Next Article in Journal
Hybrid Materials Obtained by Immobilization of Biosynthesized Ag Nanoparticles with Antioxidant and Antimicrobial Activity
Previous Article in Journal
A Clinical, Pharmacological, and Formulation Evaluation of Melatonin in the Treatment of Ocular Disorders—A Systematic Review
Previous Article in Special Issue
Mitochondria and Oxidative Stress as a Link between Alzheimer’s Disease and Diabetes Mellitus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Mitochondrial Dysfunction: A Common Trigger in Neurodegenerative and Metabolic Non-Communicable Diseases”

1
Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
2
Institute of Crystallography, Consiglio Nazionale delle Ricerche, Strada Provinciale 35d, n.9, Montelibretti, 00010 Rome, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(7), 4004; https://doi.org/10.3390/ijms25074004
Submission received: 9 January 2024 / Accepted: 26 February 2024 / Published: 3 April 2024
Non-communicable diseases (NCDs) are non-infectious and non-transmissible chronic disorders. They include cancer, neurodegenerative, autoimmune and cardiovascular diseases, as well as metabolic dysfunctions, such as diabetes and obesity. NCDs are characterized by low-grade inflammation and oxidative stress and are frequently associated with a marked mitochondrial dysfunction [1]. Mitochondrial functionality is the result of a fine-tuned balance between biogenesis, dynamics, and mitophagy [2,3,4]. The impairment of this quality control leads to the engulfment of cells in tissues and organs with dysfunctional organelles, causing their release into the cytosol and bloodstream of several mitochondrial molecules, collectively known as damage-associated molecular patterns (DAMPs) [5]. Mitochondrial DAMPs include proteins (TFAM and N-formylpeptides), lipids (cardiolipin), metabolites (succinate and ATP), and mitochondrial DNA [6]. They can activate the innate immune response, thereby contributing to the development of chronic inflammatory disorders [7].
This Special Issue, entitled “Mitochondrial Dysfunction: A Common Trigger in Neurodegenerative and Metabolic Non-Communicable Diseases” of the International Journal of Molecular Sciences, includes a total of seven contributions, composed of four research articles and three reviews. New information on the role of mitochondria and oxidative stress in both neurodegenerative and metabolic diseases, as well as in brain tumors, has been presented. The contribution of mitochondrial dysfunction has also been investigated in acute anoxia and preeclampsia.
Spermidine is an ubiquitary polyamine with well-known geroprotective properties. It can extend the health span and lifespan of different organisms, from fungi to mammals, and delay the onset of cardiovascular diseases and neurodegenerative disorders [8]. At the molecular level, spermidine seems to be a natural autophagy inducer [9]. Fairley and coworkers (contribution 1) investigated the specific role of spermidine on mitochondrial dysfunction induced by the deposition of abnormal hyper-phosphorylated tau protein, a microtubule protein. The hyper-phosphorylation of tau protein causes its dissociation from microtubules with the consequent formation of insoluble aggregates and neurofibrillary tangles, the hallmark of tauopathies, a class of human neurodegenerative diseases that includes Alzheimer’s disease (AD) and Parkinson’s disease. Using the human neuroblastoma SH-SY5Y cell line, a cellular model of tauopathies, the authors determined the effects of spermidine on the bioenergetics and cell metabolic activity versus the control counterpart, SH-SY5Y cells carrying the empty vector. They showed that spermidine improved mitochondrial oxidative phosphorylation, mitochondrial membrane potential, and ATP production in tau-expressing cells. Moreover, spermidine treatment decreased the level of free radicals, increased autophagy, and restored tau-induced impairments in mitophagy, inducing the gene expression of players involved in these mechanisms, lc3, p62, and parkin. The authors suggest that spermidine has potential therapeutic properties to counteract tau-related mitochondrial dysfunction.
