Elsevier

Biochimie

Volume 190, November 2021, Pages 91-110
Biochimie

Contribution of Apaf-1 to the pathogenesis of cancer and neurodegenerative diseases

https://doi.org/10.1016/j.biochi.2021.07.004Get rights and content

Highlights

  • Alteration in Apaf-1 level is frequently observed in many human cancers.

  • The contribution of Apaf-1 to pathology of neurodegenerative diseases is not as clear.

  • Designing screening assays of apoptosome assembly/disassembly will be useful in finding novel therapeutic agents.

  • More attention should be made to the contribution of Apaf-1 isoforms to the above mentioned pathologies.

Abstract

Deregulation of apoptosis is associated with various pathologies, such as neurodegenerative disorders at one end of the spectrum and cancer at the other end. Generally speaking, differentiated cells like cardiomyocytes, skeletal myocytes and neurons exhibit low levels of Apaf-1 (Apoptotic protease activating factor 1) protein suggesting that down-regulation of Apaf-1 is an important event contributing to the resistance of these cells to apoptosis. Nonetheless, upregulation of Apaf-1 has not emerged as a common phenomenon in pathologies associated with enhanced neuronal cell death, i.e., neurodegenerative diseases. In cancer, on the other hand, Apaf-1 downregulation is a common phenomenon, which occurs through various mechanisms including mRNA hyper-methylation, gene methylation, Apaf-1 localization in lipid rafts, inhibition by microRNAs, phosphorylation, and interaction with specific inhibitors. Due to the diversity of these mechanisms and involvement of other factors, defining the exact contribution of Apaf-1 to the development of cancer in general and neurodegenerative disorders, in particular, is complicated. The current review is an attempt to provide a comprehensive image of Apaf-1's contribution to the pathologies observed in cancer and neurodegenerative diseases with the emphasis on the therapeutic aspects of Apaf-1 as an important target in these pathologies.

Introduction

Apoptosis is a well-organized active process of cell death playing a central role in the development and homeostasis of multicellular organisms. Apoptosis is characterized by morphological hallmarks including cellular shrinkage, chromatin condensation, membrane blebbing, nuclear fragmentation, and apoptotic body formation engulfed by phagocytic cells [1]. Apoptosis is induced by extrinsic and intrinsic stimuli and as such is classified into the receptor and mitochondrial-mediated pathways, also known as the extrinsic and the intrinsic pathway, respectively [2]. In both pathways, all morphological changes result from the action of cysteine aspartyl proteases, known as caspases. These enzymes are synthesized as inactive procaspases and stored in the cytosol, suggesting the requirement for a fast response should the need arise. The two major classes of apoptotic caspases are initiator caspases of caspases-2, -8, -9, -10 and effector caspases of caspases-3, -6, -7. All initiator caspases require a specific complex for activation. Caspase-2, the most highly conserved caspase across all species, is activated by a large protein complex termed the PIDDosome in response to a range of different stresses including heat, metabolic disruption, and DNA damage [[3], [4], [5], [6]]. Caspase-2 has been shown to mediate and amplify mitochondria-dependent apoptosis via cleaving BID, a Bcl-2 family protein with a BH3 domain only [7]. The truncated BID acts by both enhancing channel formation by Bax and Bak to release cytochrome c from the mitochondria and neutralizing antiapoptotic Bcl-2 proteins [8]. Caspase-9 is the initiator caspase of the intrinsic pathway or the mitochondrial pathway, while the caspases of −8 and −10 are the initiator caspases of the extrinsic or receptor-mediated pathways. These pathways merge at the effector caspases of caspase-3 and to some extent caspase-7. Proteolytic processing of the effector procaspases by the initiator caspases leads to their activation. These events are followed by the cleavage of vital cellular proteins at specific sites, culminating in cell death [9]. Procaspase-6 acts as one of many substrates of caspase-3 contributing to the apoptotic events in the nucleus by cleaving nuclear mitotic apparatus protein (NuMA) and Lamin A [10]. The activation of the initiator caspase-9 is achieved through a specific complex named apoptosome. Unlike caspase-9, the activation of caspase-8 and -10 is achieved through the formation of various complexes whose main component is a distinct subset of the tumor necrosis factor receptor (TNFR) superfamily members characterized by the presence of death domain (DD) [11]. Diversity of TNFR, their interacting ligands, and intracellular regulatory elements create many complexes with various signaling outcomes. The formation of death inducing signaling complexes, DISCs, is associated with the activation of mostly caspase-8 and in some instances caspase-10 leading to apoptosis. Other complexes can also form, the most notable of which is the 2-megadalton ripoptosome complex. Once again, various ripoptosomes can form with RIP kinase 1 acting as a major common component. Homo association of caspase-8 in the complex leads to apoptosis, while its hetero association with a caspase-8 homolog FLIPL, which lacks active site cysteine, prevents cell death. Necroptosis can also occur if caspase-8 heterodimerizes with the shorter form of FLIP [12]. Moreover, caspase-8 participates in inflammation through a complex known as FADDosome. FADDosome is a complex of caspase-8, FADD and RIP1 which are assembled in response to Tumor Necrosis Factor (TNF)-related apoptosis-inducing ligand (TRAIL) and leads to NF-κB-dependent expression of pro-inflammatory cytokines and chemokines. Caspase-8 appears to be a scaffold for the formation of this complex, because the activity of this complex is not dependent on the enzymatic activity of caspase-8 [13]. On other hand, it has been reported that FADDosome is an apoptosis-inducing complex in cancer cells via caspase-8 activation, which occurs in response to DNA single-strand breaks. It has been proposed that the presence of caspase-10 is essential for FADDosome assembly and caspase-8 activity in this complex [14].

