Next Article in Journal
CRISPR-Cas9 Technology for the Creation of Biological Avatars Capable of Modeling and Treating Pathologies: From Discovery to the Latest Improvements
Next Article in Special Issue
Aged G Protein-Coupled Receptor Kinase 3 (Grk3)-Deficient Mice Exhibit Enhanced Osteoclastogenesis and Develop Bone Lesions Analogous to Human Paget’s Disease of Bone
Previous Article in Journal
Label-Free Imaging Analysis of Patient-Derived Cholangiocarcinoma Organoids after Sorafenib Treatment
Previous Article in Special Issue
HIF-1α Regulates Bone Homeostasis and Angiogenesis, Participating in the Occurrence of Bone Metabolic Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Comment

Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882

1
Team ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ EA7270/Inserm, University Bourgogne Franche-Comté, 21000 Dijon, France
2
Lab-NAFS ‘Nutrition—Functional Food & Vascular Health’, Faculty of Medicine, University of Monastir, LR12ES05, Monastir 5000, Tunisia
3
Faculty of Sciences of Tunis, University Tunis-El Manar, Tunis 2092, Tunisia
4
Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
*
Author to whom correspondence should be addressed.
Cells 2022, 11(22), 3612; https://doi.org/10.3390/cells11223612
Submission received: 29 September 2022 / Revised: 7 November 2022 / Accepted: 11 November 2022 / Published: 15 November 2022
(This article belongs to the Special Issue Advances in Bone Metabolism)
Due to the increase in life span and life expectancy, which can, however, be more or less pronounced depending on the economic, social and cultural context [1,2], age-related diseases (cardiovascular, neurodegenerative and ocular diseases, osteoporosis and sarcopenia) are becoming more and more frequent, requiring adapted care structures and leading to an increase in individual and collective health expenses.
Therefore, a better understanding of the intrinsic and extrinsic factors that promote the transition from physiological to pathological aging is essential to prevent age-related diseases in order to make the best use of the additional time that longevity brings [3].
The results presented by Jing Ouyang et al. in “7-ketocholesterol induces oxiapoptophagy and inhibits osteogenic differentiation in MC3T3-E1” [4] are important in the context of the pathophysiology of age-related diseases mainly at three levels:
  • they reinforce the hypothesis that 7-ketocholesterol (7KC), essentially formed by cholesterol auto-oxidation [5], is a potential risk factor that could promote pathological aging and contribute to several age-related diseases [6,7,8] (Figure 1);
  • they confirm that 7KC can induce oxiapoptophagy, originally described in 2003 [9], involving oxidative stress, apoptosis and autophagy [10]; the signaling pathways associated with this type of death are well studied [11], which makes it possible to consider identifying specific therapeutic targets to prevent this type of cell death, which could occur in several age-related diseases. In addition, the in vitro study of oxiapoptophagy has permitted the identification of cytoprotective natural and synthethic molecules as well as a mixture of molecules (especially Mediterranean oils) that counteract 7KC-induced oxiapoptophagy and that can be of therapeutic and nutraceutic interest [12,13,14];
  • they provide new evidence of the potentially negative impact of 7KC in osteoporosis [15,16]; indeed, Jing Ouyang et al. show on mouse osteoblastic MCT3-E1 cells that 7KC decreases alkaline activity (ALP), used to judge osteogenic differentiation, reduces mineralization, and induces a decrease of OPN (osteopontin) and RUNX2 (Runt-2 transcription factor), two proteins involved in osteogenic differentiation [4].
