Skip to main content
Log in

Proteochemometrics modeling for prediction of the interactions between caspase isoforms and their inhibitors

  • Original Article
  • Published:
Molecular Diversity Aims and scope Submit manuscript

Abstract

Caspases (cysteine-aspartic proteases) play critical roles in inflammation and the programming of cell death in the form of necroptosis, apoptosis, and pyroptosis. The name of these enzymes has been chosen in accordance with their cysteine protease activity. They act as cysteines in nucleophilically active sites to attack and cleave target proteins in the aspartic acid and amino acid C-terminal. Based on the substrate’s structure and the specificity, the physiological activity of caspases is divided. However, in apoptosis, the division of caspases into initiating caspases (caspase 2, 8, 9, and 10) and executive caspases (caspase 3, 6, and 7) is essential. The present study aimed to perform Proteochemometrics Modeling to generalize the data on caspases, which could predict ligand and protein interactions. In this study, we employed protein and ligand descriptors. Moreover, protein descriptors were computed using the Protr R package, while PADEL-Descriptor was employed for the computation of ligand descriptors. In addition, NCA (Neighborhood Component Analyses) was used for descriptor selection, and SVR, decision tree, and ensemble methods were utilized for the proteochemometrics modeling. This study shows that the ensemble model demonstrates superior performance compared with other models in terms of R2, Q2, and RMSE criteria.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Lamkanfi M, Dixit VM (2014) Mechanisms and functions of inflammasomes. Cell 157(5):1013–1022. https://doi.org/10.1016/j.cell.2014.04.007

    Article  CAS  Google Scholar 

  2. Man SM, Kanneganti TD (2016) Converging roles of caspases in inflammasome activation, cell death and innate immunity. Nat Rev Immunol 16(1):7. https://doi.org/10.1038/nri.2015.7

    Article  CAS  Google Scholar 

  3. Fuchs Y, Steller H (2015) Live to die another way: modes of programmed cell death and the signals emanating from dying cells. Nat Rev Mol Cell Biol 16(6):329–344. https://doi.org/10.1038/nrm3999

    Article  CAS  Google Scholar 

  4. Hoste E, Kemperman P, Devos M, Denecker G, Kezic S, Yau N, Declercq W (2011) Caspase-14 is required for filaggrin degradation to natural moisturizing factors in the skin. J Investig Dermatol 131(11):2233–2241. https://doi.org/10.1038/jid.2011.153

    Article  CAS  Google Scholar 

  5. Stowe I, Lee B, Kayagaki N (2015) Caspase-11: arming the guards against bacterial infection. Immunol Rev 265(1):75–84. https://doi.org/10.1111/imr.12292

    Article  CAS  Google Scholar 

  6. Fuentes-Prior P, Salvesen GS (2004) The protein structures that shape caspase activity, specificity, activation and inhibition. Biochem J 384(2):201–232. https://doi.org/10.1042/BJ20041142

    Article  CAS  Google Scholar 

  7. Shalini S, Dorstyn L, Dawar S, Kumar S (2015) Old, new and emerging functions of caspases. Cell Death Differ 22(4):526–539. https://doi.org/10.1038/cdd.2014.216

    Article  CAS  Google Scholar 

  8. Sanchez I, Xu CJ, Juo P, Kakizaka A, Blenis J, Yuan J (1999) Caspase-8 is required for cell death induced by expanded polyglutamine repeats. Neuron 22(3):623–633. https://doi.org/10.1016/S0896-6273(00)80716-3

    Article  CAS  Google Scholar 

  9. Squitieri F, Maglione V, Orobello S, Fornai F (2011) Genotype-, aging-dependent abnormal caspase activity in Huntington disease blood cells. J Neural Transm 118(11):1599–1607. https://doi.org/10.1007/s00702-011-0646-1

    Article  CAS  Google Scholar 

  10. Narula J, Pandey P, Arbustini E, Haider N, Narula N, Kolodgie FD, Kharbanda S (1999) Apoptosis in heart failure: release of cytochrome c from mitochondria and activation of caspase-3 in human cardiomyopathy. Proc Natl Acad Sci 96(14):8144–8149. https://doi.org/10.1073/pnas.96.14.8144

