Early 35 kDa protein

Last updated
Early 35 kDa protein (AcMNPV)
P35 structure by Fisher 1999, PDB 1P35.jpg
P35 structure by Fisher et al. 1999 [1]
Identifiers
Organism Autographa californica nuclear polyhedrosis virus (AcMNPV)
SymbolP35
Entrez 1403968
PDB 1P35
RefSeq (mRNA) NC_001623.1
RefSeq (Prot) NP_054165.1
UniProt P08160
Other data
Chromosome 0: 0.12 - 0.12 Mb
Search for
Structures Swiss-model
Domains InterPro
Early 35 kDa protein (BmNPV)
Identifiers
Organism Bombyx mori nuclear polyhedrosis virus (BmNPV)
SymbolP35
Entrez 1488744
RefSeq (mRNA) NC_001962.1
RefSeq (Prot) NP_047533.1
UniProt P31354
Other data
Chromosome 0: 0.11 - 0.11 Mb
Search for
Structures Swiss-model
Domains InterPro

The Early 35 kDa protein, or P35 in short, is a baculoviral protein that inhibits apoptosis in the cells infected by the virus. Although baculoviruses infect only invertebrates in nature, ectopic expression of P35 in vertebrate animals and cells also results in inhibition of apoptosis, thus indicating a universal mechanism. P35 has been shown to be a caspase inhibitor with a very wide spectrum of activity both in regard to inhibited caspase types and to species in which the mechanism is conserved.

Contents

Species distribution

P35 has been found in different strains of the nuclear polyhedrosis virus, a species of baculovirus that infects insects. Two orthologs of P35 that have been studied in detail are the ones from the Autographa californica multicapsid nuclear polyhedrosis virus (AcMNPV) and from the Bombyx mori nuclear polyhedrosis virus (BmNPV). The P35 ortholog from AcMNPV was found to block apoptosis in mammalian cells much more efficiently as compared to the ortholog from BmNPV. [2]

Function

The P35 protein inhibits apoptosis by acting as a competitive, irreversible inhibitor of caspases. [3] P35 first serves as a caspase substrate and is cleaved between the amino acids D87 and G88, i.e. after the sequence DQMD in P35 from AcMNPV and after the sequence DKID in P35 from BmNPV, resulting in two polypeptide products of about 10 kDa and 25 kDa in size. [3] The cleavage site is situated in a solvent-exposed loop that extends from the protein's beta sheet core, thus ensuring good accessibility to the caspase. [1] [4] However, unlike other caspase substrate proteins, the fragments of P35 do not dissociate from the caspase after cleavage. Instead, the N-terminal, 10 kDa cleavage fragment remains bound to the caspase by a covalent, stable thioester bond between the cleavage residue D87 of P35 and the cysteine residue at the active site of the caspase. [5]

While the formation of a thioester intermediate between the aspartate of the substrate's recognition site and the cysteine of the caspase's active site is a normal event in caspase-mediated protein cleavage, the resulting bond is normally quickly hydrolysed so that the cleaved products can detach. In the case of P35, however, the caspase-substrate complex remains stable. Cleavage of P35 triggers rapid conformational changes that reposition its N-terminus, which is normally buried in the protein's beta-sheet core, to the caspase's active site. As a consequence of this rearrangement, the N-terminal P35 residues C2 and V3 interact with the caspase's active site to displace water and prevent the hydrolysis reaction. The P35 residue C2 competes with the caspase's active site cysteine residue for binding of the P35 residue D87, keeping the reaction trapped in an equilibrium state. [5] [6] [7] [8]

Interactions

In insect cells, P35 inhibits an enzyme called Sf caspase-1, which was identified as a structural and functional ortholog of human CASP3 (CPP32) and CASP7 (MCH3). [9] Studies using purified human caspases in vitro found that the protein is able to also inhibit several of these, including CASP1, CASP3, CASP6, CASP7, CASP8, and CASP10. [10]

Clinical significance

Since baculoviridae infect only insects and not humans, the function of P35 in the immune evasion of infected cells is not clinically relevant. However, P35 has been considered as a potential tool in gene therapy to suppress apoptosis where it is not wanted, such as in the protection of transplanted tissue from immune rejection or in the killing of bystander cells in cancer therapy; such methods are still far from clinical application though. [11]

