Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2348 2023-06-17 19:58:35 |
2 format correct Meta information modification 2348 2023-06-19 03:06:55 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Frolova, A.S.; Chepikova, O.E.; Deviataikina, A.S.; Solonkina, A.D.; Zamyatnin, A.A. Role of Nuclear Proteases in Cell Death Pathways. Encyclopedia. Available online: https://encyclopedia.pub/entry/45752 (accessed on 27 July 2024).
Frolova AS, Chepikova OE, Deviataikina AS, Solonkina AD, Zamyatnin AA. Role of Nuclear Proteases in Cell Death Pathways. Encyclopedia. Available at: https://encyclopedia.pub/entry/45752. Accessed July 27, 2024.
Frolova, Anastasia S., Olga E. Chepikova, Anna S. Deviataikina, Alena D. Solonkina, Andrey A. Zamyatnin. "Role of Nuclear Proteases in Cell Death Pathways" Encyclopedia, https://encyclopedia.pub/entry/45752 (accessed July 27, 2024).
Frolova, A.S., Chepikova, O.E., Deviataikina, A.S., Solonkina, A.D., & Zamyatnin, A.A. (2023, June 17). Role of Nuclear Proteases in Cell Death Pathways. In Encyclopedia. https://encyclopedia.pub/entry/45752
Frolova, Anastasia S., et al. "Role of Nuclear Proteases in Cell Death Pathways." Encyclopedia. Web. 17 June, 2023.
Role of Nuclear Proteases in Cell Death Pathways
Edit

Multiple factors can trigger cell death via various pathways, and nuclear proteases have emerged as essential regulators of these processes. While certain nuclear proteases have been extensively studied and their mechanisms of action are well understood, others remain poorly characterized. Regulation of nuclear protease activity is a promising therapeutic strategy that could selectively induce favorable cell death pathways in specific tissues or organs.

protease nucleus cell death apoptosis

1. Introduction

Multicellular organisms are comprised of individual cells acting in tight and regulated cooperation. The development of an organism involves numerous rounds of cell division, but the number of cell divisions is limited, and as a result, the cell that gave rise to a large population dies. Cell death is the final stage in the life of a cell [1]. In some cases, cell death is delayed or, conversely, occurs prematurely. Tumor cells can avoid cell death, thereby replacing healthy tissues and migrating throughout the body [2]. In the case of neurodegenerative diseases, an increase in the occurrence of cell death is observed [3]. Therefore, a complete understanding of the mechanisms of cell death will provide an opportunity to find new options for the treatment of many diseases.
Every compartment in the cell has its own distinct function, with the nucleus serving as a crucial hub for genetic information storage and processing. The nucleus is responsible for controlling numerous cellular functions, including gene expression, DNA replication, and repair. One of the most significant changes that occurs during cell death is the alteration of chromatin structure, DNA degradation, and disassembly of nuclear structural proteins [4][5].
The destruction of nuclear and other proteins is accomplished by proteases, which hydrolyze peptide bonds between amino acids. Proteases are traditionally divided into specific groups according to the type of reaction they catalyze, amino acids in the active center, structure, and other features [6]. The key role of proteases is the ability to cleave proteins and thereby control individual protein levels. This allows proteases to participate in the regulation of many cellular processes, such as cell proliferation and differentiation [7], DNA replication, transcription and repair [8], angiogenesis and extracellular matrix remodeling [9][10], immunity [11][12], cell death [13], etc. Proteases are also involved in pathological conditions. In cancer, proteases regulate proteolytic extracellular matrix (EMC) remodeling, altering cell–cell and cell–matrix interactions that facilitate invasion and metastasis [14][15][16][17][18].
Proteases have been identified in the nucleus [19][20][21]. These proteases play a significant role in the regulation of gene expression [22][23], as well as in the immune response, carcinogenesis [24], and cell death [25]. Recent research has revealed that proteases in the nucleus perform a variety of functions, which include processing and activation of transcription factors, chromatin remodeling, histone modification, and DNA repair. Proteases also play a role in maintaining the structural integrity of the nucleus. The activity of nuclear proteases is tightly regulated to ensure the proper functioning of the cell. Dysregulation of these enzymes can lead to a wide range of diseases, including cancer or neurodegenerative disorders.

