This Event is licensed under the Creative Commons BY-SA license. This license allows reusers to distribute, remix, adapt, and build upon the material in any medium or format, so long as attribution is given to the creator. The license allows for commercial use. If you remix, adapt, or build upon the material, you must license the modified material under identical terms.
Event: 1825
Key Event Title
Increase, Cell death
Short name
Biological Context
Level of Biological Organization |
---|
Cellular |
Cell term
Cell term |
---|
cell |
Organ term
Organ term |
---|
organ |
Key Event Components
Key Event Overview
AOPs Including This Key Event
AOP Name | Role of event in AOP | Point of Contact | Author Status | OECD Status |
---|---|---|---|---|
Mitochondrial ATP synthase antagonism leading to growth inhibition (2) | KeyEvent | Arthur Author (send email) | Under development: Not open for comment. Do not cite | |
Mitochondrial complex III antagonism leading to growth inhibition (2) | KeyEvent | Allie Always (send email) | Under development: Not open for comment. Do not cite | |
Cytochrome oxidase inhibition to nasal tissues outcomes | KeyEvent | Agnes Aggy (send email) | Under development: Not open for comment. Do not cite | |
TLR9 activation leading to Multi Organ Failure and ARDS | KeyEvent | Cataia Ives (send email) | Under development: Not open for comment. Do not cite | |
GSK3beta inactivation leads to increased mortality | KeyEvent | Cataia Ives (send email) | Open for citation & comment | |
AHR activation decreasing lung function via AHR-ARNT tox path | KeyEvent | Arthur Author (send email) | Under development: Not open for comment. Do not cite | |
Calcium overload driven development of parkinsonian motor deficits | KeyEvent | Cataia Ives (send email) | Under development: Not open for comment. Do not cite | |
Cytopathic SARS-CoV-2 leads to hyperinflammation | KeyEvent | Allie Always (send email) | Under development: Not open for comment. Do not cite | |
Deposition of energy leading to bone loss | KeyEvent | Cataia Ives (send email) | Open for citation & comment |
Taxonomic Applicability
Life Stages
Life stage | Evidence |
---|---|
All life stages | High |
Sex Applicability
Term | Evidence |
---|---|
Unspecific | High |
Key Event Description
Cell death is part of normal development and maturation cycle, and is the component of many response patterns of living tissues to xenobiotic agents (i.e.. micro organisms and chemicals) and to endogenous modulations, such as inflammation and disturbed blood supply (Kanduc et al., 2002). Many physiological processes require cell death for their function (e.g.., embryonal development and immune selection of B and T cells) (Bertheloot et al., 2021). Defects in cells that result in their inappropriate survival or untimely death can negatively impact development or contribute to a variety of human pathologies, including cancer, AIDS, autoimmune disorders, and chronic infection. Cell death may also occur following exposure to environmental toxins or cytotoxic chemicals. Although this is often harmful, it can be beneficial in some cases, such as in the treatment of cancer (Crowley et al., 2016).
Cell death can be divided into: programmed cell death (cell death as a normal component of development) and non-programmed cell death (uncontrolled death of the cell). Although this simplistic view has blurred the intricate mechanisms separating these forms of cell death, studies have and will uncover new effectors, cell death pathways and reveal a more complex and intertwined landscape of processes involving cell death (Bertheloot et al., 2021).
Programmed cell death: is a form of cell death in which the dying cell plays an active part in its own demise (Cotter & Al-Rubeai, 1995).
