti-
tuted basement membranes, Biochim. Biophys. Acta 1178 (1993)
55e62.
[71] V. Gocheva, W. Zeng, D. Ke, D. Klimstra, T. Reinheckel, C. Peters,
D. Hanahan, J.A. Joyce, Distinct roles for cysteine cathepsin genes in
multistage tumorigenesis, Genes Dev. 20 (2006) 543e556.
[72] M. Sameni, K. Moin, B.F. Sloane, Imaging proteolysis by living human
breast cancer cells, Neoplasia 2 (2000) 496e504.
[73] A.M. Szpaderska, A. Frankfater, An intracellular form of cathepsin B
contributes to invasiveness in cancer, Cancer Res. 61 (2001) 3493e3500.
[74] M. Sameni, J. Dosescu, B.F. Sloane, Imaging proteolysis by living hu-
man glioma cells, Biol. Chem. 382 (2001) 785e788.
[75] V. Gocheva, J.A. Joyce, Cysteine cathepsins and the cutting edge of can-
cer invasion, Cell Cycle 6 (2007) 60e64.
[76] S.W. Lowe, A.W. Lin, Apoptosis in cancer, Carcinogenesis 21 (2000)
485e495.
[77] J. Liu, Q. Guo, B. Chen, Y. Yu, H. Lu, Y.Y. Li, Cathepsin B and its
interacting proteins, bikunin and TSRC1, correlate with TNF-induced
apoptosis of ovarian cancer cells OV-90, FEBS Lett. 580 (2006)
245e250.
[78] N. Vigneswaran, J. Wu, N. Nagaraj, K. Adler-Storthz, W. Zacharias, Dif-
ferential susceptibility of metastatic and primary oral cancer cells to
TRAIL-induced apoptosis, Int. J. Oncol. 26 (2005) 103e112.
[79] M.E. Guicciardi, S.F. Bronk, N.W. Werneburg, G.J. Gores, cFLIPL pre-
vents TRAIL-induced apoptosis of hepatocellular carcinoma cells by in-
hibiting the lysosomal pathway of apoptosis, Am. J. Physiol.
Gastrointest. Liver Physiol. 292 (2007) G1337eG1346.
[80] T.A. Taha, K. Kitatani, J. Bielawski, W. Cho, Y.A. Hannun, L.M. Obeid,
Tumor necrosis factor induces the loss of sphingosine kinase-1 by a cathep-
sin B-dependent mechanism, J. Biol. Chem. 280 (2005) 17196e17202.
[81] J.P. Liu, N.S. Liu, H.Y. Yuan, Q. Guo, H. Lu, Y.Y. Li, Human homologue
of SETA binding protein 1 interacts with cathepsin B and participates in
TNF-induced apoptosis in ovarian cancer cells, Mol. Cell. Biochem. 292
(2006) 189e195.
[82] X. Zheng, P.M. Chou, B.L. Mirkin, A. Rebbaa, Senescence-initiated re-
versal of drug resistance: specific role of cathepsin L, Cancer Res. 64
(2004) 1773e1780.
[83] I. Zajc, I. Hreljac, T. Lah, Cathepsin L affects apoptosis of glioblastoma
cells: a potential implication in the design of cancer therapeutics, Anti-
cancer Res. 26 (2006) 3357e3364.
[84] C.S. Gondi, N. Kandhukuri, S. Kondraganti, M. Gujrati, W.C. Olivero,
D.H. Dinh, J.S. Rao, RNA interference-mediated simultaneous down-
regulation of urokinase-type plasminogen activator receptor and cathep-
sin B induces caspase-8-mediated apoptosis in SNB19 human glioma
cells, Mol. Cancer Ther. 5 (2006) 3197e3208.
[85] M.E. Guicciardi, H. Miyoshi, S.F. Bronk, G.J. Gores, Cathepsin B knock-
out mice are resistant to tumor necrosis factor-alpha-mediated hepatocyte
apoptosis and liver injury: implications for therapeutic applications, Am.
J. Pathol. 159 (2001) 2045e2054.
[86] C. Fan, W. Wang, B. Zhao, S. Zhang, J. Miao, Chloroquine inhibits cell
growth and induces cell death in A549 lung cancer cells, Bioorg. Med.
