Elsevier

Biochimie

Volume 92, Issue 11, November 2010, Pages 1635-1643
Biochimie

Review
Pathophysiological functions of cathepsin D: Targeting its catalytic activity versus its protein binding activity?

https://doi.org/10.1016/j.biochi.2010.05.009Get rights and content

Abstract

The lysosomal aspartic protease cathepsin D (cath-D) is overexpressed and hyper-secreted by epithelial breast cancer cells. This protease is an independent marker of poor prognosis in breast cancer as it is correlated with the incidence of clinical metastasis. In normal cells, cath-D is localized in intracellular vesicles (lysosomes and endosomes). In cancer cells, overexpressed cath-D accumulates in cells, where it may affect their degradative capacities, and the pro-enzyme is hyper-secreted in the tumor micro-environment. In addition, during apoptosis, lysosomal cath-D is released into the cytosol, where it may interact with and/or cleave pro-apoptotic, anti-apoptotic, or nuclear proteins. Several studies have shown that cath-D affects various different steps in tumor progression and metastasis. Cath-D stimulates cancer cell growth in an autocrine manner, and also cath-D plays a crucial paracrine role in the tumor micro-environment by stimulating fibroblast outgrowth and tumor angiogenesis. A mutant D231N-cath-D, which is devoid of catalytic activity, remained mitogenic, indicating an additional action of cath-D by protein–protein interaction. Targeting cath-D in cancer may require the use of inhibitors of its catalytic activity, but also the development of new tools to inhibit its protein binding functions. Thus, elucidation of the mechanism of action of cath-D is crucial if an appropriate strategy is to be developed to target this protease in cancer. The discovery of new physiological substrates of cath-D using proteomic approaches can be expected to generate new critical targets. The aim of this review is to describe the roles of the cath-D protease in cancer progression and metastasis, as well as its function in apoptosis, and to discuss how it can be targeted in cancer by inhibiting its proteolytic activity and/or its binding protein activity.

Introduction

Proteases irreversibly hydrolyze the peptide bond in proteins, which results in an important and irreversible post-translational modification. The human genome encodes over 569 proteolytic enzymes or homologs, which constitute the second largest class of human enzymes. Proteases are assigned to five classes on the basis of the active site residue that executes the nucleophilic attack on the target peptide bond: aspartic, cysteine, serine, threonine and metalloproteinases. These proteases are implicated in normal physiological processes, but deregulation of their expression and/or enzyme activity in disorders such as cancer has profound consequences.

Different families of proteases have been implicated in motility, invasion, extravasation, proliferation and metastasis: the serine proteases (uPA, uPAR and PAI1) [1], the metalloproteinase family [2], the cysteine cathepsins (cathepsin-B and cathepsin-L) [3], and the aspartic cathepsin D (cath-D) [4], respectively. Cathepsin proteases are lysosomal hydrolases that degrade proteins at acidic pH in the lysosomes, or extracellularly in the matrix. Cathepsins can be divided into three subgroups, based on their active site amino acid (i.e., cysteine (B, C, H, F, K, L, O, S, V, X, and W), aspartate (D and E), or serine (A and G) cathepsins). The possible involvement of cysteine and aspartic cathepsins in cancer has been the subject of more debate than that of metalloproteases and serine proteases. This might result from the assumption that only secreted proteases that are proteolytically active at neutral pH play an active role in cancer, whereas cathepsins, which require a more acidic pH to be proteolytically active, are thought to be likely to play only a minor role. However, it has been demonstrated that cathepsins are hyper-secreted in cancer, and cath-B and cath-D have been described as being associated with the cell surface [5], [6]. Recent studies using transgenic mouse models have stimulated fresh interest in the fundamental roles of cathepsins in cancer [7], [8], [9], [10]. Although historically studies have tended to focus on the role of lysosomal proteases within the endocytic and lysosomal compartments, recent discoveries have shown that these proteases play a critical role in other intracellular compartments, such as the cytosol [11] or the nucleus [12], and within the extracellular milieu in the tumoral stroma [13]. It has become clear that their pattern of expression and their substrate specificities are more complex than was originally envisaged. In cancer, lysosomal proteases are overexpressed and their cellular localizations are profoundly altered, leading to major changes in their targets and consequently in their biological activities.

In addition, cathepsins, metalloproteases and serine proteases act in a cascade-like manner and as part of a proteolytic pathway rather than simply functioning individually [14]. Elucidating the cascade of enzymatic activities that contribute to overall proteolysis during carcinogenesis may identify rate-limiting steps or pathways that could be targeted by anti-cancer treatments [14]. The proteolytic cascade of activation of the different classes of proteases strongly suggests that anti-cancer strategies intended to target several classes of proteases simultaneously might be more promising than those that target a single protease or class of proteases.