Apoptosis At a morphological level, it is characterized by cell shrinkage rather than the swelling seen in necrotic cell death. It is characterized by a number of characteristic morphological changes in the structure of the cell, together with a number of enzyme‐dependent biochemical processes. The result of it being the clearance of cells from the body, with minimal damage to surrounding tissues. An essential feature of apoptosis is the release of cytochrome c from mitochondria, regulated by a balance between proapoptotic and antiapoptotic proteins of the BCL-2 family, initiator caspases (caspase-8, -9 and -10) and effector caspases (caspase-3, -6 and -7). Apoptosis culminates in the breakdown of the nuclear membrane by caspase-6, the cleavage of many intracellular proteins (e.g., PARP and lamin), membrane blebbing, and the breakdown of genomic DNA into nucleosomal structures (Bertheloot et al., 2021). Mechanistically, two main pathways contribute to the caspase activation cascade downstream of mitochondrial cytochrome c release:
- Intrinsic pathway is triggered by dysregulation of or imbalance in intracellular homeostasis by toxic agents or DNA damage. It is characterized by mitochondrial outer membrane permeabilization (MOMP), which results in the release of cytochrome c into the cytosol.
- Extrinsic pathway is initiated by activation of cell surface death receptors. Proapoptotic death receptors include TNFR1/2, Fas and the TNF-related apoptosis-inducing ligand (TRAIL) receptors DR4 and DR5.
Other pathways of programmed cell death are called »non-apoptotic programmed cell-death« or »caspase-independent programmed cell-death« (Blank & Shiloh, 2007).
Necroptosis: This type of regulated cell death, occurs following the activation of the tumor necrosis receptor (TNFR1) by TNFα. Activation of other cellular receptors triggers necroptosis. These receptors include death receptors (i.e., Fas/FasL), Toll-like receptors (TLR4 and TLR3) and cytosolic nucleic acid sensors such as RIG-I and STING, which induce type I interferon (IFN-I) and TNFα production and thus promote necroptosis in an autocrine feedback loop. Most of these pathways trigger NFκB- dependent proinflammatory and prosurvival signals.
Pyroptosis is a type of cell death culminating in the loss of plasma membrane integrity and induced by activation of so-called inflammasome sensors. These include the Nod-like receptor (NLR) family, the DNA receptor Absent in Melanoma 2 (AIM2) and the Pyrin receptor.
Autophagy: is a process where cellular components such as macro proteins or even whole organelles are sequestered into lysosomes for degradation (Mizushima et al., 2008; Shintani & Klionsky, 2004). The lysosomes are then able to digest these substrates, the components of which can either be recycled to create new cellular structures and/or organelles or alternatively can be further processed and used as a source of energy (D’Arcy, 2019).
Anoikis is apoptosis induced by loss of attachment to substrate or to other cells (anoikis). Anoikis overlaps with apoptosis in molecular terms, but is classified as a separate entity because of its specific form od induction (Blank & Shiloh, 2007). Induction of anoikis occurs when cells lose attachment to ECM, or adhere to an inappropriate type of ECM, the latter being the more relevant in vivo (Gilmore, 2005).
Cornification: is programmed cell death of keratinocytes. Cell death in the context of cornification involves distinct enzyme classes such as transglutaminases, proteases, DNases and others (Eckhart et al., 2013).
Non-programmed cell death: occurs accidentally in an unplanned manner.
Necrosis is generally characterized to be the uncontrolled death of the cell, usually following a severe insult, resulting in spillage of the contents of the cell into surrounding tissues and subsequent damage thereof (D’Arcy, 2019).
How It Is Measured or Detected
Assays for Quantitating Cell Death:
- Cell death can be measured by staining a sample of cells with trypan blue, assay is described in protocol: Measuring Cell Death by Trypan Blue Uptake and Light Microscopy (Crowley, Marfell, Christensen, et al., 2015d). Or with propidium Iodide, assay is described in protocol: Measuring Cell Death by Propidium Iodide (PI) Uptake and Flow Cytometry (Crowley & Waterhouse, 2015a)
- TUNEL technique: in situ terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling can be used to detect apoptotic cells (Bever & Fekete, 1999; Uribe et al., 2013).
Assays for Quantitating Cell Survival
Colony-forming assay can be used to define the number of cells in a population that are capable of proliferating and forming large groups of cells. Described in Protocol: Measuring Survival of Adherent Cells with the Colony-Forming Assay (Crowley, Christensen, & Waterhouse, 2015c); Measuring Survival of Hematopoietic Cancer Cells with the Colony-Forming Assay in Soft Agar (Crowley & Waterhouse, 2015b).