Chem. 14 (2006) 3218e3222.
[87] R.K. Amaravadi, D. Yu, J.J. Lum, T. Bui, M.A. Christophorou, G.I. Evan,
A. Thomas-Tikhonenko, C.B. Thompson, Autophagy inhibition enhances
therapy-induced apoptosis in a Myc-induced model of lymphoma, J.
Clin. Invest. 117 (2007) 326e336.
[88] J. Sotelo, E. Briceno, M.A. Lopez-Gonzalez, Adding chloroquine to
conventional treatment for glioblastoma multiforme: a randomized, dou-
ble-blind, placebo-controlled trial, Ann. Intern. Med. 144 (2006) 337e343.
[89] M. Charley, D.L. Thiele, M. Bennett, P.E. Lipsky, Prevention of lethal
murine graft versus host disease by treatment of donor cells with L-
leucyl-L-leucine methyl ester, J. Clin. Invest. 78 (1986) 1415e1420.
386 O. Vasiljeva, B. Turk / Biochimie 90 (2008) 380e386
... It is principally involved in the podosome-mediated degradation of the basement membrane and connective tissue via the secretion of lysosomes that eventually promotes tumor cell growth and nearby tissue progression. Thus, it clears the biological barriers for tissue invasion and finally metastasized through lymphatic and systemic circulation [81][82][83][84][85]. The indinavir's molecular interaction with cathepsin B revealed higher intermolecular interaction with low binding energy than the native ligand Diphenyl acetic Acid. ...
Article
Purpose The objective of the present investigation was to establish a molecular association of different proteases as cancer targets with the indinavir and how the physicochemical characteristics of the indinavir sulfate microsphere vary with different process variables was systemically established. Methods Molecular interactions with indinavir were identified and established by molecular simulation docking studies. Indinavir sulfate-loaded microspheres were prepared by the oil-in-oil emulsion solvent evaporation technique. Results Results indicated that indinavir could interact with all four proteases at the active binding site of receptors. Indinavir was found to show significantly higher interaction with Matrix Metalloproteases, Aspartate Proteases, and Cysteine Proteases with a binding energy of -8.80, -8.19, and -6.87, respectively, as compared to their native ligand. However, serine proteases exhibit less but significant interaction with a binding energy of -5.92 than the native ligand. The microspheres exhibited 72%-93% of entrapment and prolonged drug release (DR), up to 9 h. The drug-loaded microspheres showed invariable character by the Fourier-transform infrared spectroscopy (FTIR), differential scanning calorimetry (DSC), and thermographs and revealed no drug-polymer interactions. The decrease in the drug's crystallinity was observed in X-ray diffraction (XRD). The scanning electron microscope (SEM) study revealed the spherical and porous nature of microspheres. Conclusion Indinavir could act as a potential inhibitor of different proteases associated with tumor growth initiation, progression, and metastasis, and microspheres with sustained DR could be utilized to deliver an anticancer drug in a more targeted way as an emerging cancer microsphere technology.
... Through interactions with other proteases and their inhibitors, cathepsins can alter general proteolytic activity within the tumor microenvironment. In addition to direct regulation of multiple processes involved in tumor progression and metastasis, it is now evident that many proteases can have an indirect impact through activation of multiple cascades of enzymatic activities [236,237]. It can be illustrated by the processing of urokinase-type plasminogen activator (pro-uPA) pro-form by cathepsin B, leading to conversion of plasminogen into plasmin [238], which may activate zymogens of matrix metalloproteinases, and thus together with precursor proteases of this proteolytic activation cascade execute their numerous functions associated with tumor progression and metastasis [239]. ...