It is worth noting that, at the moment, several in vitro studies have shown that natural and synthetic oxysterols act on osteogenesis by inhibiting or activating it, especially some oxysterols [17]. Deleterious effects on osteogenesis have already been observed with 7KC [16], cholestene-3β,5α,6β-triol and 27-hydroxycholesterol [18,19,20,21,22], which is an endogenous selective estrogen receptor modulator with side effects in bone. In contrast, 7α,25-hydroxycholesterol [23], 22S-hydroxycholesterol, 22R-hydroxycholesterol as well as 20S-hydroxycholesterol and some of its synthetic derivatives have bone-inducing properties [17,24]. As several oxysterols are simultaneously present in biological fluids and tissues, it would be necessary to define the oxysterol profiles in patients with osteoporosis and then to better define the activity of these oxysterols not only in vitro but also in vivo. While the use of a single oxysterol in cellular models is an essential step in specifying its biological activities and in identifying pharmacological targets and therapeutic molecules, appropriate mixtures of oxysterols (already used to address age-related or inflammatory bowel diseases) [25,26] could also be of interest to study osteoporosis in a more physiological context. Thus, to better understand the pathophysiology of osteoporosis, it seems first necessary to identify and quantify oxysterols in patients and then to address the biological activities of these molecules in appropriate cellular models. Studies on primary cultures, bone explants or organoids associated with microfluidic approaches using 7KC alone or in mixture could complement the results obtained on MC3T3-E1 and are now required.
In the context of cell death, the present study demonstrates again that oxiapoptophagy can concern different cell types from different species [10]. This type of cell death has often been described in the presence of 7KC, but also with other oxysterols (7β-hydroxycholesterol, 24S-hydroxycholesterol, 25-hydroxycholesterol, 7α,25-dihydrocholesterol, 5,6-epoxycholesterol), on human monocytic U937 cells [9], human JJN3 and U266 myeloma cells [27], bone marrow mesenchymal cells from patients with acute leukemia [28], L929 mouse fibroblast cells [29,30], and murine nerve cells (158N oligodendrocytes, microglial BV-2 cells, N2a neuronal cells) [31,32,33,34]. The study by Jing Ouyang et al. also clearly establishes an induction of oxiapoptophagy by 7KC on mouse osteoblastic MC3T3-E1 cells.
In addition, when MC3T3-E1 cells were cultured in the presence of 7KC and pre-treated with N-acetylcysteine (NAC) (a powerful antioxidant that was initially described as preventing 7KC-induced apoptosis on human U937 monocytic cells) [35], it was observed that NAC greatly reduces oxidative stress induced by 7KC and restores RedOx balance. Thus, the increases in the expression of NADPH-oxidase 4 (NOX4), the level of malonedialdehyde (MDA, a marker of lipid peroxidation) and the superoxide dismutase (SOD) activity were almost normalized. Apoptosis and autophagy were also greatly reduced. Therefore, oxidative stress plays a major role in the induction of 7KC-induced oxiapoptophagy on MC3T3-L1 cells, which is consistent with the results obtained on many other cells treated with 7KC [36]. It is important to underline that using natural or synthetic molecules, an inhibition of oxiapoptophagy has always been observed to act directly or indirectly on oxidative stress and normalize the RedOx balance [12].
Consequently, the data obtained by Jing Ouyang et al. brings additional evidence that supports the fact that 7KC could contribute (at least in part via oxiapoptophagy induction) to osteoporosis, which is a frequent age-related disease in post-menopausal women. These data constitute an additional in vitro proof of concept on the potential role of 7KC in age-related disease. There is now a need for in vivo evidence in appropriate animal models and in humans.

Author Contributions

Conceptualization, G.L.; writing—original draft preparation, G.L.; writing—review and editing, G.L., I.G., T.N., A.Z. and A.V.; supervision, G.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by: Université de Bourgogne (Dijon, France), University of Monastir (Monastir, Tunisia) and University Tunis El Manar (Tunis, Tunisia).

Conflicts of Interest

The authors declare no conflict of interest. The main data discussed can be found in the following research paper: [4].