ASSAYS TO DISTINGUISH APOPTOSIS FROM NECROSIS AND OTHER DEATH MODALITIES
Detecting Nuclear Condensation: The nucleus is generally round in healthy cells but fragmented in apoptotic cells. Dyes such as Giemsa or hematoxylin, which are purple in color and therefore easily viewed using light microscopy, are commonly used to stain the nucleus. Other features of apoptosis and necrosis, such as plasma membrane blebbing or rupture, can be identified by staining the cytoplasm with eosin. Eosin is pinkish in color and can also be viewed using light microscopy. Hematoxylin and eosin are, therefore, commonly used together to stain cells. Assay is described in Protocol: Morphological Analysis of Cell Death by Cytospinning Followed by Rapid Staining (Crowley, Marfell, & Waterhouse, 2015c); Analyzing Cell Death by Nuclear Staining with Hoechst 33342 (Crowley, Marfell, & Waterhouse, 2015a).
Detection of DNA Fragmentation: Apoptotic cells with fragmented DNA can be identified and distinguished from live cells by staining with Propidium Iodide (PI) and measuring DNA content by flow cytometry. This assay is described in Protocol: Measuring the DNA Content of Cells in Apoptosis and at Different Cell-Cycle Stages by Propidium Iodide Staining and Flow Cytometry (Crowley, Chojnowski, & Waterhouse, 2015a). TUNEL technique can also be used: in situ terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling can be used to detect apoptotic cells (Bever & Fekete, 1999; Crowley, Marfell, & Waterhouse, 2015b; Uribe et al., 2013).
Detecting Phosphatidylserine Exposure: Apoptosis is also characterized by exposure of phosphatidylserine (PS) on the outside of apoptotic cells, which acts as a signal that triggers removal of the dying cell by phagocytosis. Annexin V, can selectively bind to PS to label apoptotic cells in which PS is exposed. Purified annexin V can be conjugated to various fluorochromes, which can then be visualized by fluorescence microscopy or detected by flow cytometry. This assay is described in protocol: Quantitation of Apoptosis and Necrosis by Annexin V Binding, Propidium Iodide Uptake, and Flow Cytometry (Crowley, Marfell, Scott, et al., 2015e).
Detecting Caspase Activity: antibodies that specifically recognize the cleaved fragments of caspases and their substrates can be used to specifically detect caspase activity in apoptotic cells by immunocytochemistry. Flow cytometry (using primary antibodies conjugated to fluorescent molecules, or by counter staining with fluorescently labeled antibodies against the primary antibody) can then be used to quantitate the number of apoptotic cells. This assay is described in protocol: Detecting Cleaved Caspase-3 in Apoptotic Cells by Flow Cytometry (Crowley & Waterhouse, 2015a).
Detecting Mitochondrial Damage: flow cytometry can be used to quantitate the number of cells that have reduced mitochondrial transmembrane potential, which is commonly associated with cytochrome c release during apoptosis. For this assay see protocol: Measuring Mitochondrial Transmembrane Potential by TMRE Staining (Crowley, Christensen, & Waterhouse, 2015b).
Listed below are common methods for detecting the KE, however there may be other comparable methods that are not listed.