    Article  CAS  Google Scholar 

  11. McIlwain DR, Berger T, Mak TW (2013) Caspase functions in cell death and disease. Cold Spring Harb Perspect Biol 5(4):a008656. https://doi.org/10.1101/cshperspect.a026716

    Article  Google Scholar 

  12. Yordanov V, Dimitrov I, Doytchinova I (2017) Proteochemometrics for the prediction of binding to the MHC proteins. Lett Drug Des Discov 14(1):2–9. https://doi.org/10.2174/1570180813666160630122341

    Article  CAS  Google Scholar 

  13. Lapinsh M, Prusis P, Gutcaits A, Lundstedt T, Wikberg JE (2001) Development of proteo-chemometrics: a novel technology for the analysis of drug-receptor interactions. Biochim Biophys Acta (BBA)-Gen Subj 1525(1–2):180–190. https://doi.org/10.1016/S0304-4165(00)00187-25

    Article  CAS  Google Scholar 

  14. Gottfries J, Eriksson L (2010) Extensions to amino acid description. Mol Divers 14(4):709–718. https://doi.org/10.1007/s11030-009-9204-2

    Article  CAS  Google Scholar 

  15. Sheikhpour R, Gharaghani S, Nazarshodeh E (2020) Sparse feature selection in multi-target modeling of carbonic anhydrase isoforms by exploiting shared information among multiple targets. Chemom Intell Lab Syst 200:104000. https://doi.org/10.1016/j.chemolab.2020.104000

    Article  CAS  Google Scholar 

  16. Lapinsh M, Prusis P, Mutule I, Mutulis F, Wikberg JE (2003) QSAR and proteo-chemometric analysis of the interaction of a series of organic compounds with melanocortin receptor subtypes. J Med Chem 46(13):2572–2579. https://doi.org/10.1021/jm020945m

    Article  CAS  Google Scholar 

  17. Bessonnet S, El Mansori M, Mezghani S, Coniglio N, Pee R, Pinault S (2020) Features selection approaches for an objective control of cosmetic quality of coated surfaces. Surf Topogr Metrol Prop 8(2):024007. https://doi.org/10.1088/2051-627x/ab8500

    Article  Google Scholar 

  18. Parks C, Gaieb Z, Amaro RE (2020) An analysis of proteochemometric and conformal prediction machine learning protein- ligand binding affinity models. Front Mol Biosci 7:93. https://doi.org/10.3389/fmolb.2020.00093

    Article  CAS  Google Scholar 

  19. Scapin G, Patel D, Arnold E (eds) (2015) Multifaceted roles of crystallography in modern drug discovery. Springer, Cham

    Google Scholar 

  20. Liu T, Lin Y, Wen X, Jorissen RN, Gilson MK (2007) BindingDB: a web-accessible database of experimentally determined protein–ligand binding affinities. Nucleic Acids Res 35(suppl_1):D198–D201. https://doi.org/10.1093/nar/gkl999

  21. Berman HM (2008) The protein data bank: a historical perspective. Acta Crystallogr A. https://doi.org/10.1107/S0108767307035623

    Article  Google Scholar 

  22. O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR (2011) Open Babel: An open chemical toolbox. J Cheminform. https://doi.org/10.1186/1758-2946-3-33

  23. Xiao N, Cao DS, Zhu MF, Xu QS (2015) protr/ProtrWeb: R package and web server for generating various numerical representation schemes of protein sequences. Bioinformatics 31(11):1857–1859. https://doi.org/10.1093/bioinformatics/btv042

    Article  CAS  Google Scholar 

  24. Yap CW (2011) PaDEL-descriptor: an open source software to calculate molecular descriptors and fingerprints. J Comput Chem 32(7):1466–1474. https://doi.org/10.1002/jcc.21707

    Article  CAS  Google Scholar 

  25. Chandrashekar G, Sahin F (2014) A survey on feature selection methods. Comput Electr Eng 40(1):16–28. https://doi.org/10.1016/j.compeleceng.2013.11.024