References

  1. Vaupel, J.W.; Villavicencio, F.; Bergeron-Boucher, M.P. Demographic perspectives on the rise of longevity. Proc. Natl. Acad. Sci. USA 2021, 118, 132. [Google Scholar] [CrossRef]
  2. Aburto, J.M.; Villavicencio, F.; Basellini, U.; Kjærgaard, S.; Vaupel, J.W. Dynamics of life expectancy and life span equality. Proc. Natl. Acad. Sci. USA 2020, 117, 5250–5259. [Google Scholar] [CrossRef] [Green Version]
  3. Scott, A.J. The longevity society. Lancet Healthy Longev. 2021, 2, e820–e827. [Google Scholar] [CrossRef]
  4. Ouyang, J.; Xiao, Y.; Ren, Q.; Huang, J.; Zhou, Q.; Zhang, S.; Li, L.; Shi, W.; Chen, Z.; Wu, L. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882. [Google Scholar] [CrossRef]
  5. Anderson, A.; Campo, A.; Fulton, E.; Corwin, A.; Jerome, W.G., 3rd; O’Connor, M.S. 7-Ketocholesterol in disease and aging. Redox. Biol. 2020, 29, 101380. [Google Scholar] [CrossRef]
  6. Zarrouk, A.; Vejux, A.; Mackrill, J.; O’Callaghan, Y.; Hammami, M.; O’Brien, N.; Lizard, G. Involvement of oxysterols in age-related diseases and ageing processes. Ageing Res. Rev. 2014, 18, 148–162. [Google Scholar] [CrossRef] [PubMed]
  7. Samadi, A.; Sabuncuoglu, S.; Samadi, M.; Isikhan, S.Y.; Chirumbolo, S.; Peana, M.; Lay, I.; Yalcinkaya, A.; Bjørklund, G. A Comprehensive Review on Oxysterols and Related Diseases. Curr. Med. Chem. 2021, 28, 110–136. [Google Scholar] [CrossRef] [PubMed]
  8. Ali, J.; Aziz, M.A.; Rashid, M.M.O.; Basher, M.A.; Islam, M.S. Propagation of age-related diseases due to the changes of lipid peroxide and antioxidant levels in elderly people: A narrative review. Health Sci. Rep. 2022, 5, e650. [Google Scholar] [CrossRef]
  9. Monier, S.; Samadi, M.; Prunet, C.; Denance, M.; Laubriet, A.; Athias, A.; Berthier, A.; Steinmetz, E.; Jürgens, G.; Nègre-Salvayre, A.; et al. Impairment of the cytotoxic and oxidative activities of 7 beta-hydroxycholesterol and 7-ketocholesterol by esterification with oleate. Biochem. Biophys. Res. Commun. 2003, 303, 814–824. [Google Scholar] [CrossRef]
  10. Nury, T.; Zarrouk, A.; Yammine, A.; Mackrill, J.J.; Vejux, A.; Lizard, G. Oxiapoptophagy: A type of cell death induced by some oxysterols. Br. J. Pharm. 2021, 178, 3115–3123. [Google Scholar] [CrossRef] [PubMed]
  11. Vejux, A.; Abed-Vieillard, D.; Hajji, K.; Zarrouk, A.; Mackrill, J.J.; Ghosh, S.; Nury, T.; Yammine, A.; Zaibi, M.; Mihoubi, W.; et al. 7-Ketocholesterol and 7β-hydroxycholesterol: In vitro and animal models used to characterize their activities and to identify molecules preventing their toxicity. Biochem. Pharm. 2020, 173, 113648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Nury, T.; Yammine, A.; Ghzaiel, I.; Sassi, K.; Zarrouk, A.; Brahmi, F.; Samadi, M.; Rup-Jacques, S.; Vervandier-Fasseur, D.; Pais de Barros, J.P.; et al. Attenuation of 7-ketocholesterol- and 7β-hydroxycholesterol-induced oxiapoptophagy by nutrients, synthetic molecules and oils: Potential for the prevention of age-related diseases. Ageing Res. Rev. 2021, 68, 101324. [Google Scholar] [CrossRef] [PubMed]
