Download PDF
Commentary  |  Open Access  |  10 Jun 2024

Mitochondria-derived vesicles: potential nano-batteries to recharge the cellular powerhouse

Views: 158 |  Downloads: 26 |  Cited:  0
Extracell Vesicles Circ Nucleic Acids 2024;5:271-5.
10.20517/evcna.2023.71 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Mitochondria dysfunction is increasingly recognized as a critical factor in various pathogenic processes. The mechanism governing mitochondrial quality control serves as an adaptive response, ensuring the preservation of mitochondrial morphology, quantity, and overall function, crucial for cell survival. The generation of mitochondria-derived vesicles (MDVs) is one of the processes of mitochondrial quality control. Recent literature has suggested MDV heterogeneity; however, the detailed characteristics of various MDV subtypes still need to be studied better. Recent studies have shown that MDVs also play a role in inter-organelle communication for mitochondria besides quality control. For instance, Hazan et al. demonstrated that functional mitochondria from Saccharomyces cerevisiae release vesicles independent of the fission machinery. These vesicles, falling within the typical size range of MDVs, were selectively loaded with mitochondrial proteins, especially with functional ATP synthase subunits. Intriguingly, these MDVs maintained membrane potential and could generate ATP. Moreover, MDVs could fuse with naïve mitochondria, transferring their ATP generation machinery. Lastly, this study revealed a potential delivery mechanism of ATP-producing vesicles, presenting a promising avenue to rejuvenate ATP-deficient mitochondria. Overall, this study unveils a novel mechanism for inter-organelle communication by vesicles, which is crucial for maintaining cellular homeostasis and could also be important in pathological conditions.

Keywords

Mitochondria, mitochondria-derived vesicle, inter-organelle communication, F1-F0 ATP synthase

MAIN TEXT

Mitochondria, a double membrane-bound organelle, regulate a plethora of cellular functions, encompassing oxidative ATP production, β-oxidation, calcium signaling, and apoptosis. Mitochondria crucially influence energy metabolism, redox status, molecular signaling, and cell survival and death. Owing to their crucial involvement in multiple cellular processes, mitochondrial damage has been considered one of the main driving factors for diseases such as aging-associated pathologies, neurodegenerative diseases, cancer, metabolic diseases, etc.[1-5]. It is emerging that under both normal physiological conditions and stress situations, mitochondria could communicate with other organelles within the cell and with organelles of other cells. One possible mode of this communication is via mitochondria-derived vesicles (MDVs)[6-8]. MDVs are single or double-membrane vesicles generated either from the outer mitochondrial membrane or from the inner and outer mitochondrial membrane with a portion of the mitochondrial matrix, respectively[9-11]. MDVs are characterized by these three major criteria: (i) a relatively uniform diameter ranging from 70-150 nm; (ii) biogenesis independent of the mitochondrial fission protein Drp1 (Dnm1 in yeast); and (iii) selective incorporation of protein cargo. While MDV generation is a basal housekeeping process under normal physiological settings, it escalates during pathological circumstances[7,8,12] and is reported to gradually decline with aging[13].

