КЛІТИННА ЗАГИБЕЛЬ: СУЧАСНИЙ ПОГЛЯД НА АПОПТОЗ ТА АУТОФАГІЮ

Автор(и)

  • В.О. Срібна

DOI:

https://doi.org/10.24061/1727-4338.XXII.2.84.2023.11

Ключові слова:

клітинна загибель, апоптоз, автофагія

Анотація

Мета роботи – здійснити аналіз даних літератури стосовно сучасного стану
досліджень клітинної загибелі, взаємозв’язку сигнальних шляхів апоптозу й
аутофагії в умовах різних патологій, а також ключових посередників обох процесів.
Висновки. Дані літератури засвідчують про наявність чіткого взаємозв’язку
сигнальних шляхів апоптозу й аутофагії в умовах різних патологічних процесів,
який здійснюється через ключові посередники, зокрема кальцій (Ca2+), ліпідні
рафти, ERLIN, PINK1 тощо. Клітинна загибель є перспективною терапевтичною
мішенню для лікування захворювань у людини.

Посилання

Galluzzi L, Blomgren K, Kroemer G. Mitochondrial membrane

permeabilization in neuronal injury. Nat Rev Neurosci.

;10(7):481-94. doi: 10.1038/nrn2665

Kers J, Leemans JC, Linkermann A. An overview of pathways

of regulated necrosis in acute kidney injury. Semin Nephrol.

;36(3):139-52. doi: 10.1016/j.semnephrol.2016.03.002

Fuchs Y, Steller H. Programmed cell death in animal development and

disease. Cell. 2011;147(4):742-58. doi: 10.1016/j.cell.2011.10.033

Delbridge ARD, Grabow S, Strasser A, Vaux DL. Thirty years

of BCL-2: translating cell death discoveries into novel cancer

therapies. Nat Rev Cancer. 2016;16(2):99-109. doi: 10.1038/

nrc.2015.17

Dillon CP, Green DR. Molecular cell biology of apoptosis and

necroptosis in cancer. Adv Exp Med Biol. 2016;930:1-23. doi:

1007/978-3-319-39406-0_1

Linkermann A, Stockwell BR, Krautwald S, Anders HJ. Regulated

cell death and inflammation: an auto-amplification loop causes

organ failure. Nat Rev Immunol. 2014;14(11):759-67. doi:

1038/nri3743

Yatim N, Cullen S, Albert ML. Dying cells actively regulate

adaptive immune responses. Nat Rev Immunol. 2017;17(4):262-75.

doi: 10.1038/nri.2017.9

Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis

P. Inducers of immunogenic cancer cell death. Cytokine Growth

Factor Rev. 2013;24(4):319-33. doi: 10.1016/j.cytogfr.2013.01.005

Inoue H, Tani K. Multimodal immunogenic cancer cell death as

a consequence of anticancer cytotoxic treatments. Cell Death

Differ. 2014;21(1):39-49. doi: 10.1038/cdd.2013.84

Conrad M, Angeli JPF, Vandenabeele P, Stockwel BR. Regulated

necrosis: disease relevance and therapeutic opportunities. Nat Rev

Drug Discov. 2016;15(5):348-66. doi: 10.1038/nrd.2015.6

Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in

development, inflammation and disease. Nat Rev Mol Cell Biol.

;18(2):127-36. doi: 10.1038/nrm.2016.149

Galluzzi L, Kepp O, Chan FKM, Kroemer G. Necroptosis:

mechanisms and relevance to disease. Annu Rev Pathol.

;12:103-30. doi: 10.1146/annurev- pathol-052016-100247

Maskarinec SF, McKelvy M, Boyle K, Hotchkiss H, Duarte ME,

Addison B, et al. Neutrophil functional heterogeneity is a fixed

phenotype and is associated with distinct gene expression profiles.

