Caspase-8 and Cancer

 

Simone Fulda

University Children's Hospital, Eythstr.24, 89075 Ulm, Germany

To whom correspondence and reprint requests should be addressed:
Prof. Dr. Simone Fulda, University Children's Hospital,
Eythstr.24, D-89075 Ulm, Germany
Tel.: +49-731 50057034
Fax: +49-731 50057058
E-mail: simone.fulda@uniklinik-ulm.de

 

December 2009

 

 

Abstract

Caspase-8 belongs to the family of cysteine proteases that plays a critical role in the regulation of programmed cell death (apoptosis). For example, caspase-8 function is required for proper signaling via the death receptor (extrinsic) pathway. Accordingly, the disturbance of caspase-8 expression or function may contribute to cancer formation and progression. In addition, inactivation of caspase-8 may promote resistance to current treatment approaches, which critically require signaling via the death receptor pathway. Thus, restoration of caspase- 8 function presents a promising approach to overcome resistance to apoptosis in human cancers.


Introduction

Apoptosis (programmed cell death) is one of the most common forms of cell death in multicellular organisms and is a crucial component of normal development as well as a variety of physiological processes (Lockshin and Zakeri, 2007). This implies that too little or too much apoptosis can shift the tightly regulated balance between cell growth and cell death towards one or the other side and disturbs cellular homeostasis (Evan and Vousden, 2001). For example, a decreased rate of apoptosis favors tumor formation as well as its progression (Lowe and Lin, 2000). Accordingly, evasion of apoptosis can be found as a characteristic feature in most human malignancies (Hanahan and Weinberg, 2000). Over the last years key signaling pathways and molecules have been identified. For example, one of these critical regulators of apoptosis is caspase-8, a member of the caspase family of proteases (Degterev et al., 2003). Caspase-8 plays a central role in the transmission of the death signal in the death receptor (extrinsic) pathway of apoptosis by coupling the stimulation of death receptors to the activation of intracellular signaling cascades that eventually lead to cell death (Barnhart et al., 2003). Consequently, reduced expression or dysfunction of caspase-8 fosters carcinogenesis as well as the progression and treatment resistance in various different cancers (Fulda, 2008). This review focuses on the role of caspase-8 in cancer.


Apoptosis signaling pathways

There are two principle apoptosis signaling pathways: the death receptor (extrinsic) pathway and the mitochondrial (intrinsic) pathway (Fulda and Debatin, 2006). In the death receptor pathway, ligation of cell surface receptors of the tumor necrosis factor (TNF) receptor superfamily, including CD95 (APO-1/Fas) or TRAIL receptors, leads to the activation of caspase-8 in the death-inducing signaling complex (DISC) that contains besides caspase-8 and activated death receptors also the adaptor molecule FADD (Ashkenazi, 2008). Within the caspase family, caspase-8 is an initiator that following its activation can either directly cleave downstream effector caspases such as caspase-3 or, alternatively, can indirectly promote effector caspase activation via the cleavage of Bid (Ashkenazi, 2008). Bid is a pro-apoptotic protein of the Bcl-2 family with a BH3 domain only, which moves to mitochondria upon its cleavage to stimulate the mitochondrial signaling pathway (Adams and Cory, 2007). This involves the release of intermembrane space proteins such as cytochrome c and second mitochondria-derived activator of caspase (Smac)/direct IAP Binding protein with Low pI (DIABLO) from the mitochondria into the cytosol, which in turn triggers activation of caspase-3 (Kroemer et al., 2007). Once in the cytosol, cytochrome c causes caspase-3 activation via the formation of the apoptosome complex, a multimeric protein complex that contains Apaf-1 and caspase-9 besides cytochrome c (Kroemer et al., 2007). By comparison, activation of caspases is triggered by Smac/DIABLO via its opposing effect against IAPs (Kroemer et al., 2007).


Caspase-8: structure and function

Caspase-8 is one of the initiator caspases and belongs to the family of cysteine proteases (Degterev et al., 2003). As enzymes, caspases function as effector molecules of many forms of cell death and as such, play a central role in the execution phase of apoptosis (Degterev et al., 2003). In humans, the caspase-8 gene maps to chromosome 2q33 (Kischkel et al., 1998). From the structural point of view, caspase-8 is evolutionary highly conserved. For example, there is about 20% identity between mammalian caspase-8 and its homologue in the nematode c. elegans, which is the ced-3 protein (Degterev et al., 2003). Caspase-8 contains 480 amino acids and is a 55 kDa protein with two N-terminal death-effector domains (DED), which function as platforms for protein-protein interaction (Barnhart et al., 2003). The proteolytic domain is located at the C-terminus with a preferred substrate specificity of I/L/V/E X D, as caspase-8 belongs to the class of cysteine proteases (Degterev et al., 2003). Activation of caspase-8 results in the formation of an active heterotetramer of caspase-8 with two large and two small subunits (Degterev et al., 2003).

A number of different isoforms of caspase-8 with distinct functions can be distinguished. Isoforms caspase-8a and -8b are the pro-apoptotic variants that are most commonly expressed in mammalian cells (Scaffidi et al., 1997). Caspase-8 long (caspase-8L) is a splice variant, where a 136 bp insertion between exon 8 and exon 9 of full-length caspase-8 mRNA generates a premature stop codon via alternative splicing (Horiuchi et al., 2000; Himeji et al., 2002). This leads to the production of a truncated protein, which contains only the two Nterminal DED domains, but lacks the C-terminal protease domain (Horiuchi et al., 2000; Himeji et al., 2002). Accordingly, caspase-8L can be recruited into the DISC via its DED domains, but remains proteolytically inert due to absence of its protease domain (Miller et al., 2006; Mohr et al., 2005). Consequently, caspase-8L interferes in a dominant-negative manner with the transduction of the death signal from activated death receptors.

Besides its key role in the regulation of apoptosis, caspase-8 also has non-apoptotic properties, e.g. in the regulation of T cell homeostasis via control of IL-2 generation (Algeciras-Schimnich et al., 2002; Launay et al., 2005), in the control of differentiation and proliferation in the hematopoietic system (Algeciras-Schimnich et al., 2002; Launay et al., 2005) or in the regulation of adhesion, migration and metastasis in a complex manner (Stupack et al., 2006; Stupack and Cheresh, 2002; Senft et al., 2007; Helfer et al., 2006; Finlay and Vuori, 2007; Stegh et al., 2000). These data highlight the relevance of caspase-8 in various physiological contexts.


Caspase-8 is frequently inactivated in human cancers

Evasion of apoptosis, a hallmark of human cancers, can be caused by the inactivation of caspase-8 via multiple mechanisms. These comprise genetic alterations, epigenetic modifications, alternative splicing or posttranslational changes. Mutations of caspase-8 have been detected at relatively low frequency, for example in head and neck carcinoma or colorectal and gastric cancer (Kim et al., 2003; Mandruzzato et al., 1997; Soung et al., 2005). In its mutated form, caspase-8 interferes with the recruitment of wild-type caspase-8 to activated death receptors in a dominant-negative fashion (Kim et al., 2003; Mandruzzato et al., 1997). Additionally, homo- or heterozygous genomic deletions of caspase-8 as well as allelic imbalance on chromosome 2q associated with alterations of the caspase-8 gene have also been described, e.g. in neuroblastoma (Teitz et al., 2000; Takita et al., 2001).

As far as epigenetic mechanisms are concerned, silencing of caspase-8 expression by hypermethylation of regulatory sequences of the caspase-8 gene has been detected in multiple cancers, including several pediatric cancers such as neuroblastoma, medulloblastoma, Ewing tumor , retinoblastoma and rhabdomyosarcoma as well as glioblastoma or lung carcinoma (Teitz et al., 2000; Fulda et al., 2001; Hopkins-Donaldson et al., 2003; Pingoud-Meier et al., 2003; Harada et al., 2002; Hopkins-Donaldson et al., 2000; Iolascon et al., 2003; Takita et al., 2000; Grotzer et al., 2000; Shivapurkar et al., 2002a; Shivapurkar et al., 2002b). As discussed in more detail in the previous section, alternative splicing of caspase-8 can result in the production of caspase-8L as a dominant-negative splice variant, for example in leukemia and neuroblastoma (Miller et al., 2006; Mohr et al., 2005). Another mechanism of inactivation is caused by inhibitory phosphorylation on tyrosine 308 of caspase-8, e.g. via Src kinase (Cursi et al., 2006). This phosphorylation event may also promote cell migration via caspase-8 (Barbero et al., 2008). Recent evidence suggests that loss of caspase-8 favors cellular transformation, at least in in vitro models (Krelin et al., 2008).


Conclusions

Since caspase-8 presents a key regulator of apoptosis, inactivation of caspase-8 can confer resistance to cell death. Genetic, epigenetic as well as posttranslational changes can contribute to inactivation of caspase-8 in human malignancies. Thus, restoration of caspase-8 function presents a promising strategy to either directly trigger apoptosis in cancer cells or to restore sensitivity for apoptotic stimuli.


Acknowledgements

This work has been partially supported by grants from the Deutsche Forschungsgemeinschaft, Deutsche Krebshilfe, Bundesministerium für Forschung und Technologie, the European Community (ApopTrain, APO-SYS), and IAP6/18.

Bibliography

A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma.
Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen P.
J Exp Med. 1997 Aug 29;186(5):785-93.
PMID 9271594
 
FLICE is predominantly expressed as two functionally active isoforms, caspase-8/a and caspase-8/b.
Scaffidi C, Medema JP, Krammer PH, Peter ME.
J Biol Chem. 1997 Oct 24;272(43):26953-8.
PMID 9341131
 
Assignment of CASP8 to human chromosome band 2q33-->q34 and Casp8 to the murine syntenic region on chromosome 1B-proximal C by in situ hybridization.
Kischkel FC, Kioschis P, Weitz S, Poustka A, Lichter P, Krammer PH.
Cytogenet Cell Genet. 1998;82(1-2):95-6.
PMID 9763668
 
Resistance to TRAIL-induced apoptosis in primitive neuroectodermal brain tumor cells correlates with a loss of caspase-8 expression.
Grotzer MA, Eggert A, Zuzak TJ, Janss AJ, Marwaha S, Wiewrodt BR, Ikegaki N, Brodeur GM, Phillips PC.
Oncogene. 2000 Sep 21;19(40):4604-10.
PMID 11030149
 
The hallmarks of cancer.
Hanahan D, Weinberg RA.
Cell. 2000 Jan 7;100(1):57-70. (REVIEW)
PMID 10647931
 
Loss of caspase-8 expression in highly malignant human neuroblastoma cells correlates with resistance to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis.
Hopkins-Donaldson S, Bodmer JL, Bourloud KB, Brognara CB, Tschopp J, Gross N.
Cancer Res. 2000 Aug 15;60(16):4315-9.
PMID 10969767
 
Dominant expression of a novel splice variant of caspase-8 in human peripheral blood lymphocytes.
Horiuchi T, Himeji D, Tsukamoto H, Harashima S, Hashimura C, Hayashi K.
Biochem Biophys Res Commun. 2000 Jun 16;272(3):877-81.
PMID 10860845
 
Apoptosis in cancer.
Lowe SW, Lin AW.
Carcinogenesis. 2000 Mar;21(3):485-95. (REVIEW)
PMID 10688869
 
Identification of the cytolinker plectin as a major early in vivo substrate for caspase 8 during CD95- and tumor necrosis factor receptor-mediated apoptosis.
Stegh AH, Herrmann H, Lampel S, Weisenberger D, Andra K, Seper M, Wiche G, Krammer PH, Peter ME.
Mol Cell Biol. 2000 Aug;20(15):5665-79.
PMID 10891503
 
Absent or reduced expression of the caspase 8 gene occurs frequently in neuroblastoma, but not commonly in Ewing sarcoma or rhabdomyosarcoma.
Takita J, Yang HW, Bessho F, Hanada R, Yamamoto K, Kidd V, Teitz T, Wei T, Hayashi Y.
Med Pediatr Oncol. 2000 Dec;35(6):541-3.
PMID 11107112
 
Caspase 8 is deleted or silenced preferentially in childhood neuroblastomas with amplification of MYCN.
Teitz T, Wei T, Valentine MB, Vanin EF, Grenet J, Valentine VA, Behm FG, Look AT, Lahti JM, Kidd VJ.
Nat Med. 2000 May;6(5):529-35.
PMID 10802708
 
Proliferation, cell cycle and apoptosis in cancer.
Evan GI, Vousden KH.
Nature. 2001 May 17;411(6835):342-8. (REVIEW)
PMID 11357141
 
Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer.
Fulda S, Kufer MU, Meyer E, van Valen F, Dockhorn-Dworniczak B, Debatin KM.
Oncogene. 2001 Sep 13;20(41):5865-77.
PMID 11593392
 
Allelic imbalance on chromosome 2q and alterations of the caspase 8 gene in neuroblastoma.
Takita J, Yang HW, Chen YY, Hanada R, Yamamoto K, Teitz T, Kidd V, Hayashi Y.
Oncogene. 2001 Jul 19;20(32):4424-32.
PMID 11466626
 
Apoptosis-independent functions of killer caspases.
Algeciras-Schimnich A, Barnhart BC, Peter ME.
Curr Opin Cell Biol. 2002 Dec;14(6):721-6. (REVIEW)
PMID 12473345
 
Deregulation of caspase 8 and 10 expression in pediatric tumors and cell lines.
Harada K, Toyooka S, Shivapurkar N, Maitra A, Reddy JL, Matta H, Miyajima K, Timmons CF, Tomlinson GE, Mastrangelo D, Hay RJ, Chaudhary PM, Gazdar AF.
Cancer Res. 2002 Oct 15;62(20):5897-901.
PMID 12384554
 
Characterization of caspase-8L : a novel isoform of caspase-8 that behaves as an inhibitor of the caspase cascade.
Himeji D, Horiuchi T, Tsukamoto H, Hayashi K, Watanabe T, Harada M.
Blood. 2002 Jun 1;99(11):4070-8.
PMID 12010809
 
Loss of expression of death-inducing signaling complex (DISC) components in lung cancer cell lines and the influence of MYC amplification.
Shivapurkar N, Reddy J, Matta H, Sathyanarayana UG, Huang CX, Toyooka S, Minna JD, Chaudhary PM, Gazdar AF.
Oncogene. 2002a Dec 5;21(55):8510-4.
PMID 12466971
 
Differential inactivation of caspase-8 in lung cancers.
Shivapurkar N, Toyooka S, Eby MT, Huang CX, Sathyanarayana UG, Cunningham HT, Reddy JL, Brambilla E, Takahashi T, Minna JD, Chaudhary PM, Gazdar AF.
Cancer Biol Ther. 2002b Jan-Feb;1(1):65-9.
PMID 12170765
 
Get a ligand, get a life : integrins, signaling and cell survival.
Stupack DG, Cheresh DA.
J Cell Sci. 2002 Oct 1;115(Pt 19):3729-38. (REVIEW)
PMID 12235283
 
The death effector domain protein family.
Barnhart BC, Lee JC, Alappat EC, Peter ME.
Oncogene. 2003 Nov 24;22(53):8634-44.
PMID 14634625
 
A decade of caspases.
Degterev A, Boyce M, Yuan J.
Oncogene. 2003 Nov 24;22(53):8543-67. (REVIEW)
PMID 14634618
 
Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation.
Hopkins-Donaldson S, Ziegler A, Kurtz S, Bigosch C, Kandioler D, Ludwig C, Zangemeister-Wittke U, Stahel R.
Cell Death Differ. 2003 Mar;10(3):356-64.
PMID 12700635
 
Caspase 3 and 8 deficiency in human neuroblastoma.
Iolascon A, Borriello A, Giordani L, Cucciolla V, Moretti A, Monno F, Criniti V, Marzullo A, Criscuolo M, Ragione FD.
Cancer Genet Cytogenet. 2003 Oct 1;146(1):41-7.
PMID 14499695
 
Inactivating mutations of caspase-8 gene in colorectal carcinomas.
Kim HS, Lee JW, Soung YH, Park WS, Kim SY, Lee JH, Park JY, Cho YG, Kim CJ, Jeong SW, Nam SW, Kim SH, Lee JY, Yoo NJ, Lee SH.
Gastroenterology. 2003 Sep;125(3):708-15.
PMID 12949717
 
Loss of caspase-8 protein expression correlates with unfavorable survival outcome in childhood medulloblastoma.
Pingoud-Meier C, Lang D, Janss AJ, Rorke LB, Phillips PC, Shalaby T, Grotzer MA.
Clin Cancer Res. 2003 Dec 15;9(17):6401-9.
PMID 14695141
 
Vital functions for lethal caspases.
Launay S, Hermine O, Fontenay M, Kroemer G, Solary E, Garrido C.
Oncogene. 2005 Aug 4;24(33):5137-48. (REVIEW)
PMID 16079910
 
Caspase-8L expression protects CD34+ hematopoietic progenitor cells and leukemic cells from CD95-mediated apoptosis.
Mohr A, Zwacka RM, Jarmy G, Buneker C, Schrezenmeier H, Dohner K, Beltinger C, Wiesneth M, Debatin KM, Stahnke K.
Oncogene. 2005 Mar 31;24(14):2421-9.
PMID 15735742
 
CASPASE-8 gene is inactivated by somatic mutations in gastric carcinomas.
Soung YH, Lee JW, Kim SY, Jang J, Park YG, Park WS, Nam SW, Lee JY, Yoo NJ, Lee SH.
Cancer Res. 2005 Feb 1;65(3):815-21.
PMID 15705878
 
Src kinase phosphorylates Caspase-8 on Tyr380: a novel mechanism of apoptosis suppression.
Cursi S, Rufini A, Stagni V, Condo I, Matafora V, Bachi A, Bonifazi AP, Coppola L, Superti-Furga G, Testi R, Barila D.
EMBO J. 2006 May 3;25(9):1895-905. Epub 2006 Apr 13.
PMID 16619028
 
Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy.
Fulda S, Debatin KM.
Oncogene. 2006 Aug 7;25(34):4798-811. (REVIEW)
PMID 16892092
 
Caspase-8 promotes cell motility and calpain activity under nonapoptotic conditions.
Helfer B, Boswell BC, Finlay D, Cipres A, Vuori K, Bong Kang T, Wallach D, Dorfleutner A, Lahti JM, Flynn DC, Frisch SM.
Cancer Res. 2006 Apr 15;66(8):4273-8.
PMID 16618751
 
Caspase 8L, a novel inhibitory isoform of caspase 8, is associated with undifferentiated neuroblastoma.
Miller MA, Karacay B, Zhu X, O'Dorisio MS, Sandler AD.
Apoptosis. 2006 Jan;11(1):15-24.
PMID 16374545
 
Potentiation of neuroblastoma metastasis by loss of caspase-8.
Stupack DG, Teitz T, Potter MD, Mikolon D, Houghton PJ, Kidd VJ, Lahti JM, Cheresh DA.
Nature. 2006 Jan 5;439(7072):95-9.
PMID 16397500
 
The Bcl-2 apoptotic switch in cancer development and therapy.
Adams JM, Cory S.
Oncogene. 2007 Feb 26;26(9):1324-37. (REVIEW)
PMID 17322918
 
Novel noncatalytic role for caspase-8 in promoting SRC-mediated adhesion and Erk signaling in neuroblastoma cells.
Finlay D, Vuori K.
Cancer Res. 2007 Dec 15;67(24):11704-11.
PMID 18089800
 
Mitochondrial membrane permeabilization in cell death.
Kroemer G, Galluzzi L, Brenner C.
Physiol Rev. 2007 Jan;87(1):99-163. (REVIEW)
PMID 17237344
 
Cell death in health and disease.
Lockshin RA, Zakeri Z.
J Cell Mol Med. 2007 Nov-Dec;11(6):1214-24. Epub 2007 Nov 20. (REVIEW)
PMID 18031301
 
Caspase-8 interacts with the p85 subunit of phosphatidylinositol 3-kinase to regulate cell adhesion and motility.
Senft J, Helfer B, Frisch SM.
Cancer Res. 2007 Dec 15;67(24):11505-9.
PMID 18089778
 
Targeting the extrinsic apoptosis pathway in cancer.
Ashkenazi A.
Cytokine Growth Factor Rev. 2008 Jun-Aug;19(3-4):325-31. Epub 2008 May 20. (REVIEW)
PMID 18495520
 
Identification of a critical tyrosine residue in caspase 8 that promotes cell migration.
Barbero S, Barila D, Mielgo A, Stagni V, Clair K, Stupack D.
J Biol Chem. 2008 May 9;283(19):13031-4. Epub 2008 Jan 23.
PMID 18216014
 
Caspase-8.
Fulda S.
Schwab M, editor. Encyclopedia of Cancer. Berlin: Springer. 2008.
 
Caspase-8 deficiency facilitates cellular transformation in vitro.
Krelin Y, Zhang L, Kang TB, Appel E, Kovalenko A, Wallach D.
Cell Death Differ. 2008 Sep;15(9):1350-5. Epub 2008 Jun 20.
PMID 18566604
 
Written2009-12Simone Fulda
Children's Hospital, Eythstr.24, D-89075 Ulm, Germany

Citation

This paper should be referenced as such :
Fulda, S
Caspase-8, Cancer
Atlas Genet Cytogenet Oncol Haematol. 2010;14(9):894-897.
Free journal version : [ pdf ]   [ DOI ]
On line version : http://AtlasGeneticsOncology.org/Deep/Casp8inCancerID20080.htm