    Article  Google Scholar 

  26. Malan NS, Sharma S (2019) Feature selection using regularized neighbourhood component analysis to enhance the classification performance of motor imagery signals. Comput Biol Med 107:118–126. https://doi.org/10.1016/j.compbiomed.2019.02.009

    Article  Google Scholar 

  27. Kavya N, Sriraam N, Usha N, Sharath D, Hiremath B, Menaka M, Venkatraman B (2020) Feature selection using neighborhood component analysis with support vector machine for classification of breast mammograms. International conference on communication, computing and electronics systems. Springer, Singapore, pp 253–260

    Chapter  Google Scholar 

  28. Ren J, Qiu Z, Fan W, Cheng H, Yu PS (2008) Forward semi-supervised feature selection. Pacific-Asia conference on knowledge discovery and data mining. Springer, Berlin, Heidelberg, pp 970–976

    Chapter  Google Scholar 

  29. Giblin KA, Hughes SJ, Boyd H, Hansson P, Bender A (2018) Prospectively validated proteochemometric models for the prediction of small-molecule binding to bromodomain proteins. J Chem Inf Model 58(9):1870–1888. https://doi.org/10.1021/acs.jcim.8b00400

    Article  CAS  Google Scholar 

  30. Tresadern G, Trabanco AA, Pérez-Benito L, Overington JP, van Vlijmen HW, van Westen GJ (2017) Identification of allosteric modulators of metabotropic glutamate 7 receptor using proteochemometric modeling. J Chem Inf Model 57(12):2976–2985. https://doi.org/10.1021/acs.jcim.7b00338

    Article  CAS  Google Scholar 

  31. Shao J (1993) Linear model selection by cross-validation. J Am Stat Assoc 88(422):486–494. https://doi.org/10.1080/01621459.1993.10476299

    Article  Google Scholar 

  32. Berrar D (2019) Cross-validation. Encycl Bioinf Comput Biol 1:542–545. https://doi.org/10.1016/B978-0-12-809633-8.20349-X

    Article  Google Scholar 

  33. Rasti B, Mazraedoost S, Panahi H, Falahati M, Attar F (2019) New insights into the selective inhibition of the β-carbonic anhydrases of pathogenic bacteria Burkholderia pseudomallei and Francisella tularensis: a proteochemometrics study. Mol Divers 23(2):263–273. https://doi.org/10.1007/s11030-018-9869-5

    Article  CAS  Google Scholar 

  34. Boser BE, Guyon IM, Vapnik VN (1992) A training algorithm for optimal margin classifiers. In Proceedings of the 5th Annual ACM Workshop on Computational Learning Theory (pp. 144–152). Pennsylvania, Pittsburgh, USA

  35. Xu Z, Gao Y, Jin Y (2014) Application of an optimized SVR model of machine learning. Int J Multimed Ubiquitous Eng 9(6):67–80. https://doi.org/10.1155/2016/6212951

    Article  Google Scholar 

  36. Kisi O, Cimen M (2012) Precipitation forecasting by using wavelet-support vector machine conjunction model. Eng Appl Artif Intell 25(4):783–792. https://doi.org/10.1016/j.engappai.2011.11.003

    Article  Google Scholar 

  37. Caputo B, Sim K, Furesjo F, Smola A (2002) Appearance-based object recognition using SVMs: which kernel should I use?. In: Proc of NIPS workshop on Statistical methods for computational experiments in visual processing and computer vision, Whistler Vol. 2002

  38. Podgorelec V, Kokol P, Stiglic B, Rozman I (2002) Decision trees: an overview and their use in medicine. J Med Syst 26(5):445–463. https://doi.org/10.1023/A:1016409317640

    Article  Google Scholar 

  39. Kalyan S, François A (2018) 5. Freeing the comparative method from the tree model: a framework for historical glottometry. Senri Ethnol Stud 98:59–89. https://doi.org/10.15021/00009005