Recent studies have focused on extracellular proteases as primary targets for drug discovery, because of their differential expression in many pathophysiological processes, including cancer, cardiovascular conditions, and inflammatory, pulmonary, and periodontal diseases [3], [15]. Interestingly, new extracellular inhibitors of metalloproteases, serine proteases and cysteine proteases are currently under clinical investigation [15]. The aim of this review is to present the role of the cath-D protease in cancer progression and metastasis, as well as its function in apoptosis, and to discuss how it could be targeted in cancer by inhibiting its proteolytic activity and/or its protein binding activity.

Section snippets

Structure and trafficking of cath-D

Cath-D [E.C. 3.4.23.5] is a ubiquitous, lysosomal, aspartic endo-proteinase that requires an acidic pH to be proteolytically active (Fig. 1). The human cath-D gene contains 9 exons, and is located on chromosome 11p15 [16]. During its transportation to lysosomes, the 52-kDa human pro-cath-D is proteolytically processed to form a 48-kDa, single-chain, intermediate which is an active enzyme located in the endosomes. Further proteolytic processing yields the mature active lysosomal protease, which

The tight control of cath-D expression and catalytic function is fundamental in normal cells

During fetal development, the level of cath-D increases gradually in all tissues, suggesting a gradual maturation of the lysosomal system [34]. A reduction of cath-D expression or catalytic activity leads to devastating neurodegenerative disorders. Cath-D knockout mice die shortly after birth, and display a neuronal accumulation of ceroid lipofuscin, accompanied by neurodegeneration in the retina and central nervous system, and the accumulation of autophagic vacuoles [35], [36], [37], [38].

Function of cath-D in apoptosis

Cysteine and aspartic cathepsins play key roles in tumor cell death via the mediation of apoptosis [4], [11], [49], [50], [51]. The function of cath-D in apoptosis needs further investigation, since this protease has both anti-apoptotic and pro-apoptotic functions.

Cath-D is an independent marker of a poor prognosis in breast cancer

In the 1990s, several independent clinical studies showed that the cath-D level in primary breast cancer cytosols is an independent prognostic parameter correlated with the incidence of clinical metastasis and shorter survival times [76]. A meta-analysis of studies on node-negative breast cancer [77], as well as a complete study of 2810 patients in Rotterdam [78], indicate that high concentrations of cath-D are an effective marker of aggressiveness. Cath-D is now recognized as an independent

Targeting cath-D in cancer

Cathepsins have long been known to play an important role in the progression and metastasis of cancer. Cath-D stimulates cancer cell proliferation, fibroblast outgrowth, tumor angiogenesis, and metastasis. In cancer cells, overexpressed cath-D accumulates in cells where it may affect their degradation capacities, and the pro-enzyme is hyper-secreted in the tumor micro-environment (Fig. 2). Therefore, inhibiting cath-D action requires the development of inhibitors targeting extracellular cath-D,

Conclusion

Cath-D is a key protease that affects many fundamental functions in cells. The molecular mechanism by which cath-D affects cancer progression remains largely unknown. Furthermore, we still do not have any specific cath-D inhibitors that could be used to target its action in cancer. Since this protease may also act by protein–protein interaction, it will be crucial to identify its partners in order to develop inhibitors to block its protein binding function.

Acknowledgement

Grant sponsors ‘ANR Jeunes chercheurs Jeunes chercheuses’ ANR-05-JCJC-0215-01, and EU FP6; Grant number: LSHC-CT-2007-037665.

References (169)

  • M. Canuel et al.

    Sortilin mediates the lysosomal targeting of cathepsins D and H

    Biochem. Biophys. Res. Commun.

    (2008)
  • M. Koike et al.

    Involvement of two different cell death pathways in retinal atrophy of cathepsin D-deficient mice

    Mol. Cell Neurosci.

    (2003)
  • L. Myllykangas et al.

    Cathepsin D-deficient Drosophila recapitulate the key features of neuronal ceroid lipofuscinoses

    Neurobiol. Dis.

    (2005)
  • T. Awano et al.

    A mutation in the cathepsin D gene (CTSD) in American Bulldogs with neuronal ceroid lipofuscinosis

    Mol. Genet. Metab.

    (2006)
  • R. Steinfeld et al.

    Cathepsin D deficiency is associated with a human neurodegenerative disorder

    Am. J. Hum. Genet.

    (2006)
  • J. Tyynela et al.

    Congenital ovine neuronal ceroid lipofuscinosis–a cathepsin D deficiency with increased levels of the inactive enzyme

    Eur. J. Paediatr. Neurol.

    (2001)
  • D. Hilfiker-Kleiner et al.

    A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy

    Cell

    (2007)
  • A.M. Sheikh et al.

    Cathepsin D and apoptosis related proteins are elevated in the brain of autistic subjects

    Neuroscience

    (2010)
  • O. Vasiljeva et al.

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

    Biochimie

    (2008)
  • K. Ollinger

    Inhibition of cathepsin D prevents free-radical-induced apoptosis in rat cardiomyocytes

    Arch. Biochem. Biophys.

    (2000)
  • K. Takuma et al.

    Roles of cathepsins in reperfusion-induced apoptosis in cultured astrocytes

    Neurochem. Int.