Preprint
Full-text available
Papain-like lysosomal cysteine proteases include 11 human cysteine cathepsins which act as endopeptidases and/or exopeptidases. They are involved in numerous physiological and pathological processes. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Their activities are tightly regulated in various ways to prevent potentially hazardous effects on the cell. In this review, we focus on the structural and functional aspects of these four cysteine cathepsins and their role in neurodegeneration and cancer. Neurodegenerative disorders of aging share an endolysosomal dysfunction and accumulation and spread of oligomeric forms of neurotoxic proteins. The accumulation of various protein aggregates activates the microglia, thus inducing the activation and release of cysteine cathepsins and proinflammatory cytokines, leading to neurodegeneration. In cancer, cysteine cathepsins participate in tumor progression and metastasis. Tumor–stromal crosstalk leads to activation of the stroma and overexpression and secretion of proteolytic enzymes, triggering extracellular matrix degradation and the release of soluble factors. Activated stromal cells (i.e., macrophages, fibroblasts, mast cells) secrete additional growth factors, cytokines, and chemokines that are responsible for the regulation of processes leading to tumor progression and metastasis. Therefore, a better understanding of the role of cysteine cathepsins in neurodegeneration and cancer could lead to novel targeted therapeutic approaches.
... Each group of proteases acts on different protein substrates and is involved in regulating different stages in the progression of cancer. For example, cysteine proteases (e.g. the cathepsin family of proteases) can degrade both intracellular and extracellular matrix (ECM) proteins [3], and assist tumour cell invasion into nearby tissues, blood and lymph vessels [4]. Serine proteases can degrade extracellular matrix proteins as well as growth factors such as epidermal growth factor (EGF), fibroblast growth factor-2 (FGF-2) and hepatocyte growth factor/scatter factor (HGF-SF) [5] and are involved in tumour cell invasion, angiogenesis and metastasis [6]. ...
Article
Full-text available
Galectin-3 is a galactoside-binding protein that is commonly overexpressed in many epithelial cancers. It is increasingly recognized as a multi-functional, multi-mode promoter in cancer development, progression, and metastasis. This study reports that galectin-3 secretion by human colon cancer cells induces cancer cell secretion, in an autocrine/paracrine manner, of a number of proteases including cathepsin-B, MMP-1 and MMP-13. The secretion of these proteases causes disruption of epithelial monolayer integrity, increases its permeability and promotes tumour cell invasion. This effect of galectin-3 is shown to be mediated through induction of cellular PYK2-GSK3α/β signalling and can be prevented by the presence of galectin-3 binding inhibitors. This study thus reveals an important mechanism in galectin-3-mediated promotion of cancer progression and metastasis. It provides further evidence to the increased realization of galectin-3 as a potential therapeutic target for the treatment of cancer. Cell Death and Disease (2023) 14:268 ; https://doi.
... As a consequence of these various agents, damaged mitochondria can generate increased amounts of reactive oxygen species, and their effects on lysosomal membranes can result in the release of lysosomal contents into the cytoplasm. It has also been shown that reactive oxygen species can activate phospholipase A2 (PLA2), which can cause destabilization and increased permeability of the lysosomal membrane [23,41,67,70,72,79,82,87,138,[149][150][151][152][153][154]. The free radicals show the ability to interact with free intralysosomal iron, creating highly reactive hydroxyl radicals in the Fenton-type reaction, which contributes to the induction of LMP through the ongoing lipid peroxidation of lysosomal membranes, resulting in the formation of lipofuscin and further damage to the lysosomal membrane proteins [79,155]. ...
Article
Full-text available
Lysosomes are organelles containing acidic hydrolases that are responsible for lysosomal degradation and the maintenance of cellular homeostasis. They play an important role in autophagy, as well as in various cell death pathways, such as lysosomal and apoptotic death. Various agents, including drugs, can induce lysosomal membrane permeability, resulting in the translocation of acidic hydrolases into the cytoplasm, which promotes lysosomal-mediated death. This type of death may be of great importance in anti-cancer therapy, as both cancer cells with disturbed pathways leading to apoptosis and drug-resistant cells can undergo it. Important compounds that damage the lysosomal membrane include lysosomotropic compounds, antihistamines, immunosuppressants, DNA-damaging drugs, chemotherapeutics, photosensitizers and various plant compounds. An interesting approach in the treatment of cancer and the search for ways to overcome the chemoresistance of cancer cells may also be combining lysosomotropic compounds with targeted modulators of autophagy to induce cell death. These compounds may be an alternative in oncological treatment, and lysosomes may become a promising therapeutic target for many diseases, including cancer. Understanding the functional relationships between autophagy and apoptosis and the possibilities of their regulation, both in relation to normal and cancer cells, can be used to develop new and more effective anticancer therapies.