History and discovery

The role of P35 in the inhibition of apoptosis was first described by Rollie J. Clem in the research group of Lois K. Miller at the Department of Genetics at the University of Georgia in 1991. [12] Four years later, in 1995, the reason for apoptosis inhibition by P35 was identified as its ability to bind and inhibit caspases (then still called ICE homologs) by Nancy J. Bump and co-workers at the BASF Bioresearch Corporation in Worcester, Massachusetts. [13] The mechanism of caspase inhibition was discovered by Guozhou Xu in the team of Hao Wu at the Department of Biochemistry at Weill Cornell Medical College in 2001. [5]

Related Research Articles

<span class="mw-page-title-main">Apoptosis</span> Programmed cell death in multicellular organisms

Apoptosis is a form of programmed cell death that occurs in multicellular organisms and in some eukaryotic, single-celled microorganisms such as yeast. Biochemical events lead to characteristic cell changes (morphology) and death. These changes include blebbing, cell shrinkage, nuclear fragmentation, chromatin condensation, DNA fragmentation, and mRNA decay. The average adult human loses between 50 and 70 billion cells each day due to apoptosis. For an average human child between eight and fourteen years old, each day the approximate lost is 20 to 30 billion cells.

<i>Baculoviridae</i> Family of viruses

Baculoviridae is a family of viruses. Arthropods, among the most studied being Lepidoptera, Hymenoptera and Diptera, serve as natural hosts. Currently, 85 species are placed in this family, assigned to four genera.

<span class="mw-page-title-main">Apoptosome</span>

The apoptosome is a large quaternary protein structure formed in the process of apoptosis. Its formation is triggered by the release of cytochrome c from the mitochondria in response to an internal (intrinsic) or external (extrinsic) cell death stimulus. Stimuli can vary from DNA damage and viral infection to developmental cues such as those leading to the degradation of a tadpole's tail.

<span class="mw-page-title-main">BH3 interacting-domain death agonist</span> Protein-coding gene in the species Homo sapiens

The BH3 interacting-domain death agonist, or BID, gene is a pro-apoptotic member of the Bcl-2 protein family. Bcl-2 family members share one or more of the four characteristic domains of homology entitled the Bcl-2 homology (BH) domains, and can form hetero- or homodimers. Bcl-2 proteins act as anti- or pro-apoptotic regulators that are involved in a wide variety of cellular activities.

<span class="mw-page-title-main">Caspase-9</span> Protein-coding gene in the species Homo sapiens

Caspase-9 is an enzyme that in humans is encoded by the CASP9 gene. It is an initiator caspase, critical to the apoptotic pathway found in many tissues. Caspase-9 homologs have been identified in all mammals for which they are known to exist, such as Mus musculus and Pan troglodytes.

<span class="mw-page-title-main">Caspase 8</span> Protein-coding gene in the species Homo sapiens

Caspase-8 is a caspase protein, encoded by the CASP8 gene. It most likely acts upon caspase-3. CASP8 orthologs have been identified in numerous mammals for which complete genome data are available. These unique orthologs are also present in birds.

Inhibitors of apoptosis are a group of proteins that mainly act on the intrinsic pathway that block programmed cell death, which can frequently lead to cancer or other effects for the cell if mutated or improperly regulated. Many of these inhibitors act to block caspases, a family of cysteine proteases that play an integral role in apoptosis. Some of these inhibitors include the Bcl-2 family, viral inhibitor crmA, and IAP's.

<span class="mw-page-title-main">Caspase 2</span> Protein-coding gene in the species Homo sapiens

Caspase 2 also known as CASP2 is an enzyme that, in humans, is encoded by the CASP2 gene. CASP2 orthologs have been identified in nearly all mammals for which complete genome data are available. Unique orthologs are also present in birds, lizards, lissamphibians, and teleosts.

<span class="mw-page-title-main">XIAP</span> Protein-coding gene in the species Homo sapiens

X-linked inhibitor of apoptosis protein (XIAP), also known as inhibitor of apoptosis protein 3 (IAP3) and baculoviral IAP repeat-containing protein 4 (BIRC4), is a protein that stops apoptotic cell death. In humans, this protein (XIAP) is produced by a gene named XIAP gene located on the X chromosome.

<span class="mw-page-title-main">Caspase 3</span> Protein-coding gene in the species Homo sapiens

Caspase-3 is a caspase protein that interacts with caspase-8 and caspase-9. It is encoded by the CASP3 gene. CASP3 orthologs have been identified in numerous mammals for which complete genome data are available. Unique orthologs are also present in birds, lizards, lissamphibians, and teleosts.

<span class="mw-page-title-main">Caspase 7</span> Protein-coding gene in the species Homo sapiens

Caspase-7, apoptosis-related cysteine peptidase, also known as CASP7, is a human protein encoded by the CASP7 gene. CASP7 orthologs have been identified in nearly all mammals for which complete genome data are available. Unique orthologs are also present in birds, lizards, lissamphibians, and teleosts.