    (2003)
  • K. Roberg et al.

    Microinjection of cathepsin d induces caspase-dependent apoptosis in fibroblasts

    Am. J. Pathol.

    (2002)
  • K. Roberg et al.

    Lysosomal release of cathepsin D precedes relocation of cytochrome c and loss of mitochondrial transmembrane potential during apoptosis induced by oxidative stress

    Free Radic. Biol. Med.

    (1999)
  • N. Bidere et al.

    Cathepsin D triggers Bax activation, resulting in selective apoptosis-inducing factor (AIF) relocation in T lymphocytes entering the early commitment phase to apoptosis

    J. Biol. Chem.

    (2003)
  • M. Shibata et al.

    Participation of cathepsins B and D in apoptosis of PC12 cells following serum deprivation

    Biochem. Biophys. Res. Commun.

    (1998)
  • S. Greco et al.

    Co-expression of thymidine kinase and cathepsin D in 200 primary breast carcinomas

    Cancer Lett.

    (2000)
  • J.T. Anim et al.

    Relationship between the expression of various markers and prognostic factors in breast cancer

    Acta Histochem.

    (2005)
  • P. Roger et al.

    Cathepsin D immunostaining in paraffin-embedded breast cancer cells and macrophages: correlation with cytosolic assay

    Hum. Pathol.

    (1994)
  • V. Vetvicka et al.

    Effect of human procathepsin D on proliferation of human cell lines

    Cancer Lett.

    (1994)
  • H.W. Smith et al.

    Regulation of cell signalling by uPAR

    Nat. Rev. Mol. Cell Biol.

    (2010)
  • R. Roy et al.

    Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer

    J. Clin. Oncol.

    (2009)
  • M.M. Mohamed et al.

    Cysteine cathepsins: multifunctional enzymes in cancer

    Nat. Rev. Cancer

    (2006)
  • J.A. Joyce et al.

    Multiple roles for cysteine cathepsins in cancer

    Cell Cycle

    (2004)
  • V. Gocheva et al.

    Cysteine cathepsins and the cutting edge of cancer invasion

    Cell Cycle

    (2007)
  • V. Gocheva et al.

    Distinct roles for cysteine cathepsin genes in multistage tumorigenesis

    Genes Dev.

    (2006)
  • N.I. Affara et al.

    Delineating protease functions during cancer development

    Methods Mol. Biol.

    (2009)
  • M. Cudic et al.

    Extracellular proteases as targets for drug development

    Curr. Protein Pept. Sci.

    (2009)
  • P. Augereau et al.

    Cloning and sequencing of the 52K cathepsin D complementary deoxyribonucleic acid of MCF7 breast cancer cells and mapping on chromosome 11

    Mol. Endocrinol.

    (1988)
  • A.M. Samarel et al.

    Effects of cysteine protease inhibitors on rabbit cathepsin D maturation

    Am. J. Physiol.

    (1989)
  • V. Laurent-Matha et al.

    Processing of human cathepsin D is independent of its catalytic function and auto-activation: involvement of cathepsins L and B

    J. Biochem.

    (2006)
  • U. Felbor et al.

    Neuronal loss and brain atrophy in mice lacking cathepsins B and L

    Proc. Natl. Acad. Sci. U S A

    (2002)
  • A. Wille et al.

    Cathepsin L is involved in cathepsin D processing and regulation of apoptosis in A549 human lung epithelial cells

    Biol. Chem.

    (2004)
  • P. Metcalf et al.

    Two crystal structures for cathepsin D: the lysosomal targeting signal and active site

    EMBO J.

    (1993)
  • E.T. Baldwin et al.

    Crystal structures of native and inhibited forms of human cathepsin D: implications for lysosomal targeting and drug design

    Proc. Natl. Acad. Sci. U S A

    (1993)
  • N.E. Goldfarb et al.

    Electrostatic switches that mediate the pH-dependent conformational change of “short” recombinant human pseudocathepsin D

    Biochemistry

    (2005)
  • A.Y. Lee et al.

    Conformational switching in an aspartic proteinase

    Nat. Struct. Biol.

    (1998)
  • V. Cavailles et al.

    Estrogens and growth factors induce the mRNA of the 52K-pro-cathepsin-D secreted by breast cancer cells

    Nucleic Acids Res.

    (1988)
  • C. Giamarchi et al.

    Chromatin structure of the regulatory regions of pS2 and cathepsin D genes in hormone-dependent and -independent breast cancer cell lines

    Oncogene

    (1999)
  • F. Capony et al.

    Increased secretion, altered processing, and glycosylation of pro-cathepsin D in human mammary cancer cells

    Cancer Res.

    (1989)
  • R. Castino et al.

    Prolactin promotes the secretion of active cathepsin D at the basal side of rat mammary acini

    Endocrinology

    (2008)
  • Cited by (85)

    • Cancer – Proteases in Progression and Metastasis

      2022, Encyclopedia of Cell Biology: Volume 1-6, Second Edition
    View all citing articles on Scopus
    View full text