... Select cathepsins that remain functional at neutral pH, including cathepsins B, D, and L, can activate apoptotic effectors following limited release from leaky lysosomes (166). Indeed, cathepsins are implicated in apoptotic cancer cell death in a variety of tumor models (169). Apoptotic pathways triggered by intrinsic factors such as DNA damage, endoplasmic reticulum stress, and LMP ultimately converge on mitochondrial membrane permeabilization (MOMP) and subsequent release of pro-apoptotic factors to the cytosol (166,(170)(171)(172). Mechanistic understanding of primary LMP in apoptosis was developed largely from studies using the lysosomotropic agent Leu-Leu-methyl ester (LLOMe) (173) and other apoptotic stimuli (153). ...
Chapter
Full-text available
ABSTRACT While patient-specific targeting of cellular growth and viability pathways dominates current approaches in anti cancer therapeutics development, appreciation for the strategy of targeting transformation-dependent alterations in cellular organelle structure and function continues to grow. Here we discuss the lysosome as an anti-cancer target, highlighting its role as a key mediator of cell death. As the major degradative compartment of the cell, the lysosome houses dozens of destructive enzymes and is responsible for the breakdown of both internal and external molecules and particles; however, until relatively recently the contribution of the lysosome to cellular death mechanisms has been largely overlooked. Renewed interest in the therapeutic potential of lysosomal rupture to combat cancer has led to development of lysosome-disrupting agents that induce lysosomal membrane permeabilization (LMP), cathepsin protease release, and subsequent lysosome dependent cell death (LDCD), now distinguished as a bona fide cell death process. Here, we present the basic biology, structure, and function of the lysosome, with particular emphasis on the transformation-associated alterations that sensitize cancer cell lysosomes to membrane rupture. We further describe the lysosome’s role in cell death and comprehensively outline emerging therapeutic strategies that exploit lysosomes for the treatment of a variety of malignancies.
... Select cathepsins that remain functional at neutral pH, including cathepsins B, D, and L, can activate apoptotic effectors following limited release from leaky lysosomes (166). Indeed, cathepsins are implicated in apoptotic cancer cell death in a variety of tumor models (169). Apoptotic pathways triggered by intrinsic factors such as DNA damage, endoplasmic reticulum stress, and LMP ultimately converge on mitochondrial membrane permeabilization (MOMP) and subsequent release of pro-apoptotic factors to the cytosol (166,(170)(171)(172). Mechanistic understanding of primary LMP in apoptosis was developed largely from studies using the lysosomotropic agent Leu-Leu-methyl ester (LLOMe) (173) and other apoptotic stimuli (153). ...
Chapter
Full-text available
ABSTRACT Breast cancer is a genetically and clinically heterogeneous disease with multiple subtypes. The classification of these subtypes has evolved over the years. The most common and widely accepted classification of breast cancer is from an immunohistochemical perspective, based on the expression of the following hormone receptors: estrogen (ER), progesterone (PR) and human epidermal growth factor (HER2). Accordingly, the following four subtypes of breast cancer are widely recognized: luminal A, luminal B, HER2-positive, and triple negative. With the recent advances in cancer research, and an increased molecular understanding of breast cancer, the current clinical model for classification of breast cancer may be benefit from the addition of several molecular markers such as miRNAs (let-7, miR-155, miR-150, miR-153) and mutations (p53, BRCA 1 and 2 genes). This chapter provides an overview of the characteristics of these four subtypes of breast cancer.