    Article  Google Scholar 

  40. Hansen LK, Salamon P (1990) Neural network ensembles. IEEE Trans Pattern Anal Mach Intell 12(10):993–1001. https://doi.org/10.1109/34.58871

    Article  Google Scholar 

  41. Wichard JD, Ogorzalek M (2004) Time series prediction with ensemble models. In 2004 IEEE international joint conference on neural networks (IEEE Cat. No. 04CH37541) 2:1625–1630 IEEE. https://doi.org/10.1109/IJCNN.2004.1380203

  42. Kashaw SK, Agarwal S, Mishra M, Sau S, Iyer AK (2019) Molecular docking analysis of caspase-3 activators as potential anticancer agents. Curr Comput-Aided Drug Des 15(1):55–66. https://doi.org/10.2174/1573409914666181015150731

    Article  CAS  Google Scholar 

  43. Golbraikh A, Tropsha A (2000) Predictive QSAR modeling based on diversity sampling of experimental datasets for the training and test set selection. Mol Diver 5(4):231–243. https://doi.org/10.1023/a:1020869118689

    Article  CAS  Google Scholar 

  44. Wang F, Zhou B (2020) Insight into structural requirements of ACE inhibitory dipeptides: QSAR and molecular docking studies. Mol Diver 24(4):957–969. https://doi.org/10.1007/s11030-019-10005-0

    Article  CAS  Google Scholar 

  45. Hajimahdi Z, Safizadeh F, Zarghi A (2016) QSAR analysis for some 1, 2-benzisothiazol-3-one derivatives as caspase-3 inhibitors by stepwise MLR method. Iran J Pharm Res IJPR 15(2):439. https://doi.org/10.22037/IJPR.2016.1855

    Article  CAS  Google Scholar 

  46. Agyapong O, Miller WA, Wilson MD, Kwofie SK (2021) Development of a proteochemometric-based support vector machine model for predicting bioactive molecules of tubulin receptors. Mol Divers 1–12. https://doi.org/10.1007/s11030-021-10329-w

  47. Vaidya A, Jain AK, Kumar BP, Sastry GN, Kashaw SK, Agrawal RK (2017) CoMFA, CoMSIA, kNN MFA and docking studies of 1, 2, 4-oxadiazole derivatives as potent caspase-3 activators. Arab J Chem 10:S3936–S3946. https://doi.org/10.1016/j.arabjc.2014.05.034

    Article  CAS  Google Scholar 

  48. Sathya B (2018) Hologram based QSAR analysis of caspase-3 inhibitors. J Chosun Nat Sci 11(2):93–100. https://doi.org/10.13160/ricns.2018.11.2.93

    Article  Google Scholar 

  49. Sharma S, Basu A, Agrawal RK (2013) Pharmacophore modeling and docking studies on some nonpeptide-based caspase-3 inhibitors. Biomed Res Int. https://doi.org/10.1155/2013/306081

    Article  Google Scholar 

  50. Nazarshodeh E, Sheikhpour R, Gharaghani S, Sarram MA (2018) A novel proteochemometrics model for predicting the inhibition of nine carbonic anhydrase isoforms based on supervised Laplacian score and k-nearest neighbour regression. SAR QSAR Environ Res 29(6):419–437. https://doi.org/10.1080/1062936X.2018.1447995

    Article  CAS  Google Scholar 

  51. Tropsha A, Gramatica P, Gombar VK (2003) The importance of being earnest: validation is the absolute essential for successful application and interpretation of QSPR models. QSAR Comb Sci 22(1):69–77. https://doi.org/10.1002/qsar.200390007

    Article  CAS  Google Scholar 

  52. Wikberg JE, Spjuth O, Eklund M, Lapins M (2012) Chemoinformatics taking biology into account: proteochemometrics. Computational approaches in cheminformatics and bioinformatics. Wiley, Hoboken, pp 57–92

    Google Scholar 

  53. Prusis P, Uhlén S, Petrovska R, Lapinsh M, Wikberg JE (2006) Prediction of indirect interactions in proteins. BMC Bioinf 7(1):1–13. https://doi.org/10.1186/1471-2105-7-167