Measures of apoptotic cytomorphological alterations:
Apoptotic cells exhibit electron dense nuclei, nuclear fragmentation, intact cell membrane up to the disintegration phase, disorganized cytoplasmic organelles, large clear vacuoles, blebs at cell surface, and apoptotic bodies, which can be visualized with various methods. (Elmore, 2007; Watanabe et al., 2002)
Method of Measurement |
Reference |
Description |
OECD Approved Assay |
Transmission electron microscopy (TEM) / Scanning electron microscopy (SEM)/ Fluorescence microscopy |
Martinez, Reif, and Pappas, 2010; Watanabe et al., 2002 |
TEM and SEM can image the cytomorphological alterations caused by apoptosis. |
No |
Stains: |
|||
Hematoxylin with eosin |
Elmore, 2007 |
Hematoxylin stains nuclei blue and eosin stains the cytoplasm/extracellular matrix pink, allowing for the visualization of the cytomorphological alterations of cells. |
No |
Toluidine blue or methylene blue |
Watanabe et al., 2002 |
Toluidine blue stains cellular nuclei, and identifies malignant tissue, which has an increased DNA content and a higher nuclear-to-cytoplasmic ratio. Methylene blue stain applied to a healthy cell sample results in a colorless stain. This is due to the cell's enzymes, which reduce the methylene blue, thereby, reducing its color. Methylene blue stain applied to a dead cell sample turns blue. |
No |
DAPI |
Crowley, Marfell, and Waterhouse, 2016 |
Binds strongly to adenine–thymine-rich regions in the DNA. DAPI can stain live and fixed cells. It passes less efficiently through the membrane in live cells. |
Yes |
Hoescht 33342 |
Crowley, Marfell, and Waterhouse, 2016 |
Binds to DNA in live and fixed cells, used to measure DNA condensation. |
Yes |
Acridine Orange (AO) |
Watanabe et al., 2002 |
Interacts with DNA/RNA through intercalation/electrostatic interaction, is able to penetrate cell membranes. Stains live cells green and dead cells red. |
No |
Nile blue sulfate |
Watanabe et al., 2002 |
Stains cell nuclei and lysosomes, indicating apoptotic bodies. |
No |
Neutral red |
Watanabe et al., 2002 |
Measures lysosomal membrane integrity |
No |
LysoTracker Red |
Watanabe et al., 2002 |
Measures phagolysosomal activity that occurs due to the engulfment of apoptotic bodies. |
No |
DNA damage/fragmentation assays:
Assay |
Reference |
Description |
OECD Approved Assay |
Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assay |
Kressel and Groscurth, 1994 |
Apoptosis is detected with the TUNEL method to assay the endonuclease cleavage products by enzymatically end-labeling the DNA strand breaks. |
Yes |
Nicoletti Assay (SubG1 cell fragment measurement) |
Nicoletti et al., 1991 |
Measures DNA content in nuclei at the pre-G1 phase of the cell cycle (apoptotic nuclei have less DNA than nuclei in healthy cells). |
No |
Cell Death Detection ELISA kit |
Parajuli, 2014 |
Apoptotic nucleosomes are detected using the Cell Death Detection ELISA kit, which were calculated as absorbance subtraction at 405 nm and 490 nm. |
No |
Measurement of apoptotic markers through immunochemistry:
Method of Measurement |
Reference |
Description |
OECD Approved Assay |
Western blot / immunofluorescence microscopy / immunohistochemistry |
Elmore 2007; Martinez, Reif, and Pappas, 2010; Parajuli et al, 2014 |
Apoptosis can be detected with the expression of various apoptotic markers by western blotting using antibodies. Markers can include: cytosolic cytochrome-c; caspases 2, 3, 6, 7, 8, 9, 10; Bax; Bcl-2 (apoptosis inhibitor); BIRC2; BIRC3; GAPDH; PARP; CDK2; CDK4; cyclin D1; p53; p63; p73; cytokeratin-18 |