... Nitroxoline was later shown to reduce the expression of proteins associated with the epithelial-mesenchymal transition of tumour cells and the amount of myeloid-derived suppressor cells (MDSCs) in peripheral blood in bladder cancer (56). Furthermore, nitroxoline was identified in a second, independent study by high throughput compound library search and biochemical evaluation of the best hits as a potent, reversible and non-covalent inhibitor of the lysosomal cysteine peptidase cathepsin B (57), which plays an important role in the initiation and progression of cancer, its invasion and metastasis, and is known to be a promising target for the development of novel antitumour agents (58)(59)(60). The crystal structure of the nitroxoline-cathepsin B complex shows that nitroxoline binds to the active site cleft of cathepsin B and thereby selectively inhibits its endopeptidase activity at low micromolar concentrations (57). ...
Article
Full-text available
Rak je veliko breme za zdravstvene sisteme in še vedno drugi najpogostejši vzrok smrti v razvitem svetu. Zato se, kljub velikem napredku v zadnjih letih, še vedno veliko sredstev usmerja v iskanje novih učinkovitejših protitumorskih zdravil, ki bi omogočila bolj učinkovito zdravljenje, daljše preživetje bolnikov z rakom, preprečila ponovni pojav raka in zmanjšala stranske učinke, povezane z zdravljenjem raka, s tem pa povečala kakovost življenja bolnikov z rakom. Uvajanje novih zdravil v klinično uporabo pa je hkrati povezano z vse večjimi stroški. Zato je zaradi velike potrebe po prepoznavanju novih protitumorskih zdravil iskanje novih indikacij za že obstoječa zdravila na področju onkologije pritegnilo veliko pozornosti. Pri tem se je med drugim protitumorsko delovanje pokazalo tudi za nekatere antibiotike in druge protimikrobne učinkovine. Slednje se je med drugim izkazalo za doksiciklin, klorokin, nitroksolin in nekatere predstavnike fluorokinolonov. V preglednem članku povzemamo različne molekulske mehanizme, uporabljene tumorske modele in raziskave, ki opredeljujejo njihovo protitumorsko delovanje. To so prikazale številne neodvisne študije tako in vitro kot in vivo, strukture obstoječih učinkovin pa so se hkrati uporabile kot spojine za razvoj novih derivatov in identificiranje strukturnih elementov, ki izboljšajo protitumorsko delovanje. Te raziskave tako kažejo na nove možnosti za uporabo že dobro znanih učinkovin, ki bi lahko razširile svoj spekter uporabe in samostojno ali v kombinaciji z že obstoječim zdravljenjem izboljšale uspešnost protitumorskega zdravljenja.
... Lysosomal cathepsins also participate in regulating apoptosis [122,123]. Cathepsins B, L, S, H, and K, but not cathepsins C and X, act proapoptotically by activating Bid to tBid, which activates caspases [124], and antiapoptotically by cleaving and inactivating members of the Bcl-2 family [124][125][126][127]. Moreover, CatB suppresses alternative forms of cell death by cleaving the lipid signaling enzyme sphingosine kinase 1 [128] and degrading receptor-interacting Ser/Thr protein kinase 1 (RIPK1), consequently enforcing apoptosis [69,129]. ...
Article
Full-text available
Lysosomal peptidases are hydrolytic enzymes capable of digesting waste proteins that are targeted to lysosomes via endocytosis and autophagy. Besides intracellular protein catabolism, they play more specific roles in several other cellular processes and pathologies, either within lysosomes, upon secretion into the cell cytoplasm or extracellular space, or bound to the plasma membrane. In cancer, lysosomal peptidases are generally associated with disease progression, as they participate in crucial processes leading to changes in cell morphology, signaling, migration, and invasion, and finally metastasis. However, they can also enhance the mechanisms resulting in cancer regression, such as apoptosis of tumor cells or antitumor immune responses. Lysosomal peptidases have also been identified as hallmarks of aging and neurodegeneration, playing roles in oxidative stress, mitochondrial dysfunction, abnormal intercellular communication, dysregulated trafficking, and the deposition of protein aggregates in neuronal cells. Furthermore, deficiencies in lysosomal peptidases may result in other pathological states, such as lysosomal storage disease. The aim of this review is to highlight the role of lysosomal peptidases in particular pathological processes of cancer and neurodegeneration and to address the potential of lysosomal peptidases in diagnosing and treating patients.