    Article  CAS  Google Scholar 

  54. Kontijevskis A, Petrovska R, Mutule I, Uhlen S, Komorowski J, Prusis P, Wikberg JE (2007) Proteochemometric analysis of small cyclic peptides’ interaction with wild-type and chimeric melanocortin receptors. Proteins Struct Funct Bioinf 69(1):83–96. https://doi.org/10.1007/s10822-014-9809-0

    Article  CAS  Google Scholar 

  55. Lapinsh M, Veiksina S, Uhlén S, Petrovska R, Mutule I, Mutulis F, Wikberg JE (2005) Proteochemometric mapping of the interaction of organic compounds with melanocortin receptor subtypes. Mol Pharmacol 67(1):50–59. https://doi.org/10.1124/mol.104.002857

    Article  CAS  Google Scholar 

  56. Lapinsh M, Prusis P, Petrovska R, Uhlén S, Mutule I, Veiksina S, Wikberg JE (2007) Proteochemometric modeling reveals the interaction site for Trp9 modified α-MSH peptides in melanocortin receptors. Proteins Struct Funct Bioinf 67(3):653–660. https://doi.org/10.1002/prot.21323

    Article  CAS  Google Scholar 

  57. Dinkova-Kostova AT, Cory AH, Bozak RE, Hicks RJ, Cory JG (2007) Bis (2-hydroxybenzylidene) acetone, a potent inducer of the phase 2 response, causes apoptosis in mouse leukemia cells through a p53-independent, caspase-mediated pathway. Cancer Lett 245(1–2):341–349. https://doi.org/10.1016/j.canlet.2006.01.024

    Article  CAS  Google Scholar 

  58. Kim H, Moon JY, Burapan S, Han J, Cho SK (2018) Induction of ER stress-mediated apoptosis by the major component 5, 7, 4′-trimethoxyflavone isolated from kaempferia parviflora tea infusion. Nutr Cancer 70(6):984–996. https://doi.org/10.1080/01635581.2018.1491607

    Article  CAS  Google Scholar 

  59. Garcia-Calvo M, Peterson EP, Leiting B, Ruel R, Nicholson DW, Thornberry NA (1998) Inhibition of human caspases by peptide-based and macromolecular inhibitors. J Biol Chem 273(49):32608–32613. https://doi.org/10.1070/jbc.273.49.32608

    Article  CAS  Google Scholar 

  60. Eriksson A, Hermanson M, Wickström M, Lindhagen E, Ekholm C, Jensen AJ, Höglund M (2012) The novel tyrosine kinase inhibitor AKN-028 has significant antileukemic activity in cell lines and primary cultures of acute myeloid leukemia. Blood Cancer J 2(8):e81–e88. https://doi.org/10.1038/bcj.2012.28

    Article  CAS  Google Scholar 

  61. Li J, Bombeck CA, Yang S, Kim YM, Billiar TR (1999) Nitric oxide suppresses apoptosis via interrupting caspase activation and mitochondrial dysfunction in cultured hepatocytes. J Biol Chem 274(24):17325–17333. https://doi.org/10.1074/jbc.274.24.17325

    Article  CAS  Google Scholar 

  62. Paunel-Görgülü A, Lögters T, Flohé S, Cinatl J, Altrichter J, Windolf J, Scholz M (2011) Stimulation of Fas signaling down-regulates activity of neutrophils from major trauma patients with SIRS. Immunobiology 216(3):334–342. https://doi.org/10.1016/j.imbio.2010.07.005

    Article  CAS  Google Scholar 

  63. Shin HJ, Kim HL, Kim SJ, Chung WS, Kim SS, Um JY (2011) Inhibitory effects of chelidonic acid on IL-6 production by blocking NF-κB and caspase-1 in HMC-1 cells. Immunopharmacol Immunotoxicol 33(4):614–619. https://doi.org/10.3109/08923973.2011.552508