No |
Measures of altered caspase activity:
Method of Measurement |
Reference |
Description |
OECD Approved Assay |
Caspase-3 and caspase-9 activity is measured with the enzyme-catalyzed release of p-nitroanilide (pNA) and quantified at 405 nm |
Wu, 2016 |
Visualizes caspase-3 and caspase-9 activity |
No |
PhiPhiLux Assay |
Watanabe et al., 2002 |
The PhiPhiLux molecule becomes fluorescent once it is cleaved by caspase-3, indicating caspase activity. |
No |
Ferrocene reporter |
Martinez, Reif, and Pappas, 2010 |
An electrochemical method to detect apoptosis. Ferrocene is attached to a peptide. The peptide sequence is a caspase 3 cleavage site and the ferrocene acts as the electrochemical reporter. The more caspase cleavage that occurs, the more ferrocene molecules are cleaved, the stronger the signal. |
No |
Self-assembled monolayers for matrix assisted laser desorption ionization time-of-flight mass spectrometry (SAMDI-MS) assay |
Martinez, Reif, and Pappas, 2010 |
This assay detects caspase activity. |
No |
Measures of altered mitochondrial physiology:
Method of Measurement |
Reference |
Description |
OECD Approved Assay |
Laser scanning confocal microscopy (LSCM) |
Watanabe et al., 2002 |
LCSM can monitor many mitochondrial events following staining of cells, such as: mitochondrial permeability transition, depolarization of the inner mitochondrial membrane, which may be indicative of apoptosis. |
No |
Fluorescent, cationic, lipophilic mitochondrial dyes, such as: JC-1 dye, Rhodamine, DiOC6, Mitotracker red |
Martinez, Reif, and Pappas, 2010; Sivandzade, Bhalerao, and Cucullo, 2019 |
These mitochondrial dyes can indicate disintegration of the mitochondrial outer membrane’s electrochemical gradient, as different fluorescence is observed between healthy and apoptotic cells. In healthy cells the dye accumulates in aggregates, but in apoptotic cells missing the electrochemical membrane, the dye will spread out into the cytoplasm providing different fluorescent signals. |
No |
Other measures:
Method of measurement |
Reference |
Description |
OECD Approved Assay |
Apoptosis PCR microarray |
Elmore, 2007 |
A method to profile the gene expression of many apoptotic-related genes, for example: ligands, receptors, intracellular modulators, and transcription factors. |
No |
Fluorescence correlation spectroscopy (FCS) or dual-colour fluorescence cross-correlation spectroscopy (dcFCCS) |
Martinez, Reif, and Pappas, 2010 |
Used to measure protease activity. |
No |
Apoptosis is measured with Annexin V-FITC probes |
Elmore, 2007; Wu et al., 2016 |
A measure of apoptotic membrane alterations. Annexin-V detects externalized phosphatidylserine residues, a result of apoptosis. Can be conducted in conjunction with propidium iodide (PI) staining. The relative percentage of Annexin V-FITC-positive/PI-negative cells is analyzed by flow cytometry. |
Yes |
Domain of Applicability
The process of cell death is highly conserved within multi‐cellular organisms. (Lockshin & Zakeri, 2004).
Taxonomic applicability: Increased cell death is applicable to all animals. This includes vertebrates such as humans, mice and rats (Alberts et al., 2002).
Life stage applicability: There is insufficient data on life stage applicability of this KE.
Sex applicability: This key event is not sex specific (Forger and de Vries, 2010; Ortona Matarrese, and Malorni, 2014).
Evidence for perturbation by a stressor: Multiple studies show that cell death can be increased or disrupted by many types of stressors including ionizing radiation and altered gravity (Zhu et al., 2016).