Article
Full-text available
Papain-like cysteine proteases are composed of 11 human cysteine cathepsins, originally located in the lysosomes. They exhibit broad specificity and act as endopeptidases and/or exopeptidases. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Recently, cysteine cathepsins have been found to be present outside the lysosomes and often participate in various pathological processes. Hence, they have been considered key signalling molecules. Their potentially hazardous proteolytic activities are tightly regulated. This review aims to discuss recent advances in understanding the structural aspects of these four cathepsins, mechanisms of their zymogen activation, regulation of their activities, and functional aspects of these enzymes in neurodegeneration and cancer. Neurodegenerative effects have been evaluated, particularly in Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, multiple sclerosis, and neuropsychiatric disorders. Cysteine cathepsins also participate in tumour progression and metastasis through the overexpression and secretion of proteases, which trigger extracellular matrix degradation. To our knowledge, this is the first review to provide an in-depth analysis regarding the roles of cysteine cathepsins B, H, C, and X in neurodegenerative diseases and cancer. Further advances in understanding the functions of cysteine cathepsins in these conditions will result in the development of novel, targeted therapeutic strategies.
Article
Full-text available
TNF-alpha-induced apoptosis is thought to involve mediators from acidic vesicles. Cathepsin B (cat B), a lysosomal cysteine protease, has recently been implicated in apoptosis. To determine whether cat B contributes to TNF-alpha-induced apoptosis, we exposed mouse hepatocytes to the cytokine in vitro and in vivo. Isolated hepatocytes treated with TNF-alpha in the presence of the transcription inhibitor actinomycin D (AcD) accumulated cat B in their cytosol. Further experiments using cell-free systems indicated that caspase-8 caused release of active cat B from purified lysosomes and that cat B, in turn, increased cytosol-induced release of cytochrome c from mitochondria. Consistent with these observations, the ability of TNF-alpha/AcD to induce mitochondrial release of cytochrome c, caspase activation, and apoptosis of isolated hepatocytes was markedly diminished in cells from CatB(-/-) mice. Deletion of the CatB gene resulted in diminished liver injury and enhanced survival after treatment in vivo with TNF-alpha and an adenovirus construct expressing the IkappaB superrepressor. Collectively, these observations suggest that caspase-mediated release of cat B from lysosomes enhances mitochondrial release of cytochrome c and subsequent caspase activation in TNF-alpha-treated hepatocytes.
Article
Apoptosis--the regulated destruction of a cell--is a complicated process. The decision to die cannot be taken lightly, and the activity of many genes influence a cell's likelihood of activating its self-destruction programme. Once the decision is taken, proper execution of the apoptotic programme requires the coordinated activation and execution of multiple subprogrammes. Here I review the basic components of the death machinery, describe how they interact to regulate apoptosis in a coordinated manner, and discuss the main pathways that are used to activate cell death.
Article
Cathepsin B, a lysosomal cysteine proteinase, was detected within vesicles of cellular protrusions forming cell-cell contact sites between keratinocytes of the stratum spinosum of human skin. This observation suggested the possibility that secretion of the protease into the pericellular spaces could be involved in the dissociation of cell-cell contacts to enable intraepidermal keratinocyte migration. To determine whether cathepsin B is indeed secreted from migrating keratinocytes, we first used subconfluent HaCaT cells as a culture model to study spontaneous keratinocyte migration. A cathepsin B-specific fluorescent affinity label proved the association of mature cathepsin B with the surfaces of HaCaT cells at the leading edges of growing cells. Second, we used scratch-wounds of confluent HaCaT monolayers as a model of induced keratinocyte migration. Cathepsin B was detected within lysosomes, i.e. vesicles within the perinuclear region of non-wounded cells. Expression of cathepsin B was up-regulated and cathepsin B-positive vesicles showed a redistribution from perinuclear to peripheral regions of keratinocytes at the wound margins within 4 h after wounding. Enzyme cytochemistry further showed that cell surface-associated cathepsin B was proteolytically active at the leading fronts of migrating keratinocytes. In addition, increased amounts of mature forms of cathepsin B were detected within the conditioned media of HaCaT cells during the first 4 h after scratch-wounding. In contrast, and as a control, the activity of the cytosolic enzyme lactate dehydrogenase was not significantly higher in media of wounded cells as compared with non-wounded controls, arguing for a specific induction of cathepsin B secretion upon wounding and migration of the cells. This was further substantiated by applying various cathepsin B-specific inhibitors after wounding. These experiments showed that the migration ability of keratinocytes was reduced due to the blockage of functional cathepsin B. Thus, our results strongly suggest that cell surface-associated cathepsin B is a protease that contributes to the remodelling of the extracellular matrix and thereby promotes keratinocyte migration during wound healing.