Jastroch’s research group (contribution 2) investigated the role of the mitochondrial dynamics in glucose intake-dependent insulin secretion by pancreatic cells. It was already known that altered mitochondrial morphology, due to the different levels of expression of the genes involved in fusion and fission machinery, is associated with glucose-stimulated insulin secretion in β pancreatic cells. They investigated the crosstalk between mitochondrial dynamics and insulin secretion efficiency in pancreatic cells, studying the role of Drp1, the major regulator of mitochondrial fission. Unexpectedly, the overexpression of Drp1 in wild-type MIN6 pancreatic cells decreases insulin content and failed to rescue insulin secretion to wild-type levels in Drp1 knockdown cells. The authors suggest that the reduced insulin secretion observed in MIN6 cells is due to the Drp 1-dependent impairment of insulin biosynthesis by the activation of the PKA/eIF2α/Fgf21 pathway. The PKA/eIF2α/Fgf21 pathway is responsible for endoplasmic reticulum stress and consequently reduced protein translation. Therefore, the authors claimed that attention should be paid using the overexpression of Drp1 as a therapeutic tool, considering the side effects on insulin biosynthesis.
To understand whether mitochondrial morphological alterations and dysfunction are early or late events in brain malfunctions, Morozov and coworkers (contribution 3) investigated the structure and reorganization of both mitochondria and Golgi apparatus (GA) in embryonic mouse brain during acute anoxia. Analysis using electron microscopic three-dimensional reconstruction showed that the GA was the most vulnerable organelle, showing a clear deformation already after 1 h of anoxia, whereas the mitochondria maintained the normal ultrastructure. Only after 3 h of anoxia did the mitochondria assume an unconventional structure. The authors suggest that anoxia-induced GA disorganization may be responsible for the mitochondrial dysfunction observed at longer treatment times. Currently, this is the first study which suggests a potential link between GA phenotype and mitochondrial functionality.
Furthermore, the potential contribution of mitochondrial genome changes in brain tumor occurrence has been investigated by Kozakiewicz and coworkers (contribution 4). Genes encoding for respiratory chain components were analyzed by next-generation sequencing for DNA polymorphisms and mutations in blood and tumor biopsies of a cohort of 30 Caucasian patients, diagnosed with WHO grade II, III, or IV glioma. In particular, the consequences of missense mutations, able to alter the structure and biochemical properties of encoded proteins, were investigated by in silico studies. Only mitochondrial cytochrome b alterations are frequently found and seem to play a role in brain glioblastoma formation.
Xu and colleagues (contribution 5) thoroughly reviewed recent studies highlighting the crucial role of mitochondrial dysfunction in adipose tissue macrophages (ATMs) in obesity, a phenotype commonly associated with type 2 diabetes and insulin resistance. The hypertrophic expansion of adipose tissue creates local hypoxia and the release of fatty acids, leading to the accumulation of dysfunctional mitochondria in ATM. The impairment of mitochondrial quality control leads to the release of mitochondrial DAMPs responsible for the activation of the NLRP3 inflammasome with the subsequent release of proinflammatory cytokines. Interestingly, therapeutic strategies aimed at restoring homeostasis and reprogramming metabolism in ATM for the treatment of diabetes and insulin resistance have also been discussed.
Finally, Veselov et al. and Jahan and colleagues presented a growing body of evidence on the key role of mitochondrial dysfunction as a common trigger in AD and type 2 diabetes mellitus, as well as in the context of preeclampsia, highlighting how the loss of mitochondrial homeostasis can contribute to the onset and chronicity of apparently very distant NCD (contributions 6 and 7).
In conclusion, the contributions included in this Special Issue explore several aspects and different pathological conditions in which mitochondrial dysfunction seems to play a role and shed more light on many unresolved questions in this field. However, much remains to be done, mainly in terms of attempting to translate research findings into therapeutic approaches in neurodegenerative and metabolic NCDs.
Nevertheless, no studies in this Special Issue addressed microbiota–mitochondrial crosstalk. In recent years, several studies have strongly suggested a pivotal role of the microbial communities in numerous NCDs [10], and bidirectional crosstalk between gut microbiota and mitochondria has recently been discovered [11]. Gut microbiota by-products can modulate the gene expression levels of players with a role in the mitochondrial biogenesis and metabolic pathways of host cells, such as PGC-1α AMPk and SIRT1 [12]. On the other hand, the increase in mitochondrial ROS, due to oxidative phosphorylation impairment, leads to redox imbalance and gut barrier integrity loss [13]. Future research, focusing on the fine characterization of the molecular mechanisms involved in the interplay between mitochondria and microbiota in the host cells, could pave the way to new therapeutic approaches and should therefore be encouraged.