References
Alberts, B. et al. (2002), “Programmed Cell Death (Apoptosis)”, in Molecular Biology of the Cell. 4th edition, Garland Science, New York, https://www.ncbi.nlm.nih.gov/books/NBK26873/
Bertheloot, D., Latz, E., & Franklin, B. S. (2021). Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cellular & Molecular Immunology, 18, 1106–1121. https://doi.org/10.1038/s41423-020-00630-3
Bever, M. M., & Fekete, D. M. (1999). Ventromedial focus of cell death is absent during development of Xenopus and zebrafish inner ears. Journal of Neurocytology, 28(10–11), 781–793. https://doi.org/10.1023/a:1007005702187
Blank, M., & Shiloh, Y. (2007). Cell Cycle Programs for Cell Death: Apoptosis is Only One Way to Go. Cell Cycle, 6(6), 686–695. https://doi.org/10.4161/cc.6.6.3990
Cotter, T. G., & Al-Rubeai, M. (1995). Cell death (apoptosis) in cell culture systems. Trends in Biotechnology, 13(4), 150–155. https://doi.org/10.1016/S0167-7799(00)88926-X
Crowley, L. C., Chojnowski, G., & Waterhouse, N. J. (2015a). Measuring the DNA content of cells in apoptosis and at different cell-cycle stages by propidium iodide staining and flow cytometry. Cold Spring Harbor Protocols, 10, 905–910. https://doi.org/10.1101/pdb.prot087247
Crowley, L. C., Christensen, M. E., & Waterhouse, N. J. (2015b). Measuring mitochondrial transmembrane potential by TMRE staining. Cold Spring Harbor Protocols, 12, 1092–1096. https://doi.org/10.1101/pdb.prot087361
Crowley, L. C., Christensen, M. E., & Waterhouse, N. J. (2015c). Measuring survival of adherent cells with the Colony-forming assay. Cold Spring Harbor Protocols, 8, 721–724. https://doi.org/10.1101/pdb.prot087171
Crowley, L. C., Marfell, B. J., Christensen, M. E., & Waterhouse, N. J. (2015d). Measuring cell death by trypan blue uptake and light microscopy. Cold Spring Harbor Protocols, 7, 643–646. https://doi.org/10.1101/pdb.prot087155
Crowley, L. C., Marfell, B. J., Scott, A. P., Boughaba, J. A., Chojnowski, G., Christensen, M. E., & Waterhouse, N. J. (2016). Dead cert: Measuring cell death. Cold Spring Harbor Protocols, 2016(12), 1064–1072. https://doi.org/10.1101/pdb.top070318
Crowley, L. C., Marfell, B. J., Scott, A. P., & Waterhouse, N. J. (2015e). Quantitation of apoptosis and necrosis by annexin V binding, propidium iodide uptake, and flow cytometry. Cold Spring Harbor Protocols, 11, 953–957. https://doi.org/10.1101/pdb.prot087288
Crowley, L. C., Marfell, B. J., & Waterhouse, N. J. (2015a). Analyzing cell death by nuclear staining with Hoechst 33342. Cold Spring Harbor Protocols, 9, 778–781. https://doi.org/10.1101/pdb.prot087205
Crowley, L. C., Marfell, B. J., & Waterhouse, N. J. (2015b). Detection of DNA fragmentation in apoptotic cells by TUNEL. Cold Spring Harbor Protocols, 10, 900–905. https://doi.org/10.1101/pdb.prot087221
Crowley, L. C., Marfell, B. J., & Waterhouse, N. J. (2015c). Morphological analysis of cell death by cytospinning followed by rapid staining. Cold Spring Harbor Protocols, 9, 773–777. https://doi.org/10.1101/pdb.prot087197
Crowley, L. C., & Waterhouse, N. J. (2015a). Detecting cleaved caspase-3 in apoptotic cells by flow cytometry. Cold Spring Harbor Protocols, 11, 958–962. https://doi.org/10.1101/pdb.prot087312
Crowley, L. C., & Waterhouse, N. J. (2015b). Measuring survival of hematopoietic cancer cells with the Colony-forming assay in soft agar. Cold Spring Harbor Protocols, 8, 725. https://doi.org/10.1101/pdb.prot087189
D’Arcy, M. S. (2019). Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biology International, 43(6), 582–592. https://doi.org/10.1002/cbin.11137
Eckhart, L., Lippens, S., Tschachler, E., & Declercq, W. (2013). Cell death by cornification. Biochimica et Biophysica Acta - Molecular Cell Research, 1833(12), 3471–3480. https://doi.org/10.1016/j.bbamcr.2013.06.010
Elmore, S. (2007), “Apoptosis: A Review of Programmed Cell Death”, Toxical Pathology, Vol. 35/4, SAGE, https://doi.org/10.1080/01926230701320337.