  13. Brahmi, F.; Vejux, A.; Sghaier, R.; Zarrouk, A.; Nury, T.; Meddeb, W.; Rezig, L.; Namsi, A.; Sassi, K.; Yammine, A.; et al. Prevention of 7-ketocholesterol-induced side effects by natural compounds. Crit. Rev. Food Sci. Nutr. 2019, 59, 3179–3198. [Google Scholar] [CrossRef] [PubMed]
  14. Rezig, L.; Ghzaiel, I.; Ksila, M.; Yammine, A.; Nury, T.; Zarrouk, A.; Samadi, M.; Chouaibi, M.; Vejux, A.; Lizard, G. Cytoprotective activities of representative nutrients from the Mediterranean diet and of Mediterranean oils against 7-ketocholesterol- and 7β-hydroxycholesterol-induced cytotoxicity: Application to age-related diseases and civilization diseases. Steroids 2022, 187, 109093. [Google Scholar] [CrossRef]
  15. Vejux, A.; Malvitte, L.; Lizard, G. Side effects of oxysterols: Cytotoxicity, oxidation, inflammation, and phospholipidosis. Braz. J. Med. Biol. Res. 2008, 41, 545–556. [Google Scholar] [CrossRef] [Green Version]
  16. Sato, Y.; Ishihara, N.; Nagayama, D.; Saiki, A.; Tatsuno, I. 7-ketocholesterol induces apoptosis of MC3T3-E1 cells associated with reactive oxygen species generation, endoplasmic reticulum stress and caspase-3/7 dependent pathway. Mol. Genet. Metab. Rep. 2017, 10, 56–60. [Google Scholar] [CrossRef]
  17. Che, Y.; Yang, J.; Tang, F.; Wei, Z.; Chao, Y.; Li, N.; Li, H.; Wu, S.; Dong, X. New Function of Cholesterol Oxidation Products Involved in Osteoporosis Pathogenesis. Int. J. Mol. Sci. 2022, 23, 2020. [Google Scholar] [CrossRef]
  18. Liu, H.; Yuan, L.; Xu, S.; Wang, K.; Zhang, T. Cholestane-3beta,5alpha,6beta-triol inhibits osteoblastic differentiation and promotes apoptosis of rat bone marrow stromal cells. J. Cell Biochem. 2005, 96, 198–208. [Google Scholar] [CrossRef]
  19. DuSell, C.D.; Nelson, E.R.; Wang, X.; Abdo, J.; Mödder, U.I.; Umetani, M.; Gesty-Palmer, D.; Javitt, N.B.; Khosla, S.; McDonnell, D.P. The endogenous selective estrogen receptor modulator 27-hydroxycholesterol is a negative regulator of bone homeostasis. Endocrinology 2010, 151, 3675–3685. [Google Scholar] [CrossRef] [Green Version]
  20. Nelson, E.R.; DuSell, C.D.; Wang, X.; Howe, M.K.; Evans, G.; Michalek, R.D.; Umetani, M.; Rathmell, J.C.; Khosla, S.; Gesty-Palmer, D.; et al. The oxysterol, 27-hydroxycholesterol, links cholesterol metabolism to bone homeostasis through its actions on the estrogen and liver X receptors. Endocrinology 2011, 152, 4691–4705. [Google Scholar] [CrossRef]
  21. Nelson, E.R.; Wardell, S.E.; McDonnell, D.P. The molecular mechanisms underlying the pharmacological actions of estrogens, SERMs and oxysterols: Implications for the treatment and prevention of osteoporosis. Bone 2013, 53, 42–50. [Google Scholar] [CrossRef] [Green Version]