2. Nuclear Proteases

Proteases participate in many cellular processes, in which it is necessary to cleave proteins in the extracellular matrix, on the cellular membrane or in some cellular compartments. In many cases, proteases have either a specific localization, such as on the membrane [26], or their localization in cells change due to different factors [27]
Proteases that are secreted into the extracellular environment or anchored in the cell membrane can affect morpho- and angiogenesis by remodeling the extracellular matrix [9][10]. Acidic vesicles, known as lysosomes, contain pH-dependent proteases—cathepsins, napsins, and asparagine endopeptidase [28]. These lysosomal proteases are responsible for protein degradation during phagocytosis, endocytosis, and autophagy, but are also involved in growth factor signaling, and antigen presentation. Mitoproteases, which are found in mitochondria, degrade misfolded or damaged proteins, regulate mitochondrial gene expression and mitophagy, and activate or inhibit a number of other pathways [28].
Research indicates that nuclear proteases may exhibit high specificity towards nuclear proteins. For instance, nuclear cathepsins have been observed to cleave specific nuclear substrates, while lysosomal cathepsins can cleave all available proteins [29]. This specificity presents an exciting opportunity to use nuclear proteases for cleaving specific proteins, potentially modulating cell death by degrading key proteins. Furthermore, nuclear proteases might be utilized for the degradation of unwanted accumulated proteins, such as polyglutamine proteins that are associated with neuropathology [30]. Targeted degradation of these proteins using nuclear proteases could be a promising approach for treatment. Studying the nuclear substrates of nuclear proteases could also provide insight into utilizing them in targeting systems such as PROTACs (proteolysis targeting chimeras) for specific degradation of nuclear proteins [31]. The potential applications of nuclear proteases are vast, but careful study of these proteins is required to fully understand their functions and potential. Understanding the mechanism of action of nuclear proteases, their specificity, and the consequences of their activity will be critical in developing effective therapies and treatments that can harness their potential. Future research should continue to explore the role of nuclear proteases in cellular processes and their potential applications in therapeutic interventions.
Many proteases exhibit both nuclear and cytoplasmic localization, and it remains unclear why certain proteases translocate into the nucleus. Detailed analysis of protein sequences has revealed that some proteases possess a nuclear translocation signal (NLS). For example, matrix metalloproteinase-2 (MMP-2) has an NLS on its C-terminus, and amino acid substitutions in this region result in loss of nuclear localization [32]. Bioinformatic analysis of MMP proteins has shown that all members of this group contain one or more NLSs [21]. However, not all of these proteases have been found in the nucleus, indicating that the presence of an NLS alone may not be sufficient for nuclear translocation. The localization of the NLS within the protein structure plays an important role in determining its localization. In some cases, the NLS may be masked by a prodomain or linker region, preventing the protein from translocating into the nucleus [25]. Given these findings, the mechanisms that dictate whether proteases in their active or inactive forms exhibit nuclear localization are still not fully understood. Further research is necessary to elucidate the factors that influence nuclear translocation of proteases, which could have significant implications for understanding their regulation.

3. Nuclear Compartment in Cell Death

The nucleus is a defining feature of eukaryotic cells and is the largest organelle in most cells. It separates the genome and transcriptional machinery from the cytoplasm [33]. The nucleus serves as the cell’s control center by coordinating processes such as cell growth, metabolism, and cell division. In addition, the nucleus plays a role in some forms of cell death.
There are different classifications and nomenclatures of cell death, based on multiple mechanisms and phenotypes. Historically, three morphologically distinct categories (type I-III cell death), namely apoptosis, autophagy, and necrosis, have been used for classification [34][35]. This morphological classification is still extensively employed. In 2018, the Nomenclature Committee on Cell Death provided molecular marker-based definitions of cell death types. Intrinsic and extrinsic apoptosis are types of cell death that occur in response to internal or external signals. Mitochondrial permeability transition (MPT)-driven necrosis is caused by mitochondrial destruction, while necroptosis and parthanatos are mediated by specific proteins. Iron overload and lipid peroxidation are the triggers for ferroptosis, whereas pyroptosis, entotic cell death, NETosis, and immunogenic cell death are types of cell death that occur as a consequence of an inflammatory response. Two types of cell death involve specific compartments harboring various proteases, namely lysosome-dependent cell death and autophagy-dependent cell death. Cellular senescence is a form of cell death that occurs due to a state of cell division arrest, and mitotic catastrophe happens when cells perform an abortive act of cell division. Cell death can happen in two ways: due to overwhelming damage, which is called accidental cell death, or as a result of specific signaling events, also known as regulated cell death (RCD), which is the physiological form of programmed cell death [36][37]. Among all the mentioned types of cell death, only three occur with the participation of the cell nucleus: apoptosis, parthanatos, and NETosis.
Apoptosis is a multi-pathway mode of cell death that leads to the destruction of cells and the nucleus plays a crucial role in this process. The intrinsic pathway of apoptosis is initiated by internal signals, including DNA damage, which triggers the release of cytochrome c from mitochondria. The regulation of this pathway is carried out by pro- and anti-apoptotic proteins of the BCL-2 family, as well as initiator and effector caspases [38][39]. The activation of initiator caspases by cytochrome c in turn activates the main effector caspases. In contrast, extrinsic signals activate a distinct apoptosis pathway, which ultimately leads to the activation of effector caspases. Once translocated into the nucleus, effector caspases cleave several nuclear proteins, including poly(ADP-ribose) polymerase-1 (PARP-1), lamin, β-tubulin, and others [40]. Cleavage of the inhibitor of caspase-activated DNase (ICAD) by caspase-3 is a crucial event in the apoptotic pathway, allowing caspase-activated DNase (CAD) to induce oligonucleosomal DNA fragmentation [41][42][43]. Other mitochondrial proteins, such as endonuclease G (EndoG) and apoptosis-inducing factor (AIF), also enter the nucleus and initiate chromatin condensation and DNA fragmentation, which can later lead to membrane blebbing. In the final stages of apoptosis, the cell partitions into small apoptotic bodies that are eliminated by macrophages or surrounding cells. In that case, the contents of the cell is not released into the environment and does not trigger an inflammatory reaction.
Parthanatos is a distinct type of cell death that is mainly triggered by DNA damage. In response to this, PARP-1 protein begins to produce an excessive amount of poly(ADP-ribose) (PAR), which is then translocated into the mitochondria. The PAR molecules interact with the mitochondria, inducing the release of AIF [44]. Once AIF enters the nucleus, it triggers extensive DNA fragmentation and chromatin condensation, ultimately leading to cell death. The translocation of AIF from mitochondria to the nucleus, and subsequent nucleus destruction, which is characteristic of this type of cell death, highlights the critical role of the nucleus in parthanatos [45].
NETotic cell death is thought to involve a complex signaling pathway [46][47][48]. Activation of NADPH oxidase by chemical reagents or bacterial action leads to the formation of reactive oxygen species (ROS) [46]. The presence of ROS triggers the release of bactericidal proteins, such as antimicrobial peptides, cytokines, and digestive enzymes, including neutrophil elastase (NE), cathepsin G, azurocidin, and myeloperoxidase (MPO), from the azurophilic granules of neutrophils into the cytosol [49]. NE partially translocates into the nucleus and cleaves nuclear proteins [50]. Peptidyl arginine deiminase 4 (PAD4) also enters the nucleus, where it induces histone citrullination [51][52], leading to DNA decondensation. In the next stage of NETosis, decondensed chromatin, decorated with histones and antimicrobial proteins, is released into the cytoplasm as a result of rupturing of the nuclear envelope. This forms a net-like structure, termed the neutrophil extracellular trap (NET), which is then expelled from the cell [46]. The nucleus’s involvement in NETotic cell death underscores the significance of this compartment in cell death mechanisms.
The cell death mechanisms in the nucleus share a similar pattern across the three types of cell death: apoptosis, NETosis, and parthanatos (Figure 1). During apoptosis and parthanatos, DNA condensation occurs through the common protein AIF, while NETosis involves DNA decondensation, which is critical for NET formation. DNA fragmentation is exclusive to apoptosis. These three cell death types also involve disruption of the nuclear envelope to a varying degree. For example, in apoptosis, nuclear proteases cleave lamins, which leads to destruction of the nuclear envelope and the nucleus as a whole [53][54]. During NETosis, pores form in the nuclear envelope, possibly due to the insertion of gasdermin D protein into the membrane [55]. Disassembly of nuclear lamin without proteolysis is also observed [56]. The process of nuclear destruction during parthanatos has not been extensively studied, and the proteins that are responsible for this process remain unknown.
Figure 1. Role of nucleus in three types of cell death: apoptosis, NETosis and parthanatos. Although these cell death pathways have different activators, the nuclear events that occur during these processes are similar. This includes the degradation of structural and functional proteins by proteases, as well as DNA decondensation or degradation by endonucleases or modification enzymes. Apoptosis proteins are indicated in blue boxes; NETosis—pink, parthanatos—green.
All these processes involve important nuclear regulatory proteins in the cell nucleus. Apoptosis demonstrates how the degradation of such proteins by nuclear proteinases can regulate cell death (Table 1). The pathways of NETosis and parthanatos are not yet fully understood, and there are many gaps in our knowledge of the nucleus’s role in these processes that may involve the action of nuclear proteases.
Table 1. Nuclear substrates in apoptosis, parthanatos, and NETosis.
Cell Death Protease Substrate in Nucleus Substrate Cell Function What Happened after Cleavage Ref.
apoptosis caspase-3 Sp1 Transcription factor Apoptosis [57]
PARP-1 DNA repair Activation of apoptosis [58]
lamin Nuclear envelope Degradation of nucleus [53]
importin-α Import of protein in cell nucleus Downregulate DNA synthesis [59]
large subunit of the DNA replication complex C Regulation of DNA replication Decrease DNA binding [60]
Rad51 DNA repair Activation of apoptosis [61]
ICAD Inhibition of CAD DNA fragmentation [42][62]
calpain lamin A Nuclear envelope Degradation of nucleus [54]
lamin B Nuclear envelope Degradation of nucleus [54]
spectrin Skeletal protein Product of SBPD145, 150i, 120
Activation of apoptosis
[63]
cathepsin L p53 Transcription factor, regulation
of caspase-7 expression
Silencing of CtsL induce the decrease in p53 [64]
prohibitin Transcription factor, regulation
of caspase-7 expression
Silencing of CtsL induce the decrease in p53 [64]
cathepsin B - - DNA condensation and fragmentation [65]
granzyme lamin Nuclear envelope - [66]
PARP DNA repair - [67][68]
ICAD Inhibition of CAD - [67]
- - DNA fragmentation [69]
?—apoptosis calpain PARP DNA repair - [70]
CaMK4 Calcium signaling, regulates β-cell apoptosis - [71]
β-catenin Transcription factor, regular expression of Wnt pathways genes - [72]
c-Fos Transcription factor - [73][74]
c-Jun Transcription factor - [73][74]
Sp3, Sp4 Transcription factor - [75]
p53 Transcription factor - [76]
SPase Sp1 Transcription factor - [77]
Rb Regulates cell growth - [77]
NETosis calpain H3 Maintains structure of DNA Degradation of nuclear envelope [78]
HP1a Gene regulation Chromatin decondensation [78]
lamin A/C Nuclear core structure Degradation of nuclear envelope [78]
H3 Maintains structure of DNA Degradation of nuclear envelope [78]
? ? Chromatin decondensation [78]
neutrophil elastase H1, H2A, H2B, H3, H3 Maintains structure of DNA Chromatin decondensation [50][79]
?—parthanatos calpain PARP - - [70]
granzyme PARP - - [67]
cysteine protease/cathepsin AIF - - [64][80]
?—The involvement in cell death based on the substrate.

References

  1. Gudipaty, S.A.; Conner, C.M.; Rosenblatt, J.; Montell, D.J. Unconventional Ways to Live and Die: Cell Death and Survival in Development, Homeostasis, and Disease. Annu. Rev. Cell Dev. Biol. 2018, 34, 311–332.
  2. Fernald, K.; Kurokawa, M. Evading apoptosis in cancer. Trends Cell Biol. 2013, 23, 620–633.
  3. Cui, J.; Zhao, S.; Li, Y.; Zhang, D.; Wang, B.; Xie, J.; Wang, J. Regulated cell death: Discovery, features and implications for neurodegenerative diseases. Cell Commun. Signal. 2021, 19, 120.
  4. Prokhorova, E.A.; Zamaraev, A.V.; Kopeina, G.S.; Zhivotovsky, B.; Lavrik, I.N. Role of the nucleus in apoptosis: Signaling and execution. Cell. Mol. Life Sci. 2015, 72, 4593–4612.
  5. Saunders, C.A.; Parent, C.A. ScienceDirect Emerging roles for the nucleus during neutrophil signal relay and NETosis. Curr. Opin. Cell Biol. 2019, 62, 135–143.
  6. Gurumallesh, P.; Alagu, K.; Ramakrishnan, B.; Muthusamy, S. A systematic reconsideration on proteases. Int. J. Biol. Macromol. 2019, 128, 254–267.
  7. King, R.W.; Deshaies, R.J.; Peters, J.-M.; Kirschner, M.W. How Proteolysis Drives the Cell Cycle. Science 1996, 274, 1652–1659.
  8. Burby, P.E.; Simmons, Z.W.; Schroeder, J.W.; Simmons, L.A. Discovery of a dual protease mechanism that promotes DNA damage checkpoint recovery. PLoS Genet. 2018, 14, e1007512.
  9. Ghajar, C.M.; George, S.C.; Putnam, A.J. Matrix Metalloproteinase Control of Capillary Morphogenesis. Crit. Rev. Eukaryot. Gene Expr. 2008, 18, 251–278.
  10. Lakka, S.S.; Gondi, C.S.; Rao, J.S. Proteases and glioma angiogenesis. Brain Pathol. 2005, 15, 327–341.
  11. Qureshi, N.; Vogel, S.N.; Van Way, C.; Papasian, C.J.; Qureshi, A.A.; Morrison, D.C. The Proteasome: A Central Regulator of Inflammation and Macrophage Function. Immunol. Res. 2005, 31, 243–260.
  12. Lamkanfi, M.; Kanneganti, T.D. Caspase-7: A protease involved in apoptosis and inflammation. Int. J. Biochem. Cell Biol. 2010, 42, 21–24.
  13. Martin, S.J. Caspases: Executioners of Apoptosis. In Pathobiology of Human Disease; Elsevier: Amsterdam, The Netherlands, 2014; Volume 16, pp. 145–152. ISBN 9780123864567.
  14. Tan, G.-J.; Peng, Z.-K.; Lu, J.-P.; Tang, F.-Q. Cathepsins mediate tumor metastasis. World J. Biol. Chem. 2013, 4, 91–101.
  15. Tedelind, S.; Jordans, S.; Resemann, H.; Blum, G.; Bogyo, M.; Führer, D.; Brix, K. Cathepsin B trafficking in thyroid carcinoma cells. Thyroid Res. 2011, 4, S2.
  16. Mohamed, M.M.; Sloane, B.F. Cysteine cathepsins: Multifunctional enzymes in cancer. Nat. Rev. Cancer 2006, 6, 764–775.
  17. Sloane, B.F.; Rozhin, J.; Johnson, K.; Taylor, H.; Crissman, J.D.; Honn, K.V. Cathepsin B: Association with plasma membrane in metastatic tumors. Proc. Natl. Acad. Sci. USA 1986, 83, 2483–2487.
  18. Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer 2002, 2, 161–174.
  19. Mannello, F.; Medda, V. Nuclear localization of Matrix metalloproteinases. Prog. Histochem. Cytochem. 2012, 47, 27–58.
  20. Soond, S.M.; Kozhevnikova, M.V.; Frolova, A.S.; Savvateeva, L.V.; Plotnikov, E.Y.; Townsend, P.A.; Han, Y.P.; Zamyatnin, A.A. Lost or Forgotten: The nuclear cathepsin protein isoforms in cancer. Cancer Lett. 2019, 462, 43–50.
  21. Frolova, A.S.; Petushkova, A.I.; Makarov, V.A.; Soond, S.M.; Zamyatnin, A.A. Unravelling the network of nuclear matrix metalloproteinases for targeted drug design. Biology 2020, 9, 480.
  22. Sounni, N.E.; Roghi, C.; Chabottaux, V.; Janssen, M.; Munaut, C.; Maquoi, E.; Galvez, B.G.; Gilles, C.; Frankenne, F.; Murphy, G.; et al. Up-regulation of Vascular Endothelial Growth Factor-A by Active Membrane-type 1 Matrix Metalloproteinase Through Activation of Src-Tyrosine Kinases. J. Biol. Chem. 2004, 279, 13564–13574.
  23. Burton, L.J.; Dougan, J.; Jones, J.; Smith, B.N.; Randle, D.; Henderson, V.; Odero-Marah, V.A. Targeting the Nuclear Cathepsin L CCAAT Displacement Protein/Cut Homeobox Transcription Factor-Epithelial Mesenchymal Transition Pathway in Prostate and Breast Cancer Cells with the Z-FY-CHO Inhibitor. Mol. Cell. Biol. 2017, 37, e00297-16.
  24. Xie, Y.; Lu, W.; Liu, S.; Yang, Q.; Shawn Goodwin, J.; Sathyanarayana, S.A.; Pratap, S.; Chen, Z. MMP7 interacts with ARF in nucleus to potentiate tumor microenvironments for prostate cancer progression in vivo. Oncotarget 2016, 7, 47609–47619.
  25. Si-Tayeb, K.; Monvoisin, A.; Mazzocco, C.; Lepreux, S.; Decossas, M.; Cubel, G.; Taras, D.; Blanc, J.F.; Robinson, D.R.; Rosenbaum, J. Matrix metalloproteinase 3 is present in the cell nucleus and is involved in apoptosis. Am. J. Pathol. 2006, 169, 1390–1401.
  26. Knapinska, A.M.; Fields, G.B. The expanding role of mt1-mmp in cancer progression. Pharmaceuticals 2019, 12, 77.
  27. Zheng, T.S.; Schlosser, S.F.; Dao, T.; Hingorani, R.; Crispe, I.N.; Boyer, J.L.; Flavell, R.A. Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo. Proc. Natl. Acad. Sci. USA 1998, 95, 13618–13623.
  28. Müller, S.; Dennemärker, J.; Reinheckel, T. Specific functions of lysosomal proteases in endocytic and autophagic pathways. Biochim. Biophys. Acta—Proteins Proteom. 2012, 1824, 34–43.
  29. Stratigopoulos, G.; LeDuc, C.A.; Cremona, M.L.; Chung, W.K.; Leibel, R.L. Cut-like homeobox 1 (CUX1) regulates expression of the fat mass and obesity-associated and retinitis pigmentosa GTPase regulator-interacting protein-1-like (RPGRIP1L) genes and coordinates leptin receptor signaling. J. Biol. Chem. 2011, 286, 2155–2170.
  30. Havel, L.S.; Li, S.; Li, X.-J. Nuclear accumulation of polyglutamine disease proteins and neuropathology. Mol. Brain 2009, 2, 21.
  31. Békés, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200.
  32. Kwan, J.A.; Schulze, C.J.; Wang, W.; Leon, H.; Sariahmetoglu, M.; Sung, M.; Sawicka, J.; Sims, D.E.; Sawicki, G.; Schulz, R. Matrix metalloproteinase-2 (MMP-2) is present in the nucleus of cardiac myocytes and is capable of cleaving poly (ADP-ribose) polymerase (PARP) in vitro. FASEB J. 2004, 18, 690–692.
  33. Lammerding, J. Mechanics of the nucleus. Compr. Physiol. 2011, 1, 783–807.
  34. Green, D.R.; Llambi, F. Cell Death Signaling. Cold Spring Harb. Perspect. Biol. 2015, 7, a006080.
  35. Schweichel, J.-U.; Merker, H.-J. The morphology of various types of cell death in prenatal tissues. Teratology 1973, 7, 253–266.
  36. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541.
  37. Kroemer, G.; El-Deiry, W.S.; Golstein, P.; Peter, M.E.; Vaux, D.; Vandenabeele, P.; Zhivotovsky, B.; Blagosklonny, M.V.; Malorni, W.; Knight, R.A.; et al. Classification of cell death: Recommendations of the nomenclature committee on cell death. Cell Death Differ. 2005, 12, 1463–1467.
  38. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63.
  39. Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 2019, 20, 175–193.
  40. Nicholson, D.W.; Thornberry, N.A. Caspases: Killer proteases. Trends Biochem. Sci. 1997, 22, 299–306.
  41. Liu, X.; Zou, H.; Slaughter, C.; Wang, X. DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis. Cell 1997, 89, 175–184.
  42. Sakahira, H.; Enari, M.; Nagata, S. Cleavage of CAD inhibitor in CAD activation and DNA degradation during apoptosis. Nature 1998, 391, 96–99.
  43. Enari, M.; Sakahira, H.; Yokoyama, H.; Okawa, K.; Iwamatsu, A.; Nagata, S. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 1998, 391, 43–50.
  44. Andrabi, S.A.; Dawson, T.M.; Dawson, V.L. Mitochondrial and nuclear cross talk in cell death: Parthanatos. Ann. N. Y. Acad. Sci. 2008, 1147, 233–241.
  45. David, K.K.; Andrabi, S.A.; Dawson, T.M.; Dawson, V.L. Parthanatos, a messenger of death. Front. Biosci. 2009, 14, 1116.
  46. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry (Mosc.) 2020, 85, 1178.
  47. Thiam, H.R.; Wong, S.L.; Wagner, D.D.; Waterman, C.M. Cellular Mechanisms of NETosis. Annu. Rev. Cell Dev. Biol. 2021, 36, 191–218.
  48. Neubert, E.; Meyer, D.; Rocca, F.; Günay, G.; Kwaczala-tessmann, A.; Grandke, J.; Senger-sander, S.; Geisler, C.; Egner, A.; Schön, M.P.; et al. Chromatin swelling drives neutrophil extracellular trap release. Nat. Commun. 2018, 9, 3767.
  49. Sheshachalam, A.; Srivastava, N.; Mitchell, T.; Lacy, P.; Eitzen, G. Granule protein processing and regulated secretion in neutrophils. Front. Immunol. 2014, 5, 448.
  50. Papayannopoulos, V.; Metzler, K.D.; Hakkim, A.; Zychlinsky, A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 2010, 191, 677–691.
  51. Wang, Y.; Li, M.; Stadler, S.; Correll, S.; Li, P.; Wang, D.; Hayama, R.; Leonelli, L.; Han, H.; Grigoryev, S.A.; et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J. Cell Biol. 2009, 184, 205–213.
  52. Hamam, H.J.; Palaniyar, N. Post-Translational Modifications in NETosis and NETs-Mediated Diseases. Biomolecules 2019, 9, 369.
  53. Ruchaud, S.; Korfali, N.; Villa, P.; Kottke, T.J.; Dingwall, C.; Kaufmann, S.H.; Earnshaw, W.C. Caspase-6 gene disruption reveals a requirement for lamin A cleavage in apoptotic chromatin condensation. EMBO J. 2002, 21, 1967–1977.
  54. Kenji Takahashi Calpain Substrate Specificity. In Intracellular Calcium-Dependent Proteolysis; CRC Press: Boca Raton, FL, USA, 1990; p. 20. ISBN 9781351073813.
  55. Chen, K.W.; Monteleone, M.; Boucher, D.; Sollberger, G.; Ramnath, D.; Condon, N.D.; von Pein, J.B.; Broz, P.; Sweet, M.J.; Schroder, K. Noncanonical inflammasome signaling elicits gasdermin D–dependent neutrophil extracellular traps. Sci. Immunol. 2018, 3, eaar6676.
  56. Li, Y.; Li, M.; Weigel, B.; Mall, M.; Werth, V.P.; Liu, M. Nuclear envelope rupture and NET formation is driven by PKCα-mediated lamin B disassembly. EMBO Rep. 2020, 21, e48779.
  57. Torabi, B.; Flashner, S.; Beishline, K.; Sowash, A.; Donovan, K.; Bassett, G.; Azizkhan-Clifford, J. Caspase cleavage of transcription factor Sp1 enhances apoptosis. Apoptosis 2018, 23, 65–78.
  58. Tewari, M.; Quan, L.T.; O’Rourke, K.; Desnoyers, S.; Zeng, Z.; Beidler, D.R.; Poirier, G.G.; Salvesen, G.S.; Dixit, V.M. Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase. Cell 1995, 81, 801–809.
  59. Kim, B.J.; Lee, H. Caspase-mediated cleavage of importin-α increases its affinity for MCM and downregulates DNA synthesis by interrupting the binding of MCM to chromatin. Biochim. Biophys. Acta-Mol. Cell Res. 2008, 1783, 2287–2293.
  60. Ubeda, M.; Habener, J.F. The Large Subunit of the DNA Replication Complex C (DSEB/RF-C140) Cleaved and Inactivated by Caspase-3 (CPP32/YAMA) during Fas-induced Apoptosis. J. Biol. Chem. 1997, 272, 19562–19568.
  61. Huang, Y.; Nakada, S.; Ishiko, T.; Utsugisawa, T.; Datta, R.; Kharbanda, S.; Yoshida, K.; Talanian, R.V.; Weichselbaum, R.; Kufe, D.; et al. Role for Caspase-Mediated Cleavage of Rad51 in Induction of Apoptosis by DNA Damage. Mol. Cell. Biol. 1999, 19, 2986–2997.
  62. Tang, D.; Kidd, V.J. Cleavage of DFF-45/ICAD by Multiple Caspases Is Essential for Its Function during Apoptosis. J. Biol. Chem. 1998, 273, 28549–28552.
  63. Zhang, Z.; Larner, S.F.; Liu, M.C.; Zheng, W.; Hayes, R.L.; Wang, K.K.W. Multiple alphaII-spectrin breakdown products distinguish calpain and caspase dominated necrotic and apoptotic cell death pathways. Apoptosis 2009, 14, 1289–1298.
  64. Kenig, S.; Frangež, R.; Pucer, A.; Lah, T. Inhibition of cathepsin L lowers the apoptotic threshold of glioblastoma cells by up-regulating p53 and transcription of caspases 3 and 7. Apoptosis 2011, 16, 671–682.
  65. Vancompernolle, K.; Van Herreweghe, F.; Pynaert, G.; Van de Craen, M.; De Vos, K.; Totty, N.; Sterling, A.; Fiers, W.; Vandenabeele, P.; Grooten, J. Atractyloside-induced release of cathepsin B, a protease with caspase-processing activity. FEBS Lett. 1998, 438, 150–158.
  66. Zhang, D.; Beresford, P.J.; Greenberg, A.H.; Lieberman, J. Granzymes A and B directly cleave lamins and disrupt the nuclear lamina during granule-mediated cytolysis. Proc. Natl. Acad. Sci. USA 2001, 98, 5746–5751.
  67. Lu, H.; Hou, Q.; Zhao, T.; Zhang, H.; Zhang, Q.; Wu, L.; Fan, Z. Granzyme M Directly Cleaves Inhibitor of Caspase-Activated DNase (CAD) to Unleash CAD Leading to DNA Fragmentation. J. Immunol. 2006, 177, 1171–1178.
  68. Froelich, C.J.; Hanna, W.L.; Poirier, G.G.; Duriez, P.J.; D’amours, D.; Salvesen, G.S.; Alnemri, E.S.; Earnshaw, W.C.; Shah, G.M. Granzyme B/Perforin-Mediated Apoptosis of Jurkat Cells Results in Cleavage of Poly(ADP-ribose) Polymerase to the 89-kDa Apoptotic Fragment and Less Abundant 64-kDa Fragment. Biochem. Biophys. Res. Commun. 1996, 227, 658–665.
  69. Pinkoski, M.J.; Heibein, J.A.; Barry, M.; Bleackley, R.C. Nuclear translocation of granzyme B in target cell apoptosis. Cell Death Differ. 2000, 7, 17–24.
  70. McGinnis, K.M.; Gnegy, M.E.; Park, Y.H.; Mukerjee, N.; Wang, K.K.W. Procaspase-3 and Poly(ADP)ribose polymerase (PARP) are calpain substrates. Biochem. Biophys. Res. Commun. 1999, 263, 94–99.
  71. Tremper-Wells, B.; Vallano, M. Lou Nuclear calpain regulates Ca2+-dependent signaling via proteolysis of nuclear Ca2+/calmodulin-dependent protein kinase type IV in cultured neurons. J. Biol. Chem. 2005, 280, 2165–2175.
  72. Abe, K.; Takeichi, M. NMDA-Receptor Activation Induces Calpain-Mediated β-Catenin Cleavages for Triggering Gene Expression. Neuron 2007, 53, 387–397.
  73. Hirai, S.; Kawasaki, H.; Yaniv, M.; Suzuki, K. Degradation of transcription factors, c-Jun and c-Fos, by calpain. FEBS Lett. 1991, 287, 57–61.
  74. Pariat, M.; Salvat, C.; Bebien, M.; Brockly, F.; Altieri, E.; Carillo, S.; Jariel-Encontre, I.; Piechaczyk, M. The sensitivity of c-Jun and c-Fos proteins to calpains depends on conformational determinants of the monomers and not on formation of dimers. Society 2000, 138, 129–138.
  75. Mao, X.; Yang, S.-H.; Simpkins, J.W.; Barger, S.W. Glutamate receptor activation evokes calpain-mediated degradation of Sp3 and Sp4, the prominent Sp-family transcription factors in neurons. J. Neurochem. 2007, 100, 1300–1314.
  76. Kubbutat, M.H.; Vousden, K.H. Proteolytic cleavage of human p53 by calpain: A potential regulator of protein stability. Mol. Cell. Biol. 1997, 17, 460–468.
  77. Nishinaka, T.; Fu, Y.H.F.; Chen, L.I.; Yokoyama, K.; Chiu, R. A unique cathepsin-like protease isolated from CV-1 cells is involved in rapid degradation of retinoblastoma susceptibility gene product, RB, and transcription factor SP1. Biochim. Biophys. Acta-Gene Struct. Expr. 1997, 1351, 274–286.
  78. Gößwein, S.; Lindemann, A.; Mahajan, A.; Maueröder, C.; Martini, E.; Patankar, J.; Schett, G.; Becker, C.; Wirtz, S.; Naumann-Bartsch, N.; et al. Citrullination licenses calpain to decondense nuclei in neutrophil extracellular trap formation. Front. Immunol. 2019, 10, 2481.
  79. Dhaenens, M.; Glibert, P.; Lambrecht, S.; Vossaert, L.; Van Steendam, K.; Elewaut, D.; Deforce, D. Neutrophil Elastase in the capacity of the “H2A-specific protease”. Int. J. Biochem. Cell Biol. 2014, 51, 39–44.
  80. Yuste, V.J.; Moubarak, R.S.; Delettre, C.; Bras, M.; Sancho, P.; Robert, N.; D’Alayer, J.; Susin, S.A. Cysteine protease inhibition prevents mitochondrial apoptosis-inducing factor (AIF) release. Cell Death Differ. 2005, 12, 1445–1448.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 262
Revisions: 2 times (View History)
Update Date: 19 Jun 2023
1000/1000
Video Production Service