Article
We have previously reported that the microtubule stabilizing agents (MSAs) paclitaxel, epothilone B and discodermolide induce caspase-independent cell death in non-small cell lung cancer (NSCLC) cells. Here we present two lines of evidence indicating a central role for the lysosomal protease cathepsin B in mediating cell death. First, inhibition of cathepsin B, and not of caspases or other proteases, such as cathepsin D or calpains, results in a strong protection against drug-induced cell death in several NSCLC cells. Second, MSAs trigger disruption of lysosomes and release and activation of cathepsin B. Interestingly, inhibition of cathepsin B prevents the appearance of multinucleated cells, an early characteristic of MSA-induced cell death, pointing to a central, proximal role for cathepsin B in this novel cell death pathway.
Article
Background: Malignant cell clones resistant to chemotherapy and radiotherapy frequently lead to treatment failure in patients with glioblastoma multiforme. Preliminary studies suggest that adding chloroquine to conventional therapy may improve treatment outcomes. Objective: To examine the effect of adding chloroquine to conventional therapy for glioblastoma multiforme. Design: Randomized, double-blind, placebo-controlled trial. Setting: National Institute of Neurology and Neurosurgery of Mexico. Patients: 30 patients with surgically confirmed glioblastoma confined to 1 cerebral hemisphere, with a Karnofsky performance score greater than 70, no comorbid disease, and age younger than 60 years. Interventions: Oral chloroquine at 150 mg/d for 12 months beginning on postoperative day 5 or placebo. All patients received conventional chemotherapy and radiotherapy. Measurements: Primary outcome was survival after surgery; surviving patients were followed up to October 2005. Periodic evaluation using the Karnofsky scale and imaging studies, as well as hematologic tests and ophthalmologic examinations, was done in all patients. Results: Median survival after surgery was 24 months for chloroquine-treated patients and 11 months for controls. At the end of the observation period, 6 patients treated with chloroquine had survived 59, 45, 30, 27, 27, and 20 months, respectively; 3 patients from the control group had survived 32, 25, and 22 months, respectively. Although not statistically significantly different, the rate of death with time was approximately half as large in patients receiving chloroquine as in patients receiving placebo (hazard ratio, 0.52 [95% Cl, 0.21 to 1.26]; P= 0.139). Limitations: The observed advantage of chloroquine may be due to chance; differences in pretreatment characteristics and conventional treatment regimens could not be adjusted for because of the small sample size. Conclusions: Chloroquine may improve mid-term survival when given in addition to conventional therapy for glioblastoma multiforme. These results suggest that larger, more definitive studies of chloroquine as adjuvant therapy for glioblastoma are warranted.
Article
The present study was undertaken to verify whether induction of senescence could be sufficient to reverse drug resistance and, if so, to determine the underlying mechanism(s). Our findings indicated that co-treatment of drug-resistant neuroblastoma cells with doxorubicin, at sublethal concentrations, in combination with the pan-caspase inhibitor, Q-VD-OPH, elicited a strong reduction of cell viability that occurred in a caspase-independent manner. This was accompanied by the appearance of a senescence phenotype, as evidenced by increased p21/WAF1 expression and senescence-associated β-galactosidase activity. Experiments using specific inhibitors of major cellular proteases other than caspases have shown that inhibition of cathepsin L, but not proteasome or cathepsin B, was responsible for the senescence-initiated reversal of drug resistance. This phenomenon appeared to be general because it was valid for other drugs and drug-resistant cell lines. A nonchemical approach, through cell transfection with cathepsin L small interfering RNA, also strongly reversed drug resistance. Further investigation of the underlying mechanism revealed that cathepsin L inhibition resulted in the alteration of intracellular drug distribution. In addition, in vitro experiments have demonstrated that p21/WAF1 is a substrate for cathepsin L, suggesting that inhibition of this enzyme may result in p21/WAF1 stabilization and its increased accumulation. All together, these findings suggest that cathepsin L inhibition in drug-resistant cells facilitates induction of senescence and reversal of drug resistance. This may represent the basis for a novel function of cathepsin L as a cell survival molecule responsible for initiation of resistance to chemotherapy.