    Article  CAS  Google Scholar 

  64. Xu Z, Chen X, Fu S, Bao J, Dang Y, Huang M, Wang Y (2012) Dehydrocorydaline inhibits breast cancer cells proliferation by inducing apoptosis in MCF-7 cells. Am J Chin Med 40(01):177–185. https://doi.org/10.1142/S0192415X12500140

    Article  CAS  Google Scholar 

  65. Wu CC, Chen TH, Liu BL, Wu LC, Chen YC, Tzeng YM, Hsu SL (2013) Destruxin B isolated from entomopathogenic fungus Metarhizium anisopliae induces apoptosis via a Bcl-2 family-dependent mitochondrial pathway in human nonsmall cell lung cancer cells. Evid Based Complement Altern Med. https://doi.org/10.1155/2013/548929

    Article  Google Scholar 

  66. Kim DW, Lee KT, Kwon J, Lee HJ, Lee D, Mar W (2015) Neuroprotection against 6-OHDA-induced oxidative stress and apoptosis in SH-SY5Y cells by 5, 7-Dihydroxychromone: activation of the Nrf2/ARE pathway. Life Sci 130:25–30. https://doi.org/10.1016/j.lfs.2015.02.026

    Article  CAS  Google Scholar 

  67. Gracia-Sancho J, Manicardi N, Ortega-Ribera M, Maeso-Díaz R, Guixé-Muntet S, Fernández-Iglesias A, Bosch J (2019) Emricasan ameliorates portal hypertension and liver fibrosis in cirrhotic rats through a hepatocyte-mediated paracrine mechanism. Hepatol Commun 3(7):987–1000. https://doi.org/10.1002/hep4.1360

    Article  CAS  Google Scholar 

  68. Kravchenko DV, Kuzovkova YA, Kysil VM, Tkachenko SE, Maliarchouk S, Okun IM, Ivachtchenko AV (2005) Synthesis and structure− activity relationship of 4-substituted 2-(2-Acetyloxyethyl)-8-(morpholine-4-sulfonyl) pyrrolo [3, 4-c] quinoline-1, 3-diones as potent caspase-3 inhibitors. J Med Chem 48(11):3680–3683. https://doi.org/10.1021/jm048987t

    Article  CAS  Google Scholar 

  69. Wang F, Wang L, Zhao Y, Li Y, Ping G, Xiao S, Wu C (2014) A novel small-molecule activator of procaspase-3 induces apoptosis in cancer cells and reduces tumor growth in human breast, liver and gallbladder cancer xenografts. Mol Oncol 8(8):1640–1652. https://doi.org/10.1016/j.molonc.2014.06.015

    Article  CAS  Google Scholar 

  70. Kanthasamy AG, Anantharam V, Zhang D, Latchoumycandane C, Jin H, Kaul S, Kanthasamy A (2006) A novel peptide inhibitor targeted to caspase-3 cleavage site of a proapoptotic kinase protein kinase C delta (PKCδ) protects against dopaminergic neuronal degeneration in Parkinson’s disease models. Free Radical Biol Med 41(10):1578–1589. https://doi.org/10.1016/j.freeradbiomed.2006.08.016

    Article  CAS  Google Scholar 

  71. Özören N, Kim K, Burns TF, Dicker DT, Moscioni AD, El-Deiry WS (2000) The caspase 9 inhibitor Z-LEHD-FMK protects human liver cells while permitting death of cancer cells exposed to tumor necrosis factor-related apoptosis-inducing ligand. Cancer Res 60(22):6259–6265

    Google Scholar 

  72. Li H, Nookala S, Re F (2007) Aluminum hydroxide adjuvants activate caspase-1 and induce IL-1β and IL-18 release. J Immunol 178(8):5271–5276. https://doi.org/10.4049/jimmunol.178.8.5271

    Article  CAS  Google Scholar 

  73. Yao H, Yuan Z, Wei G, Chen C, Duan J, Li Y, Liu Y (2017) Thevetiaflavone from Wikstroemia indica ameliorates PC12 cells injury induced by OGD/R via improving ROS-mediated mitochondrial dysfunction. Mol Med Rep 16(6):9197–9202. https://doi.org/10.3892/mmr.2017.7712

    Article  CAS  Google Scholar 

  74. Nicolau-Galmés F, Asumendi A, Alonso-Tejerina E, Pérez-Yarza G, Jangi SM, Gardeazabal J, Boyano MD (2011) Terfenadine induces apoptosis and autophagy in melanoma cells through ROS-dependent and-independent mechanisms. Apoptosis 16(12):1253–1267. https://doi.org/10.1007/s10495-011-0640-y

    Article  CAS  Google Scholar 

  75. Xie Q, Khaoustov VI, Chung CC, Sohn J, Krishnan B, Lewis DE, Yoffe B (2002) Effect of tauroursodeoxycholic acid on endoplasmic reticulum stress–induced caspase-12 activation. Hepatology 36(3):592–601. https://doi.org/10.1053/jhep.2002.35441

    Article  CAS  Google Scholar 

  76. Ichikawa D, Nakamura M, Murota W, Osawa S, Matsushita M, Yanagawa H, Hattori Y (2020) A phenylphthalimide derivative, TC11, induces apoptosis by degrading MCL1 in multiple myeloma cells. Biochem Biophys Res Commun 521(1):252–258. https://doi.org/10.1016/j.bbrc.2019.10.119

    Article  CAS  Google Scholar 

  77. Rudolphi K et al (2003) Pralnacasan, an inhibitor of interleukin-1beta converting enzyme, reduces joint damage in two murine models of osteoarthritis. Osteoarthr Cartil 11(10):738–746. https://doi.org/10.1016/S1063-4584(03)00153-5

    Article  CAS  Google Scholar 

  78. Boxer MB, Quinn AM, Shen M, Jadhav A, Leister W, Simeonov A, Thomas CJ (2010) A highly potent and selective caspase 1 inhibitor that utilizes a key 3-cyanopropanoic acid moiety. ChemMedChem 5(5):730. https://doi.org/10.1002/cmdc.200900531

    Article  CAS  Google Scholar 

  79. Mandal SK, Biswas R, Bhattacharyya SS, Paul S, Dutta S, Pathak S, Khuda-Bukhsh AR (2010) Lycopodine from Lycopodium clavatum extract inhibits proliferation of HeLa cells through induction of apoptosis via caspase-3 activation. Eur J Pharmacol 626(2–3):115–122. https://doi.org/10.1016/j.ejphar.2009.09.033

    Article  CAS  Google Scholar 

  80. Nguyen JT, Wells JA (2003) Direct activation of the apoptosis machinery as a mechanism to target cancer cells. Proc Natl Acad Sci 100(13):7533–7538. https://doi.org/10.1073/pnas.1031631100

    Article  CAS  Google Scholar 

  81. Hachmo Y, Kalechman Y, Skornick I, Gafter U, Caspi RR, Sredni B (2017) The small tellurium compound as101 ameliorates rat crescentic glomerulonephritis: association with inhibition of macrophage caspase-1 activity via very late antigen-4 inactivation. Front Immunol 8:240. https://doi.org/10.3389/fimmu.2017.00240

    Article  CAS  Google Scholar 

  82. Yan T, Hu G, Wang A, Sun X, Yu X, Jia J (2018) Paris saponin VII induces cell cycle arrest and apoptosis by regulating Akt/MAPK pathway and inhibition of P-glycoprotein in K562/ADR cells. Phytother Res 32(5):898–907. https://doi.org/10.1002/ptr.6029

    Article  CAS  Google Scholar 

  83. Smith CE, Soti S, Jones TA, Nakagawa A, Xue D, Yin H (2017) Non-steroidal anti-inflammatory drugs are caspase inhibitors. Cell Chem Biol 24(3):281–292. https://doi.org/10.1016/j.chembiol.2017.02.003

    Article  CAS  Google Scholar 

  84. Kobori M, Yang Z, Gong D, Heissmeyer V, Zhu H, Jung YK, Yuan J (2004) Wedelolactone suppresses LPS-induced caspase-11 expression by directly inhibiting the IKK complex. Cell Death Differ 11(1):123–130. https://doi.org/10.1038/sj.cdd.4401325

    Article  CAS  Google Scholar 

  85. Shishodia S, Azu N, Rosenzweig AJ, Jackson AD (2016) Guggulsterone for chemoprevention of cancer. Curr Pharm Des 22(3):294–306. https://doi.org/10.2174/1381612822666151112153117

    Article  CAS  Google Scholar 

  86. Rohn TT, Kokoulina P, Eaton CR, Poon WW (2009) Caspase activation in transgenic mice with Alzheimer-like pathology: results from a pilot study utilizing the caspase inhibitor, Q-VD-OPh. Int J Clin Exp Med 2(4):300 (PMID: 20057974)

    CAS  Google Scholar 

  87. Kuželová K, Grebeňová D, Brodská B (2011) Dose-dependent effects of the caspase inhibitor Q-VD-OPh on different apoptosis-related processes. J Cell Biochem 112(11):3334–3342. https://doi.org/10.1002/jcb.23263

    Article  CAS  Google Scholar 

  88. Caserta TM, Smith AN, Gultice AD, Reedy MA, Brown TL (2003) Q-VD-OPh, a broad spectrum caspase inhibitor with potent antiapoptotic properties. Apoptosis 8(4):345–352. https://doi.org/10.1023/A:1024116916932

    Article  CAS  Google Scholar 

  89. Yang J, Liu Z, Wang C, Yang R, Rathkey JK, Pinkard OW, Xiao TS (2018) Mechanism of gasdermin D recognition by inflammatory caspases and their inhibition by a gasdermin D-derived peptide inhibitor. Proc Natl Acad Sci 115(26):6792–6797. https://doi.org/10.1073/pnas.1800562115

    Article  CAS  Google Scholar 

  90. Hardin EA, Stoller D, Lawley J, Howden EJ, Hieda M, Pawelczyk J, Levine BD (2020) Noninvasive assessment of cardiac output: accuracy and precision of the closed-circuit acetylene rebreathing technique for cardiac output measurement. J Am Heart Assoc 9(17):e015794. https://doi.org/10.1161/JAHA.120.0157941

    Article  CAS  Google Scholar 

  91. Roth D, Pace NL, Lee A, Hovhannisyan K, Warenits AM, Arrich J, Herkner H (2019) Bedside tests for predicting difficult airways: an abridged Cochrane diagnostic test accuracy systematic review. Anaesthesia 74(7):915–928. https://doi.org/10.1111/anae.14608

    Article  CAS  Google Scholar 

  92. Beaujean AA, Benson NF (2019) The one and the many: Enduring legacies of Spearman and Thurstone on intelligence test score interpretation. Appl Meas Educ 32(3):198–215. https://doi.org/10.1080/08957347.2019.1619560

    Article  Google Scholar 

  93. Zar JH (1972) Significance testing of the Spearman rank correlation coefficient. J Am Stat Assoc 67(339):578–580. https://doi.org/10.2307/2284441

    Article  Google Scholar 

  94. Dewitte K, Fierens C, Stockl D, Thienpont LM (2002) Application of the Bland-Altman plot for interpretation of method-comparison studies: a critical investigation of its practice. Clin Chem 48(5):799–801. https://doi.org/10.1093/clinchem/48.5.799

    Article  CAS  Google Scholar 

  95. Osborn ZT, Villalba N, Derickson PR, Sewatsky TP, Wager AP, Freeman K (2019) Accuracy of point-of-care testing for anemia in the emergency department. Respir Care 64(11):1343–1350. https://doi.org/10.4187/respcare.06364

    Article  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the University of Tehran, Kish International Campus.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sajjad Gharaghani.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bastami, Z., Sheikhpour, R., Razzaghi, P. et al. Proteochemometrics modeling for prediction of the interactions between caspase isoforms and their inhibitors. Mol Divers 27, 249–261 (2023). https://doi.org/10.1007/s11030-022-10425-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11030-022-10425-5

Keywords

Navigation