Author Contributions

Conceptualization, P.F. and B.P.; writing-original draft preparation, P.F. and B.P.; writing-review and editing, P.F. and B.P. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

List of Contributions

  • Fairley, L.H.; Lejri, I.; Grimm, A.; Eckert, A. Spermidine Rescues Bioenergetic and Mitophagy Deficits Induced by Disease-Associated Tau Protein. Int. J. Mol. Sci. 2023, 24, 5297.
  • Kabra, U.D.; Moruzzi, N.; Berggren, P.O.; Jastroch, M. Drp1 Overexpression Decreases Insulin Content in Pancreatic MIN6 Cells. Int. J. Mol. Sci. 2022, 23, 12338.
  • Morozov, Y.M.; Rakic P. Disorder of Golgi Apparatus Precedes Anoxia-Induced Pathology of Mitochondria. Int. J. Mol. Sci. 2022, 24, 4432.
  • Kozakiewicz, P.; Grzybowska-Szatkowska, L.; Ciesielka, M.; Całka, P.; Osuchowski, J.; Szmygin, P.; Jarosz, B.; Slaska, B. Mitochondrial DNA Changes in Genes of Respiratory Complexes III, IV and V Could Be Related to Brain Tumours in Humans. Int. J. Mol. Sci. 2022, 23, 12131.
  • Xu, L.; Yan, X.; Zhao, Y.; Wang, J.; Liu, B.; Yu, S.; Fu, J.; Liu, Y.; Su, J. Macrophage Polarization Mediated by Mitochondrial Dysfunction Induces Adipose Tissue Inflammation in Obesity. Int. J. Mol. Sci. 2022, 23, 9252.
  • Veselov, I.M.; Vinogradova, D.V.; Maltsev, A.V.; Shevtsov, P.N.; Spirkova, E.A.; Bachurin, S.O.; Shevtsova E.F. Mitochondria and Oxidative Stress as a Link between Alzheimer’s Disease and Diabetes Mellitus. Int. J. Mol. Sci. 2023, 24, 14450.
  • Jahan, F.; Vasam, G.; Green, A.E.; Bainbridge, S.A.; Menzies, K.J. Placental Mitochondrial Function and Dysfunction in Preeclampsia. Int. J. Mol. Sci. 2023, 24, 4177.