Article
Background & Aims: During tumor necrosis factor α–mediated hepatocyte cytotoxicity, cathepsin B is released from lysosomes and contributes to apoptosis by indirectly promoting mitochondrial dysfunction. How this lysosomal pathway mediates mitochondrial dysfunction is unclear. Because Bcl-2 family proteins and caspase 2 have been implicated in proximal apoptosis-signaling pathways, we examined the role of these proteins in tumor necrosis factor α–induced lysosomal permeabilization and cathepsin B–mediated mitochondrial dysfunction. Methods: Studies were performed in primary hepatocytes from wild-type cathepsin B knockout, Bid knockout, and caspase 2 knockout mice and in the rat hepatoma cell line McArdle7777 by using tumor necrosis factor α/actinomycin D. Results: Studies in wild-type and Bid knockout hepatocytes showed that tumor necrosis factor α–mediated lysosomal permeabilization is Bid dependent. After tumor necrosis factor α/actinomycin D treatment, caspase 2 activity increased severalfold in wild-type hepatocytes, whereas minimal activity was observed in hepatocytes from cathepsin B knockout mice or in hepatoma cells treated with a cathepsin B inhibitor. In contrast, Bax was activated independently of cathepsin B. Pharmacological, genetic, or small interfering RNA–mediated inhibition of caspase 2 attenuated tumor necrosis factor α–mediated mitochondrial dysfunction, downstream caspase activation, and hepatocyte apoptosis. Conclusions: These data suggest that tumor necrosis factor α triggers Bid-dependent lysosomal permeabilization, followed by release of cathepsin B into the cytosol and activation of caspase 2. Caspase 2 then facilitates efficient mitochondrial cytochrome c release and apoptosis.
Article
The aberrant tumor microenvironment is increasingly appreciated as a key determinant of cancer progression, involving multiple cell types and functional interactions. Several matrix-degrading enzymes have been found to modulate regulatory networks governing tumor progression, and thus their inhibition has the potential to block multiple tumor-promoting signals and effector functions. We have previously shown that several cysteine cathepsins are upregulated in the RIP-Tag mouse model of pancreatic islet cell carcinogenesis, and that tumor progression is impaired following their collective pharmacologic inhibition. However, inhibiting an entire family of proteolytic enzymes has the additional potential to produce unwanted effects, including toxicity. As such, determining the critical target genes within implicated protease families is particularly relevant. Therefore, we have now taken a genetic approach to identify the individual cathepsin(s) that are critically important. Interestingly, not all cathepsins are functionally equivalent in promoting carcinogenesis. Mutants of cathepsins B or S impaired tumor formation and angiogenesis, while cathepsin B or L knockouts significantly retarded cell proliferation and tumor growth. Absence of any one of these three genes impaired tumor invasion. In contrast, removal of cathepsin C had no effect on either tumor formation or progression. We have identified E-cadherin as a target substrate of cathepsins B, L and S, but not cathepsin C, potentially explaining their differential effects on tumor invasion. Furthermore, we found analogous increases in cathepsin expression in human pancreatic endocrine neoplasms, and a significant association between levels of cathepsin B and L and tumor malignancy. These genetic experiments have revealed novel roles for individual cathepsins in tumor angiogenesis, invasion, apoptosis and cell proliferation, and indicate that there are important functional differences between cathepsin family members that would not have been predicted from biochemical analyses. In addition to increasing our understanding of how cathepsins specifically contribute to tumor-host interactions in the cancer microenvironment, these results may help guide the design of clinical trials aimed to assess cathepsin inhibitors as cancer therapies.