J Leukoc Biol. 2022;112(6):1485-95. doi: 10.1002/jlb.4a0322-164r

Hartel JC, Merz N, Grösch S. How sphingolipids affect T cells in the

resolution of inflammation. Front Pharmacol [Internet]. 2022[cited

Aug 15];13:1002915. Available from: https://www.ncbi.nlm.

nih.gov/pmc/articles/PMC9513432/pdf/fphar-13-1002915.pdf doi:

3389/fphar.2022.1002915

Garg AD, Martin SM, Golab J, Agostinis P. Danger signalling

during cancer cell death: origins, plasticity and regulation. Cell

Death Differ. 2014;21(1):26-38. doi: 10.1038/cdd.2013.48

Vandenabeele P, Vandecasteele K, Bachert C, Krysko O,

Krysko D. Immunogenic apoptotic cell death and anticancer

immunity. Adv Exp Med Biol [Internet]. 2016[cited

Aug 17];930:133-49. Available from: https://link.

springer.com/chapter/10.1007/978-3-319-39406-0_6 doi:

1007/978-3-319-39406-0_6

Hernandez C, Huebener P, Schwabe RF. Damage- associated

molecular patterns in cancer: a double- edged sword. Oncogene.

;35(46):5931-41. doi: 10.1038/onc.2016.104

Soares M, Teixeira L, Moita LF. Disease tolerance and immunity

in host protection against infection. Nat Rev Immunol.

;17(2):83-96. doi: 10.1038/nri.2016.136

Gabusi E, Lenzi E, Manferdini C, Dolzani P, Columbaro M, Saleh Y, et

al. Autophagy Is a Crucial Path in Chondrogenesis of Adipose- Derived

Mesenchymal Stromal Cells Laden in Hydrogel. Gels [Internet].

[cited 2023 Aug 15];24;8(12):766. Available from: https://www.

mdpi.com/2310-2861/8/12/766 doi: 10.3390/gels8120766

Mu W, Rezek V, Martin H, Carrillo MA, Tomer S, Hamid P, et al.

Autophagy inducer rapamycin treatment reduces IFN-I-mediated

Inflammation and improves anti- HIV-1 T cell response in vivo. JCI

Insight [Internet]. 2022[cited 2023 Aug 17];7(22): e159136. Available

from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9746825/

pdf/jciinsight-7-159136.pdf doi: 10.1172/jci.insight.159136

Schweichel JU, Merker HJ. The morphology of various types of

cell death in prenatal tissues. Teratology. 1973;7(3):253-66. doi:

1002/tera.1420070306

Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L,

et al. Cell death modalities: classification and pathophysiological

implications. Cell Death Differ. 2007;14(7):1237-43. doi: 10.1038/

sj.cdd.4402148

Kroemer G, El-Deiry WS, Golstein P, Peter ME, Vaux D,

Vandenabeele P, et al. Classification of cell death: recommendations

of the Nomenclature Committee on Cell Death. Cell Death Differ.

;12(Suppl 2):1463-67. doi: 10.1038/sj.cdd.4401724

Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES,

Baehrecke EH, et al. Classification of cell death: recommendations

of the Nomenclature Committee on Cell Death 2009. Cell Death

Differ. 2009;16(1):3-11. doi: 10.1038/cdd.2008.150

Galluzzi L, Aaronson SA, Abrams J, Alnemri ES, Andrews DW,

Baehrecke EH, et al. Guidelines for the use and interpretation of

assays for monitoring cell death in higher eukaryotes. Cell Death

Differ. 2009;16(8):1093-107. doi: 10.1038/cdd.2009.44

Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH,

Blagosklonny MV, et al. Molecular definitions of cell death

subroutines: recommendations of the Nomenclature Committee

on Cell Death 2012. Cell Death Differ. 2012;19(1):107-20. doi:

1038/cdd.2011.96

Galluzzi L, Pedro JMBS, Vitale I, Aaronson SA, Abrams JM,

Adam D, et al. Essential versus accessory aspects of cell death:

recommendations of the NCCD 2015. Cell Death Differ.

;22(1):58-73. doi: 10.1038/cdd.2014.137

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis

P, et al. Molecular mechanisms of cell death: recommendations

of the Nomenclature Committee on Cell Death 2018. Cell Death

Differ. 2018;25(3):486-541. doi: 10.1038/s41418-017-0012-4

Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis

N. Crosstalk between apoptosis, necrosis and autophagy.

Biochim Biophys Acta. 2013;1833(12):3448-59. doi: 10.1016/j.

bbamcr.2013.06.001

Redondo M, Fùnez R, Esteban F. Apoptosis in the Development and

Treatment of Laryngeal Cancer: Role of p53, Bcl-2 and Clusterin.

In: Chen GG, Lai PBS, editors. Apoptosis in Carcinogenesis and

Chemotherapy. Dordrecht: Springer; 2009, p. 237-49.

Chaudhry GES, Akim AM, Sung YY, Sifzizul TMT. Cancer and

apoptosis: The apoptotic activity of plant and marine natural products

and their potential as targeted cancer therapeutics. Front Pharmacol

[Internet]. 2022[cited 2023 Aug 15];13:842376. Available from:

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9399632/pdf/

fphar-13-842376.pdf doi: 10.3389/fphar.2022.842376

Sheshachalam A, Srivastava N, Mitchell T, Lacy P, Eitzen

G. Granule Protein Processing and Regulated Secretion in

Neutrophils. Front Immunol [Internet]. 2014[cited 2023 Aug

;5:448. Available from: https://www.ncbi.nlm.nih.gov/pmc/

articles/PMC4168738/pdf/fimmu-05-00448.pdf doi: 10.3389/

fimmu.2014.00448

Trapani J. Granzymes: a family of lymphocyte granule serine

proteases. Genome Biol [Internet]. 2001[cited 2023 Aug

;2(12); reviews3014.1-reviews3014.7. Available from:

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC138995/

pdf/gb-2001-2-12-reviews3014.pdf doi: 10.1186/

gb-2001-2-12-reviews3014

Gudipaty SA, Conner CM, Rosenblatt J, Montell D. Unconventional

Ways to Live and Die: Cell Death and Survival in Development,

Homeostasis, and Disease. Annu Rev Cell Dev Biol.

;34:311-32. doi: 10.1146/annurev- cellbio-100616-060748

Galluzzi L, Pedro JMBS, Blomgren K, Kroemer G. Autophagy

in acute brain injury. Nat Rev Neurosci. 2016;17(8):467-84. doi:

1038/nrn.2016.51

Anding A, Baehrecke E. Autophagy in Cell Life and Cell

Death. Curr Top Dev Biol. 2015;114:67-91. doi: 10.1016/

bs.ctdb.2015.07.012

Xu T, Nicolson S, Denton D, Kumar S. Distinct requirements of

Autophagy- related genes in programmed cell death. Cell Death

Differ. 2015;22(11):1792-802. doi: 10.1038/cdd.2015.28

Galluzzi L, Pedro JMBS, Levine B, Green DR, Kroemer

G. Pharmacological modulation of autophagy: therapeutic

potential and persisting obstacles. Nat Rev Drug Discov.

;16(7):487-511. doi: 10.1038/nrd.2017.22

Gatica D, Chiong M, Lavandero S, Klionsky DJ. Molecular

mechanisms of autophagy in the cardiovascular system. Circ Res.

;116(3):456-67. doi: 10.1161/circresaha.114.303788

Kouroumalis E, Tsomidis I, Voumvouraki A. Pathogenesis of

Hepatocellular Carcinoma: The Interplay of Apoptosis and

Autophagy. Biomedicines [Internet]. 2023[cited 2023 Aug

;11(4):1166. Available from: https://www.ncbi.nlm.nih.gov/

pmc/articles/PMC10135776/pdf/biomedicines-11-01166.pdf doi:

3390/biomedicines11041166

Wang B, Wang Y, Zhang J, Hu C, Jiang J, Li Y, et al. ROS-induced

lipid peroxidation modulates cell death outcome: mechanisms

behind apoptosis, autophagy, and ferroptosis. Arch Toxicol.

;97(6):1439-51. doi: 10.1007/s00204-023-03476-6

Gao L, Loveless J, Shay C, Teng Y. Targeting ROS-Mediated

Crosstalk Between Autophagy and Apoptosis in Cancer. Adv Exp

Med Biol. 2020;1260:1-12. doi: 10.1007/978-3-030-42667-5_1

Gao G, Chen W, Yan M, Liu J, Luo H, Wang C, et al. Rapamycin

regulates the balance between cardiomyocyte apoptosis and

autophagy in chronic heart failure by inhibiting mTOR signaling.

Int J Mol Med. 2020;45(1):195-209. doi: 10.3892/ijmm.2019.4407

Dong Y, Chen H, Gao J, Liu Y, Li J, Wang J. Molecular machinery

and interplay of apoptosis and autophagy in coronary heart

disease. J Mol Cell Cardiol. 2019;136:27-41. doi: 10.1016/j.

yjmcc.2019.09.001

Liu J, Liu W, Yang H. Balancing Apoptosis and Autophagy

for Parkinson’s Disease Therapy: Targeting BCL-2.