References

  1. Peña-Oyarzun, D.; Bravo-Sagua, R.; Diaz-Vega, A.; Aleman, L.; Chiong, M.; Garcia, L.; Bambs, C.; Troncoso, R.; Cifuentes, M.; Morselli, E.; et al. Autophagy and oxidative stress in non communicable diseases: A matter of the inflammatory state? Free Radic. Biol. Med. 2018, 124, 61–78. [Google Scholar] [CrossRef] [PubMed]
  2. Ploumi, C.; Daskalaki, I.; Tavernarakis, N. Mitochondrial biogenesis and clearance: A balancing act. FEBS J. 2017, 284, 183–195. [Google Scholar] [CrossRef] [PubMed]
  3. Chan, D.C. Mitochondrial fusion and fission in mammals. Annu. Rev. Cell Dev. Biol. 2006, 22, 79–99. [Google Scholar] [CrossRef] [PubMed]
  4. Pickles, S.; Vigié, P.; Youle, R.J. Mitophagy and quality control mechanisms in mitochondrial maintenance. Curr. Biol. 2018, 28, R170–R185. [Google Scholar] [CrossRef] [PubMed]
  5. Dela Cruz, C.S.; Kang, M.J. Mitochondrial dysfunction and damage associated molecular patterns (DAMPs) in chronic inflammatory diseases. Mitochondrion 2018, 41, 37–44. [Google Scholar] [CrossRef] [PubMed]
  6. Lin, M.M.; Liu, N.; Qin, Z.; Wang, Y. Mitochondrial-derived damage-associated molecular patterns amplify neuroinflammation in neurodegenerative diseases. Acta Pharmacol. Sin. 2022, 43, 2439–2447. [Google Scholar] [CrossRef] [PubMed]
  7. Picca, A.; Lezza, A.M.S.; Leeuwenburgh, C.; Pesce, V.; Calvani, R.; Landi, F.; Bernabei, R.; Marzetti, E. Fueling inflamm-aging through mitochondrial dysfunction: Mechanisms and molecular targets. Int. J. Mol. Sci. 2017, 18, 933. [Google Scholar] [CrossRef] [PubMed]
  8. Madeo, F.; Eisenberg, T.; Pietrocola, F.; Kroemer, G. Spermidine in health and disease. Science 2018, 26, eaan2788. [Google Scholar] [CrossRef] [PubMed]
  9. Eisenberg, T.; Knauer, H.; Schauer, A.; Büttner, S.; Ruckenstuhl, C.; Carmona-Gutierrez, D.; Ring, J.; Schroeder, S.; Magnes, C.; Antonacci, L.; et al. Induction of autophagy by spermidine promotes longevity. Nat. Cell Biol. 2009, 11, 1305–1314. [Google Scholar] [CrossRef] [PubMed]
  10. Huang, Z.; Li, Y.; Park, H.; Ho, M.; Bhardwaj, K.; Sugimura, N.; Won Lee, H.; Meng, H.; Ebert, M.P.; Chao, K.; et al. Unveiling and harnessing the human gut microbiome in the rising burden of non-communicable diseases during urbanization. Gut Microbes 2023, 15, 2237645. [Google Scholar] [CrossRef] [PubMed]
  11. Chartoumpekis, D.V.; Zaravinos, A.; Apidianakis, Y.; Lagoumintzis, G. Microbiota and mitochondria: Impact on cell signaling, physiology, and disease. Front. Microbiol. 2022, 13, 1056499. [Google Scholar] [CrossRef] [PubMed]
  12. Mollica, M.P.; Mattace Raso, G.; Cavaliere, G.; Trinchese, G.; De Filippo, C.; Aceto, S.; Prisco, M.; Pirozzi, C.; Di Guida, F.; Lama, A.; et al. Butyrate Regulates Liver Mitochondrial Function, Efficiency, and Dynamics in Insulin-Resistant Obese Mice. Diabetes 2017, 66, 1405. [Google Scholar] [CrossRef] [PubMed]
  13. Ballard, J.W.O.; Towarnicki, S.G. Mitochondria, the gut microbiome and ROS. Cell. Signal. 2020, 75, 109737. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fortini, P.; Pascucci, B. Special Issue “Mitochondrial Dysfunction: A Common Trigger in Neurodegenerative and Metabolic Non-Communicable Diseases”. Int. J. Mol. Sci. 2024, 25, 4004. https://doi.org/10.3390/ijms25074004

AMA Style

Fortini P, Pascucci B. Special Issue “Mitochondrial Dysfunction: A Common Trigger in Neurodegenerative and Metabolic Non-Communicable Diseases”. International Journal of Molecular Sciences. 2024; 25(7):4004. https://doi.org/10.3390/ijms25074004

Chicago/Turabian Style

Fortini, Paola, and Barbara Pascucci. 2024. "Special Issue “Mitochondrial Dysfunction: A Common Trigger in Neurodegenerative and Metabolic Non-Communicable Diseases”" International Journal of Molecular Sciences 25, no. 7: 4004. https://doi.org/10.3390/ijms25074004

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