  22. Chang, P.Y.; Feldman, D.; Stefanick, M.L.; McDonnell, D.P.; Thompson, B.M.; McDonald, J.G.; Lee, J.S. 27-Hydroxycholesterol, an Endogenous SERM, and Risk of Fracture in Postmenopausal Women: A Nested Case-Cohort Study in the Women’s Health Initiative. J. Bone Min. Res. 2019, 34, 59–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nevius, E.; Pinho, F.; Dhodapkar, M.; Jin, H.; Nadrah, K.; Horowitz, M.C.; Kikuta, J.; Ishii, M.; Pereira, J.P. Oxysterols and EBI2 promote osteoclast precursor migration to bone surfaces and regulate bone mass homeostasis. J. Exp. Med. 2015, 212, 1931–1946. [Google Scholar] [CrossRef]
  24. Shouhed, D.; Kha, H.T.; Richardson, J.A.; Amantea, C.M.; Hahn, T.J.; Parhami, F. Osteogenic oxysterols inhibit the adverse effects of oxidative stress on osteogenic differentiation of marrow stromal cells. J. Cell Biochem. 2005, 95, 1276–1283. [Google Scholar] [CrossRef] [PubMed]
  25. Biasi, F.; Chiarpotto, E.; Sottero, B.; Maina, M.; Mascia, C.; Guina, T.; Gamba, P.; Gargiulo, S.; Testa, G.; Leonarduzzi, G.; et al. Evidence of cell damage induced by major components of a diet-compatible mixture of oxysterols in human colon cancer CaCo-2 cell line. Biochimie 2013, 95, 632–640. [Google Scholar] [CrossRef] [PubMed]
  26. Rossin, D.; Barbosa-Pereira, L.; Iaia, N.; Testa, G.; Sottero, B.; Poli, G.; Zeppa, G.; Biasi, F. A Dietary Mixture of Oxysterols Induces In Vitro Intestinal Inflammation through TLR2/4 Activation: The Protective Effect of Cocoa Bean Shells. Antioxidants 2019, 8, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Jaouadi, O.; Limam, I.; Abdelkarim, M.; Berred, E.; Chahbi, A.; Caillot, M.; Sola, B.; Ben Aissa-Fennira, F. 5,6-Epoxycholesterol Isomers Induce Oxiapoptophagy in Myeloma Cells. Cancers 2021, 13, 3747. [Google Scholar] [CrossRef]
  28. Paz, J.L.; Levy, D.; Oliveira, B.A.; de Melo, T.C.; de Freitas, F.A.; Reichert, C.O.; Rodrigues, A.; Pereira, J.; Bydlowski, S.P. 7-Ketocholesterol Promotes Oxiapoptophagy in Bone Marrow Mesenchymal Stem Cell from Patients with Acute Myeloid Leukemia. Cells 2019, 8, 151. [Google Scholar] [CrossRef] [Green Version]
  29. You, J.S.; Lim, H.; Seo, J.Y.; Kang, K.R.; Kim, D.K.; Oh, J.S.; Seo, Y.S.; Lee, G.J.; Kim, J.S.; Kim, H.J.; et al. 25-Hydroxycholesterol-Induced Oxiapoptophagy in L929 Mouse Fibroblast Cell Line. Molecules 2021, 27, 199. [Google Scholar] [CrossRef]
  30. Kim, J.S.; Lim, H.; Seo, J.Y.; Kang, K.R.; Yu, S.K.; Kim, C.S.; Kim, D.K.; Kim, H.J.; Seo, Y.S.; Lee, G.J.; et al. GPR183 Regulates 7α,25-Dihydroxycholesterol-Induced Oxiapoptophagy in L929 Mouse Fibroblast Cell. Molecules 2022, 27, 4798. [Google Scholar] [CrossRef]
  31. Nury, T.; Zarrouk, A.; Vejux, A.; Doria, M.; Riedinger, J.M.; Delage-Mourroux, R.; Lizard, G. Induction of oxiapoptophagy, a mixed mode of cell death associated with oxidative stress, apoptosis and autophagy, on 7-ketocholesterol-treated 158N murine oligodendrocytes: Impairment by α-tocopherol. Biochem. Biophys. Res. Commun. 2014, 446, 714–719. [Google Scholar] [CrossRef] [PubMed]