ACS Chem Neurosci. 2019;10(2):792-802. doi: 10.1021/

acschemneuro.8b00356

Kim D, Song J, Jin EJ. BNIP3-Dependent Mitophagy via PGC1α

Promotes Cartilage Degradation. Cells [Internet]. 2021[cited

Aug 15];10(7):1839. Available from: https://www.ncbi.nlm.

nih.gov/pmc/articles/PMC8304751/pdf/cells-10-01839.pdf doi:

3390/cells10071839

Celia AI, Colafrancesco S, Barbati C, Alessandri C, Conti

F. Autophagy in Rheumatic Diseases: Role in the Pathogenesis and

Therapeutic Approaches. Cells [Internet]. 2022[cited 2023 Aug

;11(8):1359. Available from: https://www.ncbi.nlm.nih.gov/

pmc/articles/PMC9025357/pdf/cells-11-01359.pdf doi: 10.3390/

cells11081359

Wang X, Wang L, Xiang W. Mechanisms of ovarian aging in

women: a review. J Ovarian Res [Internet]. 2023[cited 2023 Aug

;16(1):67. Available from: https://www.ncbi.nlm.nih.gov/pmc/

articles/PMC10080932/pdf/13048_2023_Article_1151.pdf doi:

1186/s13048-023-01151-z

Sukumaran P, Da Conceicao VТ, Sun Y, Ahamad N, Saraiva LК,

Selvaraj S, et al. Calcium Signaling Regulates Autophagy and

Apoptosis. Cells [Internet]. 2021[cited 2023 Aug 15];10(8):2125.

Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/

PMC8394685/pdf/cells-10-02125.pdf doi: 10.3390/cells10082125

Sezgin E, Gutmann T, Buhl T, Dirkx R, Grzybek M, Coskun

U, et al. Adaptive Lipid Packing and Bioactivity in Membrane

Domains. PLoS One [Internet]. 2015[cited 2023 Aug 19];10(4):

e0123930. Available from: https://www.ncbi.nlm.nih.gov/pmc/

articles/PMC4408024/pdf/pone.0123930.pdf doi: 10.1371/journal.

pone.0123930

Sezgin E, Levental I, Mayor S, Eggeling C. The mystery of membrane

organization: Composition, regulation and roles of lipid rafts. Nat

Rev Mol Cell Biol. 2017;18(6):361-74. doi: 10.1038/nrm.2017.16

Garofalo T, Manganelli V, Grasso M, Mattei V, Ferri A, Misasi

R, et al. Role of mitochondrial raft-like microdomains in the

regulation of cell apoptosis. Apoptosis. 2015;20(5):621-34. doi:

1007/s10495-015-1100-x

Manganelli V, Matarrese P, Antonioli M, Gambardella L,

Vescovo T, Gretzmeier C, et al. Raft-like lipid microdomains

drive autophagy initiation via AMBRA1-ERLIN1 molecular

association within MAMs. Autophagy. 2021;17(9):2528-48. doi:

1080/15548627.2020.1834207

Manganelli V, Longo A, Mattei V, Recalchi S, Riitano G,

Caissutti D, et al. Role of ERLINs in the Control of Cell Fate

through Lipid Rafts. Cells [Internet]. 2021[cited 2023 Aug

;10(9):2408. Available from: https://www.ncbi.nlm.nih.gov/

pmc/articles/PMC8470593/pdf/cells-10-02408.pdf doi: 10.3390/

cells10092408

Brunelli F, Torosantucci L, Gelmetti V, Franzone D, Grünewald

A, Krüger R, et al. PINK1 Protects against StaurosporineInduced Apoptosis by Interacting with Beclin1 and Impairing Its

Pro- Apoptotic Cleavage. Cells [Internet]. 2022[cited 2023 Aug

;11(4):678. Available from: https://www.ncbi.nlm.nih.gov/

pmc/articles/PMC8870463/pdf/cells-11-00678.pdf doi: 10.3390/

cells11040678

##submission.downloads##

Опубліковано

2023-09-23

Номер

Розділ

Статті