MDVs are highly heterogeneous and their heterogeneity is mainly dependent upon cell type secreting these vesicles and the processes in which these vesicles are involved. Biogenesis of MDVs requires the recruitment of either PINK1/Parkin or Ras-related protein (Rab9) and sorting Nexin 9 (SNX9) or β-barrel protein, mitochondrial-anchored protein ligase (MAPL), and translocase of the outer mitochondrial membrane complex subunit 20 (TOM20)[6,14-16]. Initially, MDVs were thought of as vesicles generated in response to oxidative stress as a mitochondrial quality control process where mitochondria package their oxidized cargo in these vesicles and shuttle to the lysosome for degradation, thereby regulating mitochondrial mass more rapidly than mitophagy[17,18]. However, recent studies have shown that alteration in MDV generation and release is associated with various pathological conditions such as neurodegeneration, cardiomyocyte damage, accelerated aging, autoimmune diseases, cancer, and infections[10,19-22]. During Parkinson’s disease, circulating extracellular vesicles include MDVs, as indicated by the identification of mitochondrial signatures, which could be utilized as biomarkers for disease severity[23,24]. On the other hand, Ramirez et al. have shown that cannabidiol increases MDV generation by activating mitochondrial permeability transition pore (mPTP) opening[25], thereby advocating that protective MDV formation could potentially serve as a new therapeutic target for neurodegenerative diseases associated with mitochondrial dysfunction. Recently, Li et al. showed that MDVs protect cardiomyocytes by delivering Bcl-2 to severely damaged mitochondria under hypoxic stimulation[12]. Picca et al. showed that in older population with physical frailty and sarcopenia, circulating extracellular vesicles include MDVs, as indicated by the identification of mitochondrial signatures among them, and their secretion reduced despite the increase in the small EV secretion, indicating mitochondrial dysfunction[26]. Although, in recent years, our understanding of the potential role of MDVs in normal physiology and under disease conditions has significantly improved, there is still a significant gap in understanding their characteristics and functions, especially their role in inter-organelle communication. Further, given the recent research focus on understanding mitochondrial function in various diseases, there is a pressing need to better comprehend MDVs’ role in maintaining cellular homeostasis. A better understanding of the mechanistic framework of MDV could also aid in developing novel therapeutic approaches for treating various diseases.

A recent study by Hazan et al. meticulously characterized MDVs from the isolated functional mitochondria of yeast (strain: Saccharomyces cerevisiae)[27]. MDVs were isolated by filtration and differential centrifugation and characterized for several known basic characteristics of MDVs[27]. It was demonstrated that MDVs were composed of single or potentially double membranes, and their size ranged from 80-200 nm with a peak at 105.8 ± 2.2 nm, corresponding to typical MDV size[27]. Further, no change in the MDV secretion profile was observed in Dnm1 knockout cells, confirming that MDV secretion is independent of mitochondrial fission[27]. Interestingly, only functional mitochondria were involved in MDV secretion, and over time, as mitochondria lost their ATP generation capacity, MDV concentration decreased and stabilized at a very low level at 48 h[27]. This study also identified the selectivity of MDV protein cargo by LC/MS/MS analysis[27]. Furthermore, a comparison between mitochondrial and MDVs fractions showed distinct protein distribution in MDV[27]. MDVs were more enriched for outer membrane proteins, while mitochondrial ribosomal proteins were relatively less abundant in MDVs compared to their levels in the mitochondrial fractions[27]. Additionally, specific enrichment of a few proteins was noted in MDVs[27]. For example, isocitrate lyase 1 (Icl-1) was noticeable in MDVs but was present in negligible amounts in the mitochondrial fraction. In short, this study provided compelling evidence for the specific cargo distribution in MDVs. Notably, this study also demonstrated the role of specific cargo proteins in MDV formation. Specifically, malate dehydrogenase 1 (Mdh1), a mitochondrial matrix TCA (tricarboxylic acid) enzyme, and Om45, an outer mitochondrial membrane protein, both were identified in the MDVs, and knockout of these proteins in cells resulted in decreased concentration of MDVs. Overall, this study nicely demonstrated the three major characteristics associated with MDVs, as mentioned above.

Intriguingly, the study also provided in vitro evidence about the functional importance of MDVs. Mass spectrometry data suggested the presence of multiple subunits of the F1F0-ATP synthase complex, with 13 out of 17 subunits of F1F0-ATP synthase present in MDVs[27]. Furthermore, MDVs showed membrane potential and retained significant ATP production capacity[27]. Notably, when incubated with Atp2 (ATP synthase subunit beta) respiratory deficient mitochondria, wild-type MDVs increased ATP production by 560%, while incubation with wild-type mitochondria increased ATP production by 30%. Importantly, this study also provided evidence that MDVs are incorporated into mitochondria following uptake and lend their ATP production machinery to the recipient mitochondria. Notably, a few significant observations in the yeast model were validated in MDVs from isolated mitochondria derived from HEK293 cells. Taken together, this study suggested that MDVs are one of the components of a complex cascade of mitochondrial function, which is not only involved in the quality control of mitochondria but also in the rejuvenation of damaged mitochondria, through the transfer of MDVs between healthy and damaged mitochondria.

Further studies are required to understand the potential of MDVs as a biomarker of pathophysiological diseases where mitochondrial dysfunction is the major driving factor. For clinical translation, reliable methods to isolate MDVs from various biofluids need to be developed further. Until now, the only method to collect pure and specific MDVs has been from isolated mitochondria; however, this approach is not feasible for large clinical setups, especially when dealing with archived biofluid samples. To isolate MDVs from biofluids, a better understanding of unique or specific proteins on the surface of MDVs is requisite.

In conclusion, Hazan et al. have presented a thought-provoking approach to better understanding MDVs’ physical characteristics and functional role. Further improving our understanding of the MDV biogenesis biology and their role in various pathological conditions could not only provide us with new prognostic/diagnostic markers but also emerge as a new therapeutic approach, e.g., utilizing MDVs to rejuvenate and restore damaged mitochondria.

DECLARATIONS

Author’s contribution

Conceptualized and wrote the manuscript: Mishra S, Deep G

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

Gagan Deep is the founder of LiBiCo, which has no influence or contribution to the work presented in this manuscript.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024

REFERENCES

1. Chistiakov DA, Sobenin IA, Revin VV, Orekhov AN, Bobryshev YV. Mitochondrial aging and age-related dysfunction of mitochondria. Biomed Res Int 2014;2014:238463.

2. Lima T, Li TY, Mottis A, Auwerx J. Pleiotropic effects of mitochondria in aging. Nat Aging 2022;2:199-213.

3. Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2017;1863:1066-77.

4. Javadov S, Kozlov AV, Camara AKS. Mitochondria in health and diseases. Cells 2020;9:1177.

5. Klein K, He K, Younes AI, et al. Role of mitochondria in cancer immune evasion and potential therapeutic approaches. Front Immunol 2020;11:573326.

6. Popov LD. Mitochondrial-derived vesicles: recent insights. J Cell Mol Med 2022;26:3323-8.

7. Peng T, Xie Y, Sheng H, Wang C, Lian Y, Xie N. Mitochondrial-derived vesicles: gatekeepers of mitochondrial response to oxidative stress. Free Radic Biol Med 2022;188:185-93.

8. Picca A, Guerra F, Calvani R, et al. Mitochondrial-derived vesicles: the good, the bad, and the ugly. Int J Mol Sci 2023;24:13835.

9. Sugiura A, McLelland GL, Fon EA, McBride HM. A new pathway for mitochondrial quality control: mitochondrial-derived vesicles. EMBO J 2014;33:2142-56.

10. Heyn J, Heuschkel MA, Goettsch C. Mitochondrial-derived vesicles-link to extracellular vesicles and implications in cardiovascular disease. Int J Mol Sci 2023;24:2637.

11. Soubannier V, Rippstein P, Kaufman BA, Shoubridge EA, McBride HM. Reconstitution of mitochondria derived vesicle formation demonstrates selective enrichment of oxidized cargo. PloS one 2012;7:e52830.

12. Li B, Zhao H, Wu Y, et al. Mitochondrial-derived vesicles protect cardiomyocytes against hypoxic damage. Front Cell Dev Biol 2020;8:214.

13. Guo Y, Guan T, Yu Q, et al. ALS-linked SOD1 mutations impair mitochondrial-derived vesicle formation and accelerate aging. Redox Biol 2024;69:102972.

14. König T, Nolte H, Aaltonen MJ, et al. MIROs and DRP1 drive mitochondrial-derived vesicle biogenesis and promote quality control. Nat Cell Biol 2021;23:1271-86.

15. Braschi E, Goyon V, Zunino R, Mohanty A, Xu L, McBride HM. Vps35 mediates vesicle transport between the mitochondria and peroxisomes. Curr Biol 2010;20:1310-5.

16. Matheoud D, Sugiura A, Bellemare-Pelletier A, et al. Parkinson's disease-related proteins PINK1 and parkin repress mitochondrial antigen presentation. Cell 2016;166:314-27.

17. Cadete VJ, Deschênes S, Cuillerier A, et al. Formation of mitochondrial-derived vesicles is an active and physiologically relevant mitochondrial quality control process in the cardiac system. J Physiol 2016;594:5343-62.

18. Soubannier V, McLelland GL, Zunino R, et al. A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr Biol 2012;22:135-41.

19. König T, McBride HM. Mitochondrial-derived vesicles in metabolism, disease, and aging. Cell Metab 2024;36:21-35.

20. Abuaita BH, Schultz TL, O'Riordan MX. Mitochondria-derived vesicles deliver antimicrobial reactive oxygen species to control phagosome-localized staphylococcus aureus. Cell Host Microbe 2018;24:625-636.e5.

21. Picca A, Guerra F, Calvani R, et al. Generation and release of mitochondrial-derived vesicles in health, aging and disease. J Clin Med 2020;9:1440.

22. Gagliardi S, Mitruccio M, Di Corato R, et al. Defects of mitochondria-lysosomes communication induce secretion of mitochondria-derived vesicles and drive chemoresistance in ovarian cancer cells. Cell Commun Signal 2024;22:165.

23. Picca A, Guerra F, Calvani R, et al. Mitochondrial-derived vesicles as candidate biomarkers in Parkinson’s disease: rationale, design and methods of the EXosomes in PArkiNson disease (EXPAND) study. Int J Mol Sci 2019;20:2373.

24. Picca A, Guerra F, Calvani R, et al. Mitochondrial signatures in circulating extracellular vesicles of older adults with parkinson's disease: results from the EXosomes in PArkiNson's disease (EXPAND) study. J Clin Med 2020;9:504.

25. Ramirez A, Old W, Selwood DL, Liu X. Cannabidiol activates PINK1-parkin-dependent mitophagy and mitochondrial-derived vesicles. Eur J Cell Biol 2022;101:151185.

26. Picca A, Beli R, Calvani R, et al. Older adults with physical frailty and sarcopenia show increased levels of circulating small extracellular vesicles with a specific mitochondrial signature. Cells 2020;9:973.

27. Hazan Ben-Menachem R, Lintzer D, Ziv T, et al. Mitochondrial-derived vesicles retain membrane potential and contain a functional ATP synthase. EMBO Rep 2023;24:e56114.

Cite This Article

Export citation file: BibTeX | EndNote | RIS

OAE Style

Mishra S, Deep G. Mitochondria-derived vesicles: potential nano-batteries to recharge the cellular powerhouse. Extracell Vesicles Circ Nucleic Acids 2024;5:271-5. http://dx.doi.org/10.20517/evcna.2023.71

AMA Style

Mishra S, Deep G. Mitochondria-derived vesicles: potential nano-batteries to recharge the cellular powerhouse. Extracellular Vesicles and Circulating Nucleic Acids. 2024; 5(2): 271-5. http://dx.doi.org/10.20517/evcna.2023.71

Chicago/Turabian Style

Shalini Mishra, Gagan Deep. 2024. "Mitochondria-derived vesicles: potential nano-batteries to recharge the cellular powerhouse" Extracellular Vesicles and Circulating Nucleic Acids. 5, no.2: 271-5. http://dx.doi.org/10.20517/evcna.2023.71

ACS Style

Mishra, S.; Deep G. Mitochondria-derived vesicles: potential nano-batteries to recharge the cellular powerhouse. Extracell. Vesicles. Circ. Nucleic. Acids. 2024, 5, 271-5. http://dx.doi.org/10.20517/evcna.2023.71

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
158
Downloads
26
Citations
0
Comments
0
1

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Extracellular Vesicles and Circulating Nucleic Acids
ISSN 2767-6641 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/