<span class="mw-page-title-main">Caspase 6</span> Protein-coding gene in the species Homo sapiens

Caspase-6 is an enzyme that in humans is encoded by the CASP6 gene. CASP6 orthologs have been identified in numerous mammals for which complete genome data are available. Unique orthologs are also present in birds, lizards, lissamphibians, and teleosts. Caspase-6 has known functions in apoptosis, early immune response and neurodegeneration in Huntington's and Alzheimer's disease.

<span class="mw-page-title-main">Caspase 10</span> Protein-coding gene in the species Homo sapiens

Caspase-10 is an enzyme that, in humans, is encoded by the CASP10 gene.

<span class="mw-page-title-main">Diablo homolog</span> Protein-coding gene in the species Homo sapiens

Diablo homolog (DIABLO) is a mitochondrial protein that in humans is encoded by the DIABLO gene on chromosome 12. DIABLO is also referred to as second mitochondria-derived activator of caspases or SMAC. This protein binds inhibitor of apoptosis proteins (IAPs), thus freeing caspases to activate apoptosis. Due to its proapoptotic function, SMAC is implicated in a broad spectrum of tumors, and small molecule SMAC mimetics have been developed to enhance current cancer treatments.

<span class="mw-page-title-main">APAF1</span> Mammalian protein found in Homo sapiens

Apoptotic protease activating factor 1, also known as APAF1, is a human homolog of C. elegans CED-4 gene.

<span class="mw-page-title-main">HtrA serine peptidase 2</span> Enzyme found in humans

Serine protease HTRA2, mitochondrial is an enzyme that in humans is encoded by the HTRA2 gene. This protein is involved in caspase-dependent apoptosis and in Parkinson's disease.

<span class="mw-page-title-main">Caspase-activated DNase</span> Protein-coding gene in the species Homo sapiens

Caspase-activated DNase (CAD) or DNA fragmentation factor subunit beta is a protein that in humans is encoded by the DFFB gene. It breaks up the DNA during apoptosis and promotes cell differentiation. It is usually an inactive monomer inhibited by ICAD. This is cleaved before dimerization.

<span class="mw-page-title-main">Picornain 3C</span>

Picornain 3C is a protease found in picornaviruses, which cleaves peptide bonds of non-terminal sequences. Picornain 3C’s endopeptidase activity is primarily responsible for the catalytic process of selectively cleaving Gln-Gly bonds in the polyprotein of poliovirus and with substitution of Glu for Gln, and Ser or Thr for Gly in other picornaviruses. Picornain 3C are cysteine proteases related by amino acid sequence to trypsin-like serine proteases. Picornain 3C is encoded by enteroviruses, rhinoviruses, aphtoviruses and cardioviruses. These genera of picoviruses cause a wide range of infections in humans and mammals.

<span class="mw-page-title-main">Sf caspase-1</span>

The protein Sf caspase-1 is the insect ortholog of the human effector caspases CASP3 (CPP32) and CASP7 (MCH3) in the species Spodoptera frugiperda. It was identified as the target of the baculoviral caspase inhibitor protein P35, which it cleaves and by which it is inhibited. Like other caspases, Sf caspase-1 is an aspartate-specific cysteine protease that is produced as an inactive proenzyme and becomes activated by autocatalytic cleavage. The Sf caspase-1 proenzyme is cleaved after the amino acid residues Asp-28 and Asp-195, resulting in a smaller 12 kDa fragment and a larger 19 kDa fragment. Just like with human caspases CASP3 or CASP7, the two cleavage fragments form heterodimers, which again form biologically active dimers-of-heterodimers consisting of two smaller and two larger fragments. Some experiments also showed cleavage of Sf caspase-1 at the residue Asp-184, resulting in an 18 kDa instead of 19 kDa fragment, however this result is likely an in vitro artefact. The insect immunophilin FKBP46 is a substrate of Sf caspase-1, which cleaves full length FKBP46 resulting in a ~25 kDa fragment.

<span class="mw-page-title-main">Death regulator Nedd2-like caspase</span> Type of cysteine protease

Death regulator Nedd2-like caspase was firstly identified and characterised in Drosophila in 1999 as a cysteine protease containing an amino-terminal caspase recruitment domain. At first, it was thought of as an effector caspase involved in apoptosis, but subsequent findings have proved that it is, in fact, an initiator caspase with a crucial role in said type of programmed cell death.

References

  1. 1 2 Fisher AJ, Cruz W, Zoog SJ, Schneider CL, Friesen PD (April 1999). "Crystal structure of baculovirus P35: role of a novel reactive site loop in apoptotic caspase inhibition". The EMBO Journal. 18 (8): 2031–9. doi:10.1093/emboj/18.8.2031. PMC   1171287 . PMID   10205157.
  2. Morishima N, Okano K, Shibata T, Maeda S (May 1998). "Homologous p35 proteins of baculoviruses show distinctive anti-apoptotic activities which correlate with the apoptosis-inducing activity of each virus". FEBS Letters. 427 (1): 144–8. doi:10.1016/S0014-5793(98)00389-5. PMID   9613616. S2CID   29840073.
  3. 1 2 Bertin J, Mendrysa SM, LaCount DJ, Gaur S, Krebs JF, Armstrong RC, Tomaselli KJ, Friesen PD (September 1996). "Apoptotic suppression by baculovirus P35 involves cleavage by and inhibition of a virus-induced CED-3/ICE-like protease". Journal of Virology. 70 (9): 6251–9. doi:10.1128/jvi.70.9.6251-6259.1996. PMC   190650 . PMID   8709252.
  4. Zoog SJ, Bertin J, Friesen PD (September 1999). "Caspase inhibition by baculovirus P35 requires interaction between the reactive site loop and the beta-sheet core". The Journal of Biological Chemistry. 274 (37): 25995–6002. doi: 10.1074/jbc.274.37.25995 . PMID   10473544.
  5. 1 2 3 Xu G, Cirilli M, Huang Y, Rich RL, Myszka DG, Wu H (March 2001). "Covalent inhibition revealed by the crystal structure of the caspase-8/p35 complex". Nature. 410 (6827): 494–7. doi:10.1038/35068604. PMID   11260720. S2CID   279649.
  6. Riedl SJ, Renatus M, Snipas SJ, Salvesen GS (November 2001). "Mechanism-based inactivation of caspases by the apoptotic suppressor p35". Biochemistry. 40 (44): 13274–80. doi:10.1021/bi010574w. PMID   11683637.
  7. Xu G, Rich RL, Steegborn C, Min T, Huang Y, Myszka DG, Wu H (February 2003). "Mutational analyses of the p35-caspase interaction. A bowstring kinetic model of caspase inhibition by p35". The Journal of Biological Chemistry. 278 (7): 5455–61. doi: 10.1074/jbc.M211607200 . PMID   12458208.
  8. Lu M, Min T, Eliezer D, Wu H (February 2006). "Native chemical ligation in covalent caspase inhibition by p35". Chemistry & Biology. 13 (2): 117–22. doi:10.1016/j.chembiol.2005.12.007. PMID   16492559.
  9. Ahmad M, Srinivasula SM, Wang L, Litwack G, Fernandes-Alnemri T, Alnemri ES (January 1997). "Spodoptera frugiperda caspase-1, a novel insect death protease that cleaves the nuclear immunophilin FKBP46, is the target of the baculovirus antiapoptotic protein p35". The Journal of Biological Chemistry. 272 (3): 1421–4. doi: 10.1074/jbc.272.3.1421 . PMID   8999805.
  10. Zhou Q, Krebs JF, Snipas SJ, Price A, Alnemri ES, Tomaselli KJ, Salvesen GS (July 1998). "Interaction of the baculovirus anti-apoptotic protein p35 with caspases. Specificity, kinetics, and characterization of the caspase/p35 complex". Biochemistry. 37 (30): 10757–65. doi:10.1021/bi980893w. PMID   9692966.
  11. Doloff JC, Su T, Waxman DJ (September 2010). "Adenoviral delivery of pan-caspase inhibitor p35 enhances bystander killing by P450 gene-directed enzyme prodrug therapy using cyclophosphamide+". BMC Cancer. 10: 487. doi: 10.1186/1471-2407-10-487 . PMC   2946310 . PMID   20836875.
  12. Clem RJ, Fechheimer M, Miller LK (November 1991). "Prevention of apoptosis by a baculovirus gene during infection of insect cells". Science. 254 (5036): 1388–90. Bibcode:1991Sci...254.1388C. doi:10.1126/science.1962198. PMID   1962198.
  13. Bump NJ, Hackett M, Hugunin M, Seshagiri S, Brady K, Chen P, Ferenz C, Franklin S, Ghayur T, Li P (September 1995). "Inhibition of ICE family proteases by baculovirus antiapoptotic protein p35". Science. 269 (5232): 1885–8. Bibcode:1995Sci...269.1885B. doi:10.1126/science.7569933. PMID   7569933.