Apoptosome is a mega-complex initially formed by cytochrome c, Apoptotic protease activating factor 1 (Apaf-1), (d)ATP and procaspase-9 assembly, although other proteins like X-linked inhibitor of apoptosis protein (XIAP) and caspase-3 have also been found in association with the complex. Apaf-1 is present in a locked and inactive monomeric form in the cytosol. In response to intracellular stresses such as hypoxia, growth factor deprivation, cell detachment and stress signals, such as drug-induced DNA damage, cytochrome c is released from the mitochondrial intermembrane space into the cytosol. Interaction of the cytochrome c with the WD40 domain of Apaf-1 activates the protein leading to the formation of the Apaf-1 apoptosome platform. This heptameric complex recruits procaspase-9 through a homotypic CARD-CARD association forming the apoptosome complex. Caspase-9 is a holoenzyme requiring the formation of the apoptosome complex for its activation. Activated procaspase-9 cleaves itself at D315, which in turn activates effector caspases (Fig. 1) [15]. The exact number of procaspase-9 molecules per apoptosome complex is a matter of debate but it is generally believed to be between 2 and 5 molecules per seven Apaf-1 molecules in the apoptosome depending upon the condition [[16], [17], [18], [19], [20]]. The apoptosome is also responsible for amplifying extrinsic cell death pathways through the truncation of BID, a process similar to the above-discussed mechanism for caspase-2 [21].

Deregulation of apoptosis is associated with various pathologies, such as neurodegenerative disorders and cancer. At one end of the spectrum lies inadequate apoptosis seen in cancer and in the other end lies excessive apoptosis occurring in neurodegenerative diseases [22]. Interestingly, a gradual decrease in Apaf-1 expression occurs during brain development, seemingly rendering neurons resistant to apoptosis [23]. Generally speaking, differentiated cells like cardiomyocytes, skeletal myocytes and neurons exhibit low levels of Apaf-1 protein [[24], [25], [26]]. Consequently, down-regulation of Apaf-1, as a key molecule in the intrinsic pathway of apoptosis, appears to be the strategy of choice to prevent apoptosis in non-dividing cells, which is accomplished through various mechanisms such as endogenous inhibitors, microRNAs and epigenetic alterations. In this context, it is important to examine whether or not Apaf-1 upregulation is a mechanism behind enhanced cell death in neurodegenerative diseases, and its downregulation acts as a mechanism for the reduced cell death in cancers. Pinpointing the extent of Apaf-1 participation in apoptosis dysregulation in cancer in general and neurodegenerative disorders in particular is complicated. The current review is an attempt to provide a comprehensive image of Apaf-1's contribution to these pathologies and shed light on the therapeutic aspects of Apaf-1 as an important target in these diseases.

Section snippets

Regulation of Apaf-1 activity

Under physiological conditions, the intrinsic apoptotic pathway is tightly regulated by a number of proteins and small molecules. The endogenous regulators of the apoptosome can affect the intrinsic pathway at various stages during the apoptosome formation (Table 1 and Fig. 2). The first line of these regulators function through mitochondrial permeabilization leading to cytochrome c release, as a critical stage in apoptosome formation and caspase activation in mammalian cells. A number of

Apaf-1 in pathological states

Apoptosis is a critical process for the homeostasis of multicellular organisms. Deregulation of the intrinsic apoptosis pathway is observed in various pathologic conditions. Excessive apoptosis occurs in stroke, ischemia-reperfusion damage and degenerative diseases, and conversely, deficient or inadequate apoptosis is linked to cancer and autoimmune diseases [64]. Given that Apaf-1 is a central molecule in the activation of the intrinsic apoptotic pathway, deregulation of Apaf-1 is inevitably

Apaf-1 as a therapeutic target

The critical role of Apaf-1, as a key molecule in the intrinsic apoptosis pathway, as well as its involvement in several severe pathological states including cancer and neurodegenerative diseases, make this protein a valuable therapeutic target for the development of novel strategies to fight cancer and neurodegenerative diseases [168]. In the following sections, we introduce some chemical activators and inhibitors of Apaf-1 or apoptosome relevant to cancer and neurodegenerative conditions.

Conclusion

Apaf-1 is one of the key molecules in the intrinsic pathway of apoptosis, which in response to various intracellular stresses, is activated upon cytochrome c release from the mitochondria. Heptameric Apaf-1 platform formed due to cytochrome c interaction recruits procaspase-9 molecules leading to the activation and auto-processing of this initiator caspase. Under physiological conditions, the non-dividing cells such as neurons and cardio-myocytes have reduced Apaf-1 expression/activity compared

Declaration of competing interest

The authors report no declarations of interest.

Acknowledgments

We thank the Universities of Tehran and Kurdistan for the support of this manuscript.

References (220)

  • T. Takatani et al.

    Taurine prevents the ischemia-induced apoptosis in cultured neonatal rat cardiomyocytes through Akt/caspase-9 pathway

    Biochem. Biophys. Res. Commun.

    (2004)
  • Y. Huang et al.

    Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain

    Cell

    (2001)
  • S. Karimpour et al.

    Integrity of ATP binding site is essential for effective inhibition of the intrinsic apoptosis pathway by NAIP

    Biochem. Biophys. Res. Commun.

    (2011)
  • J. Davoodi et al.

    Neuronal apoptosis inhibitory protein, NAIP, is an inhibitor of procaspase-9

    Int. J. Biochem. Cell Biol.

    (2010)
  • J. Davoodi et al.

    Neuronal apoptosis-inhibitory protein does not interact with Smac and requires ATP to bind caspase-9

    J. Biol. Chem.

    (2004)
  • C. Du et al.

    Smac, a mitochondrial protein that promotes cytochrome c–dependent caspase activation by eliminating IAP inhibition

    Cell

    (2000)
  • B.A. Abhari et al.

    A mechanistic insight into SMAC peptide interference with XIAP-Bir2 inhibition of executioner caspases

    J. Mol. Biol.

    (2008)
  • G. Vinothini et al.

    Mitochondria-mediated apoptosis in patients with adenocarcinoma of the breast: correlation with histological grade and menopausal status

    Breast

    (2011)
  • K. Lauber et al.

    The adapter protein Apaf-1 is proteolytically processed during apoptosis

    J. Biol. Chem.

    (2001)
  • B.B. Wolf et al.

    Defective cytochrome c-dependent caspase activation in ovarian cancer cell lines due to diminished or absent apoptotic protease activating factor-1 activity

    J. Biol. Chem.

    (2001)
  • A.E. Abdelrahman et al.

    Prognostic impact of Apaf-1, Cyclin D1, and AQP-5 in serous ovarian carcinoma treated with the first-line chemotherapy

    Ann. Diagn. Pathol.

    (2018)
  • P. Kamarajan et al.

    Apaf-1 overexpression partially overcomes apoptotic resistance in a cisplatin-selected HeLa cell line

    FEBS Lett.

    (2001)
  • C. Leo et al.

    Expression of Apaf-1 in cervical cancer correlates with lymph node metastasis but not with intratumoral hypoxia

    Gynecol. Oncol.

    (2005)
  • T. Ogawa et al.

    APAF-1-ALT, a novel alternative splicing form of APAF-1, potentially causes impeded ability of undergoing DNA damage-induced apoptosis in the LNCaP human prostate cancer cell line

    Biochem. Biophys. Res. Commun.

    (2003)
  • N. Honarpour et al.

    Adult Apaf-1-deficient mice exhibit male infertility

    Dev. Biol.

    (2000)
  • F. Christoph et al.

    Frequent epigenetic inactivation of p53 target genes in seminomatous and nonseminomatous germ cell tumors

    Canc. Lett.

    (2007)
  • J.F. Kerr et al.

    Apoptosis: a basic biological phenomenon with wideranging implications in tissue kinetics

    Br. J. Canc.

    (1972)
  • A. Tinel et al.

    The PIDDosome, a protein complex implicated in activation of caspase-2 in response to genotoxic stress

    Science

    (2004)
  • S. Kumar et al.

    Induction of apoptosis by the mouse Nedd2 gene, which encodes a protein similar to the product of the Caenorhabditis elegans cell death gene ced-3 and the mammalian IL-1 beta-converting enzyme

    Gene Dev.

    (1994)
  • S. Kumar

    Caspase 2 in apoptosis, the DNA damage response and tumour suppression: enigma no more?

    Nat. Rev. Canc.

    (2009)
  • L. Bouchier-Hayes et al.

    Real Time with Caspase-2

    (2010)
  • P. Lassus et al.

    Requirement for caspase-2 in stress-induced apoptosis before mitochondrial permeabilization

    Science

    (2002)
  • M.C. Wei et al.

    tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c

    Gene Dev.

    (2000)
  • P. Fuentes-Prior et al.

    The protein structures that shape caspase activity, specificity, activation and inhibition

    Biochem. J.

    (2004)
  • H. Hirata et al.

    Caspases are activated in a branched protease cascade and control distinct downstream processes in Fas-induced apoptosis

    J. Exp. Med.

    (1998)
  • C.M. Henry et al.

    Caspase-8 acts in a non-enzymatic role as a scaffold for assembly of a pro-inflammatory “FADDosome” complex upon TRAIL stimulation

    Mol. Cell

    (2017)
  • A. Mohr et al.

    Caspase-10: a molecular switch from cell-autonomous apoptosis to communal cell death in response to chemotherapeutic drug treatment

    Cell Death Differ.

    (2018)
  • L. Dorstyn et al.

    New insights into apoptosome structure and function

    Cell Death Differ.

    (2018)
  • S. Malladi et al.

    The Apaf-1• procaspase-9 apoptosome complex functions as a proteolytic-based molecular timer

    EMBO J.

    (2009)
  • Q. Hu et al.

    Molecular determinants of caspase-9 activation by the Apaf-1 apoptosome

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2014)
  • T.C. Cheng et al.

    A near atomic structure of the active human apoptosome

    Elife

    (2016)
  • P. Klus et al.

    Neurodegeneration and cancer: where the disorder prevails

    Sci. Rep.

    (2015)
  • A. Fortin et al.

    APAF1 is a key transcriptional target for p53 in the regulation of neuronal cell death

    J. Cell Biol.

    (2001)
  • D. Sanchis et al.

    Lack of Apaf-1 expression confers resistance to cytochrome c-driven apoptosis in cardiomyocytes

    Cell Death Differ.

    (2003)
  • C.E. Johnson et al.

    Differential Apaf-1 levels allow cytochrome c to induce apoptosis in brain tumors but not in normal neural tissues

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2007)
  • A.G. Yakovlev et al.

    Differential expression of apoptotic protease-activating factor-1 and caspase-3 genes and susceptibility to apoptosis during brain development and after traumatic brain injury

    J. Neurosci.

    (2001)
  • S. Heimer et al.

    Raptinal bypasses BAX, BAK, and BOK for mitochondrial outer membrane permeabilization and intrinsic apoptosis

    Cell Death Dis.

    (2019)
  • R. Singh et al.

    Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins

    Nat. Rev. Mol. Cell Biol.

    (2019)
  • S. Banjara et al.

    The Bcl-2 family: ancient origins, conserved structures, and divergent mechanisms

    Biomolecules

    (2020)
  • C.F. Warren et al.

    BCL-2 family isoforms in apoptosis and cancer

    Cell Death Dis.

    (2019)
  • Cited by (11)

    View all citing articles on Scopus
    View full text