Forger, N. G. and G. J. de Vries (2010), “Cell death and sexual differentiation of behavior: worms, flies, and mammals”, Current opinion in neurobiology, Vol. 20/6, Elsevier, Amsterdam, https://doi.org/10.1016/j.conb.2010.09.006
Gilmore, A. P. (2005). Anoikis. Cell Death and Differentiation, 12, 1473–1477. https://doi.org/10.1038/sj.cdd.4401723
Kanduc, D., Mittelman, A., Serpico, R., Sinigaglia, E., Sinha, A. A., Natale, C., Santacroce, R., Di Corcia, M. G., Lucchese, A., Dini, L., Pani, P., Santacroce, S., Simone, S., Bucci, R., & Farber, E. (2002). Cell death: apoptosis versus necrosis (review). International Journal of Oncology, 21(1), 165–170. https://doi.org/10.3892/ijo.21.1.165
Kressel, M. and P. Groscurth (1994), "Distinction of apoptotic and necrotic cell death by in situ labelling of fragmented DNA", Cell and tissue research, Vol. 278/3, Nature, https://doi.org/10.1007/BF00331373.
Lockshin, R. A., & Zakeri, Z. (2004). Apoptosis, autophagy, and more. International Journal of Biochemistry and Cell Biology, 36(12), 2405–2419. https://doi.org/10.1016/j.biocel.2004.04.011
Martinez, M. M., R. D. Reif, and D. Pappas (2010), “Detection of apoptosis: A review of conventional and novel techniques”, Analytical Methods, Vol. 2/8, Royal Society of Chemistry, https://doi.org/10.1039/C0AY00247J
Mizushima, N., Levine, B., Cuervo, A. M., & Klionsky, D. J. (2008). Autophagy fights disease through cellular self-digestion. Nature, 451(7182), 1069–1075. https://doi.org/10.1038/nature06639
Nicoletti I. et al. (1991), “A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry”, Journal of Immunological Methods, Vol. 139/2, Elsevier, Amsterdam, https://doi.org/10.1016/0022-1759(91)90198-O
Ortona, E., P. Matarrese, and W. Malorni (2014), “Taking into account the gender issue in cell death studies”, Cell Death & Disease, Vol. 5, Nature, https://doi.org/10.1038/cddis.2014.73.
Parajuli, K. R. et al. (2014), "Methoxyacetic acid suppresses prostate cancer cell growth by inducing growth arrest and apoptosis", American journal of clinical and experimental urology, Vol. 2/4, pp. 300-312.
Shintani, T., & Klionsky, D. J. (2004). Autophagy in health and disease: A double-edged sword. Science, 306(5698), 990–995. https://doi.org/10.1126/science.1099993
Sivandzade, F., A. Bhalerao and L. Cucullo (2019), “Analysis of the Mitochondrial Membrane Potential Using Cationic JC-1 Dye as a Sensitive Fluorescent Probe”, Bio Protocol, Vol. 9/1, https://doi.org/10.21769/BioProtoc.3128.
Uribe, P. M., Sun, H., Wang, K., Asuncion, J. D., & Wang, Q. (2013). Aminoglycoside-Induced Hair Cell Death of Inner Ear Organs Causes Functional Deficits in Adult Zebrafish (Danio rerio). PLoS ONE, 8(3), 58755. https://doi.org/10.1371/journal.pone.0058755
Wade, M. G. et al. (2008), "Methoxyacetic acid-induced spermatocyte death is associated with histone hyperacetylation in rats", Biology of Reproduction, Vol. 78/5, Oxford University Press, Oxford, https://doi.org/10.1095/biolreprod.107.065151
Watanabe, M., et al. (2002), “The pros and cons of apoptosis assays for use in the study of cells, tissues, and organs”, Microscopy and microanalysis, Vol. 8/5, Cambridge University Press, Cambridge, https://doi.org/10.1017/S1431927602010346.
Wu, R. et al. (2016), "microRNA-497 induces apoptosis and suppressed proliferation via the Bcl-2/Bax-caspase9-caspase 3 pathway and cyclin D2 protein in HUVECs", PLoS One, Vol. 11/12, PLOS, San Francisco, https://doi.org/10.1371/journal.pone.0167052.
Zhu, M., et al. (2021), “Immunogenic Cell Death Induction by Ionizing Radiation”, Frontiers in Immunology, Vol. 12, https://doi.org/10.3389/FIMMU.2021.705361.