  32. Nury, T.; Zarrouk, A.; Mackrill, J.J.; Samadi, M.; Durand, P.; Riedinger, J.M.; Doria, M.; Vejux, A.; Limagne, E.; Delmas, D.; et al. Induction of oxiapoptophagy on 158N murine oligodendrocytes treated by 7-ketocholesterol-, 7β-hydroxycholesterol-, or 24(S)-hydroxycholesterol: Protective effects of α-tocopherol and docosahexaenoic acid (DHA; C22:6 n-3). Steroids 2015, 99, 194–203. [Google Scholar] [CrossRef] [PubMed]
  33. Debbabi, M.; Zarrouk, A.; Bezine, M.; Meddeb, W.; Nury, T.; Badreddine, A.; Karym, E.M.; Sghaier, R.; Bretillon, L.; Guyot, S.; et al. Comparison of the effects of major fatty acids present in the Mediterranean diet (oleic acid, docosahexaenoic acid) and in hydrogenated oils (elaidic acid) on 7-ketocholesterol-induced oxiapoptophagy in microglial BV-2 cells. Chem. Phys. Lipids 2017, 207, 151–170. [Google Scholar] [CrossRef]
  34. Yammine, A.; Zarrouk, A.; Nury, T.; Vejux, A.; Latruffe, N.; Vervandier-Fasseur, D.; Samadi, M.; Mackrill, J.J.; Greige-Gerges, H.; Auezova, L.; et al. Prevention by Dietary Polyphenols (Resveratrol, Quercetin, Apigenin) Against 7-Ketocholesterol-Induced Oxiapoptophagy in Neuronal N2a Cells: Potential Interest for the Treatment of Neurodegenerative and Age-Related Diseases. Cells 2020, 9, 2346. [Google Scholar] [CrossRef] [PubMed]
  35. Lizard, G.; Gueldry, S.; Sordet, O.; Monier, S.; Athias, A.; Miguet, C.; Bessede, G.; Lemaire, S.; Solary, E.; Gambert, P. Glutathione is implied in the control of 7-ketocholesterol-induced apoptosis, which is associated with radical oxygen species production. FASEB J. 1998, 12, 1651–1663. [Google Scholar] [CrossRef]
  36. Testa, G.; Rossin, D.; Poli, G.; Biasi, F.; Leonarduzzi, G. Implication of oxysterols in chronic inflammatory human diseases. Biochimie 2018, 153, 220–231. [Google Scholar] [CrossRef]
Figure 1. Involvement of 7-ketocholesterol (7KC) in age-related diseases [6]. The presented scheme is based on in vitro and/or in vivo proofs of concept.
Figure 1. Involvement of 7-ketocholesterol (7KC) in age-related diseases [6]. The presented scheme is based on in vitro and/or in vivo proofs of concept.
Cells 11 03612 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ghzaiel, I.; Nury, T.; Zarrouk, A.; Vejux, A.; Lizard, G. Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882. Cells 2022, 11, 3612. https://doi.org/10.3390/cells11223612

AMA Style

Ghzaiel I, Nury T, Zarrouk A, Vejux A, Lizard G. Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882. Cells. 2022; 11(22):3612. https://doi.org/10.3390/cells11223612

Chicago/Turabian Style

Ghzaiel, Imen, Thomas Nury, Amira Zarrouk, Anne Vejux, and Gérard Lizard. 2022. "Oxiapoptophagy in Age-Related Diseases. Comment on Ouyang et al. 7-Ketocholesterol Induces Oxiapoptophagy and Inhibits Osteogenic Differentiation in MC3T3-E1 Cells. Cells 2022, 11, 2882" Cells 11, no. 22: 3612. https://doi.org/10.3390/cells11223612

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop