Next Article in Journal
Cytoprotective Effects of Delphinidin for Human Chondrocytes against Oxidative Stress through Activation of Autophagy
Next Article in Special Issue
Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration
Previous Article in Journal
Acknowledgement to Reviewers of Antioxidants in 2019
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitoprotective Clinical Strategies in Type 2 Diabetes and Fanconi Anemia Patients: Suggestions for Clinical Management of Mitochondrial Dysfunction

by
Giovanni Pagano
1,*,
Federico V. Pallardó
2,
Beatriz Porto
3,
Maria Rosa Fittipaldi
4,
Alex Lyakhovich
5,6 and
Marco Trifuoggi
1
1
Department of Chemical Sciences, Federico II Naples University, I-80126 Naples, Italy
2
Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, CIBERER, E-46010 Valencia, Spain
3
Institute of Biomedical Sciences, ICBAS, University of Porto, 4099-030 Porto, Portugal
4
Internal Medicine Unit, San Francesco d’Assisi Hospital, I-84020 Oliveto Citra (SA), Italy
5
Vall d’Hebron Institut de Recerca, E-08035 Barcelona, Spain
6
Institute of Molecular Biology and Biophysics of the “Federal Research Center of Fundamental and Translational Medicine”, 630117 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Antioxidants 2020, 9(1), 82; https://doi.org/10.3390/antiox9010082
Submission received: 17 December 2019 / Revised: 14 January 2020 / Accepted: 16 January 2020 / Published: 18 January 2020
(This article belongs to the Special Issue Oxidative Stress and Rare Diseases)

Abstract

:
Oxidative stress (OS) and mitochondrial dysfunction (MDF) occur in a number of disorders, and several clinical studies have attempted to counteract OS and MDF by providing adjuvant treatments against disease progression. The present review is aimed at focusing on two apparently distant diseases, namely type 2 diabetes (T2D) and a rare genetic disease, Fanconi anemia (FA). The pathogenetic links between T2D and FA include the high T2D prevalence among FA patients and the recognized evidence for OS and MDF in both disorders. This latter phenotypic/pathogenetic feature—namely MDF—may be regarded as a mechanistic ground both accounting for the clinical outcomes in both diseases, and as a premise to clinical studies aimed at counteracting MDF. In the case for T2D, the working hypothesis is raised of evaluating any in vivo decrease of mitochondrial cofactors, or mitochondrial nutrients (MNs) such as α-lipoic acid, coenzyme Q10, and l-carnitine, with possibly combined MN-based treatments. As for FA, the established knowledge of MDF, as yet only obtained from in vitro or molecular studies, prompts the requirement to ascertain in vivo MDF, and to design clinical studies aimed at utilizing MNs toward mitigating or delaying FA’s clinical progression. Altogether, this paper may contribute to building hypotheses for clinical studies in a number of OS/MDF-related diseases.

1. Introduction

Redox and mitochondrial anomalies are recognized—since the pioneering report by Luft (1994) [1] on “mitochondrial medicine”—in an extensive number of disorders that are afferent to several medical disciplines [2,3,4,5], including diabetes, aging, and genetic, neurologic and neuropsychiatric, and cardio-vascular diseases. Thus, several clinical studies have attempted, with varying success, to provide adjuvant treatments aimed at counteracting OS/MDF-related disorders by means of essential mitochondrial cofactors [6,7]. As stated by Wesselink et al. [6], mitochondria-focused treatments can provide basic support in contrasting disease progression in a number of OS/MDF-related disorders. The recognized key molecules in mitochondrial functions are α-lipoic acid (ALA), coenzyme Q10 (coQ10), and carnitine (CARN), also termed mitochondrial nutrients (MNs) [8]. These are involved in three basic mitochondrial functions, namely the Krebs cycle (ALA), the electron transport chain (coQ10), and acyl transfer (CARN), as recognized since the early report by Palade in 1964 [9].
Mitochondria’s essential tasks rely on oxidative metabolism of nutrients in producing high-energy molecules such as ATP, thus reactive oxygen species (ROS) are a necessary by-product of these biotransformations, causing an intramitochondrial pro-oxidant state. Hence, mitochondria are commonly regarded as the main cell’s “power plant” and the first victim of this pro-oxidant state. As a lucky evolutive solution for mitochondrial—and cellular—survival, MNs both exert their cofactor roles in energetic—pro-oxidant—metabolism and behave as strong antioxidants [10,11,12].
Among the extensive number of OS/MDF-related disorders [2], the present review is focused on two apparently unrelated diseases, such as a pandemic disease as T2D and a rare genetic disease as FA. These may be seen as two paradigms both offering mechanistic insights of the two disorders and providing working hypotheses toward adjuvant treatments of these and other OS/MDF-related disorders.

2. Type 2 Diabetes

The involvement of OS in T2D pathogenesis has been demonstrated in the last 30 years [13,14,15,16,17]. The mechanistic roles of OS and MDF in T2D pathogenesis both include the primitive damage to pancreatic beta cells [18,19] and the major clinical consequences of T2D progression, such as micro- and macro-angiopathies, and retinal damage [20,21].
As a concurrent feature of T2D-associated prooxidant state, defective respiration and oxidative phosphorylation was reported since early studies of diabetic patients and in animal studies. At the purpose to counteract T2D-associated OS, the use of several antioxidants was suggested [22,23,24,25,26,27,28,29,30]. Thereafter, and up to recent studies, MDF was further investigated as a salient feature of T2D, while a mechanistic role of the antidiabetic drug metformin, and of an herbal antidiabetic formulation, was associated with improvement of mitochondrial function [31,32,33].
In view of the recognized roles of MNs in mitochondrial functions, a body of literature has focused on the beneficial outcomes of treating diabetic patients with each MN, leading to clinical improvements following patients’ treatment with ALA, or coQ10, or CARN, as summarized in Table 1 (reviewed in [7]). Since early clinical studies in the 1990s, ALA has been demonstrated to provide adjuvant support in ameliorating the clinical conditions of T2D patients [34,35,36,37,38,39,40,41,42,43,44,45,46,47,48], with the recognition of ALA as a prescription drug in diabetic patients by the German Drug Index [49].
Other clinical studies have investigated the adjuvant effects of either coQ10 [50,51,52,53,54,55] or CARN (or acyl-CARN) [56,57,58,59,60,61,62,63,64,65] in T2D patients, by improving blood pressure and glycemic control.
Unlike the above clinical trials, only utilizing one MN, a pilot study by Palacka et al. in 2010 [66] reported on the adjuvant treatment of T2D patients by administering limited doses of ALA (100 mg/day) and of coQ10 (60 mg/day). To the best of our knowledge, no other clinical studies focused on two-MN administration in T2D patients, and no report was found reporting on triple MN administration. Nevertheless, it should be mentioned that MN combinations were reported in either in vitro or in animal studies aimed at counteracting MDF and OS in experimental diabetes or in other MDF-related disorders [67,68,69,70,71,72,73]. This was the case, e.g., for the studies by Shen et al. [69,70] reporting on combined ALA and CARN mixture-induced protective effects in rat or murine cells; MN mixtures were also found to protect diabetic rats from immuno- and liver-dysfunction [72,73].
Based on the available knowledge of safe and successful use of each MN in T2D, and in several other disorders, one may suggest that safe administration of triple MN combinations might provide successful outcomes in counteracting T2D-related symptoms and progression. This working hypothesis warrants design of ad hoc experimental and clinical studies.

3. Fanconi Anemia

According to an extensive consensus in the scientific community, FA was defined by Grover Bagby in 2018 [74] as a «rare inherited syndrome characterized by progressive bone marrow failure (BMF) and a high relative risk of hematopoietic and epithelial malignancies in children who sometimes have characteristic developmental abnormalities, including short stature and radial ray defects. Since the first clinical report of this disease 90 years ago, important clinical advances have included the development of the gold-standard diagnostic test (chromosomal breakage responses when lymphocytes or fibroblasts are exposed to low doses of either mitomycin C or diepoxybutane).
Beyond the “canonical” definition linking FA to defective DNA repair [74], three major features of FA phenotype are herein discussed, both in the attempt to understand FA pathogenesis and to develop improvements in the clinical management of FA patients. These phenotypic features include excess T2D prevalence in FA patients, the in vitro/in vivo evidence for OS, and the occurrence of MDF, though so far confined to in vitro and molecular investigations.

3.1. Excess T2D Prevalence

An excess T2D prevalence is a well-established phenotypic feature in FA patients [75,76,77], as assessed by the International Fanconi Anemia Registry [75], along with excess prevalence (81%) of multiple endocrine abnormalities. This finding was confirmed by further studies [76,77], both in FA patients and in FA mice [78]. These links between FA and T2D are consistent with shared pathogenetic mechanisms, first including the role(s) of OS both in T2D and in FA. It is worth noting that excess T2D prevalence is not confined to FA, as far as a number of other genetic diseases [79,80,81,82,83] display excess T2D prevalence (along with OS features), independently of their multiple and different genetic deficiencies. Thus, the co-occurrence of T2D in these disorders suggests common deficiency(ies) focused on OS/MDF abnormalities.

3.2. FA: In Vitro and In Vivo OS

The occurrence of redox abnormalities in FA is recognized since early in vitro studies by Nordenson [84] and by Joenje et al. [85]. Subsequently, an extensive body of literature has assessed evidence for the pathogenetic roles of OS in FA gene products [86,87,88,89,90,91,92,93,94], gene transcripts [95], cultured cells [96,97,98,99,100,101,102,103], and in freshly drawn blood cells, plasma and urine from FA patients [104,105,106]. In brief, OS, as a “non-canonical” feature of FA [74], can be reasonably considered as a major pathogenetic feature of this disorder.

3.3. FA: In Vitro and Molecular Evidence for MDF

After the pioneering report by Mukhopadhyay et al. in 2006 [107], it became clear that a FA gene product (FA-G) was associated with MDF, opening the way toward the assumption that FA proteins may be related to the control of mitochondrial activity [108,109,110]. In recent years, a growing body of evidence has been provided assessing that other FA gene products, such as FANCA, FANCC, and FANCD2 are connected to mitochondrial function [111,112,113,114] such as energetic function, altogether providing a link between OS and MDF in FA as well as in a number of cancer-prone genetic diseases, as discussed by Perrone et al. [115].
An involvement of MN in FA was notably suggested by Ponte et al. [102] who reported that L-CARN elicited a significant protective effect against DEB-induced OS, which was potentiated by ALA. Additionally, the same authors reported that ALA, in combination with N-acetyl-cysteine, improved the genetic stability in FA lymphocytes in vitro, significantly decreasing the spontaneous and DEB-induced chromosome instability associated with the cellular FA phenotype [103]. Thus, ALA-related function in MDF prevention may be consistent with a delayed disease progression.
One may conclude that the present state-of-art shows a persuasive link between OS and MDF in FA’s cellular and molecular phenotype. Thus, one may raise an imperative question about the in vivo relevance of these combined deficiencies, so far confined to molecular and in vitro investigations. An in vivo confirmation of the previously reported molecular and cellular MDF in FA is badly needed and very likely. Should a confirmation be provided, this would open gateways to innovative, mitochondrial-based clinical management for FA patients.

4. Safety and Toxicity of Mitochondrial Nutrients

4.1. ALA

α-Lipoic acid (ALA) is synthesized de novo in mitochondria from an 8-carbon fatty (octanoic) acid and functions as a cofactor for several mitochondrial complexes involved in the tricarboxylic acid cycle (TCA) cycle [116]. However, due to sterical differences only R-lipoic acid is endogenously synthesized and covalently bound to the acyl-carrier protein (ACP) in each multienzyme complex [117]. Such a non-protein cofactor known as “prosthetic group” activates the glycine cleavage system and four α-ketoacid dehydrogenase complexes, including: the pyruvate dehydrogenase, the α-ketoglutarate dehydrogenase, the branched-chain α-ketoacid dehydrogenase, and the 2-oxoadipate dehydrogenase complex.
ALA is a physiological compound produced in mammalian cells as part of their basic metabolism that can be safely administered to humans. In some countries ALA is considered a dietary supplement and in others a pharmaceutical drug. ALA is metabolized in different ways when given as a dietary supplement in mammals in various combinations; degradation of ALA is similar in humans and in rats [118]. The safety of ALA has been demonstrated in multiple clinical studies, including SYDNEY, SYDNEY 2, ALADIN I, II, and III, and NATHAN I and II [34,35,36,37,38,39,40,119].
One report of acute ALA-induced toxicity [120] was related to a suicidal attempt following ingestion of 18 g ALA that was, however, reversed after a 3-d supportive treatment. On the other hand, a body of literature has assessed the protective action of ALA against a number of xenobiotics in in vivo [121,122] and in vitro [102,103] investigations.

4.2. CoQ10

Coenzyme Q10 (coQ10) is a natural—and indispensable—compound present in mitochondria.
The synthesis of coQ10 starts with either tyrosine or phenylalanine to synthesize 4-hydroxybenzoate and the benzoquinone followed by the polyisoprenoid side chain synthesis from acetyl-coenzyme A (CoA) via the mevalonate pathway, and by the condensation of these two structures [123]. The benzoquinone group of coQ10 is critical for its function in OXPHOS and ATP production. Accepting electrons from reducing equivalents helps generate fatty acids and glucose metabolism, while transferring protons from the mitochondrial matrix to the intermembrane space creates a proton gradient across the inner mitochondrial membrane [123]. This feature also helps coQ10 to transport protons across lysosomal membranes to maintain the optimal pH [124].
The use of coQ10 as a dietary supplement offers very low toxicity and does not induce serious adverse effects in humans [125]. The acceptable daily intake is 12 mg/kg/day, calculated from the no-observed-adverse-effect level (NOAEL) of 1200 mg/kg/day derived from a 52-week chronic toxicity study in rats, i.e., 720 mg/day for a person weighing 60 kg according to Hidaka et al. [126]. coQ10 was well tolerated at up to 900 mg/day according to Ikematsu et al. [127]. In addition, administration of exogenous coQ10 does not inhibit the physiological production of coQ10 [128,129]. A recent study by Galeshkalami et al. [130] reported on the benefits of ALA and coQ10 combination on experimental diabetic neuropathy by modulating OS and apoptosis.

4.3. CARN

The amino acid derivative carnitine (CARN) is primarily synthesized in the liver in its L-form from lysine and methionine and transported via the bloodstream to cardiac and skeletal muscle [131]. It is required for mitochondrial fatty acid β-oxidation and transport of long-chain fatty acids across the inner membrane of the mitochondria, in the form of acyl-carnitine, where they can be metabolized for energy.
CARN and its active stereoisomer acetyl-L-carnitine (ALC) have been used in a number of human studies alone or as part of a combination therapy since early reports [132]. Administration of CARN in clinical studies including Alzheimer’s disease, depression, aging, diabetes, ischemia, and other neurological diseases did not report major toxic effects (reviewed in [7]). CARN/ALC and different chemical analogs have been used extensively as adjuvant treatment in neurological disorders or to prevent adverse side effects of different chemotherapeutic treatments, such as reducing brain injury after hypoxic-ischemia in newborn rats [133]. Song et al. [134] performed a metanalysis of randomized controlled trials and reported that CARN had good tolerance in patients with chronic heart failure improving clinical symptoms and cardiac functions.
Altogether, we can conclude that separate administration of ALA, or coQ10, or CARN is safe in human and in animal health. Although there are no reports of the combined use of the three MNs in humans, any combined administration should not present potential problems when administered in patients suffering from FA or T2D.

5. Clinical Determination of Mitochondrial Dysfunction

Traditionally any clinical evaluation of mitochondrial function in patients, especially for the diagnosis of neuromuscular diseases, has been performed using skeletal muscle biopsies due to the high number of mitochondria in this tissue and the relatively easy way to collect samples with very few side effects. However, due to the impaired hemostasis in FA patients, the use of clinical muscle biopsies is not advisable. Thus, it is recommended to use only non-invasive methods. A consensus statement of the Mitochondrial Medicine Society on diagnosis of mitochondrial diseases established some diagnostic methods that included the following measurements: (a) lactate and pyruvate levels in plasma; (b) plasma acylcarnitines, (c) urine organic acids, and (d) coQ10 levels in white blood cells (WBC) [135].
It would be advisable to study MDF and OS prevention by MNs in FA patients using biomarkers of mitochondrial function in plasma or blood cells from FA patients. We recommend—as it has been performed in other pathologies—the determination in white blood cells of mitochondrial OS-related enzymes like SOD2, the determination of the mitochondrial membrane potential from isolated white blood cells [136], or 8 hydroxy-deoxyguanosine as a marker of mitochondrial oxidative DNA damage [137]. It would be also advisable to determine well known markers of systemic OS in plasma samples as an indirect marker of mitochondrial impairment, such as the determination of the lipoperoxide biomarker malondialdehyde or calculation of the reduced/oxidized glutathione ratio, the most important antioxidant in mammalian cells (GSH/GSSG) in whole blood of patients [105,111].

6. Conclusions

As predicted by Luft in 1994 [1], “mitochondrial medicine” has met a broad and growing flow of basic and clinical investigations, with extensive implications in a number of disorders related to several medical disciplines [2,7]. This is the case for both T2D and FA, through different pathogenetic mechanisms and background information, and pointing to the prospects of utilizing mitochondrial-targeted adjuvant treatments. Thus, clinical studies are warranted in the improvement of life quality and expectation of patients affected by T2D and FA.
Highlights:
T2D shows excess prevalence in patients with Fanconi anemia;
Both T2D and FA display in vitro and in vivo oxidative stress;
Mitochondrial dysfunction is recognized in T2D patients;
Successful adjuvant treatments in T2D patients used one mitochondrial nutrient;
The use of MN mixtures in treating T2D patients is suggested as a working hypothesis;
MDF in FA was confined to molecular/in vitro studies as in vivo data are unavailable;
Research into in vivo MDF in FA patients is warranted.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Luft, R. The development of mitochondrial medicine. Proc. Natl. Acad. Sci. USA 1994, 91, 8731–8738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pagano, G.; Talamanca, A.A.; Castello, G.; Cordero, M.D.; d’Ischia, M.; Gadaleta, M.N.; Pallardó, F.V.; Petrović, S.; Tiano, L.; Zatterale, A. Oxidative stress and mitochondrial dysfunction across broad-ranging pathologies: Toward a rational design of chemoprevention strategies by means of mitochondrial nutrients. Oxid. Med. Cell. Longev. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
  3. Picard, M.; Wallace, D.C.; Burelle, Y. The rise of mitochondria in medicine. Mitochondrion 2016, 30, 105–116. [Google Scholar] [CrossRef]
  4. Cipak Gasparovic, A.; Zarkovic, N.; Zarkovic, K.; Semen, K.; Kaminskyy, D.; Yelisyeyeva, O.; Bottari, S.P. Biomarkers of oxidative and nitro-oxidative stress: Conventional and novel approaches. Br. J. Pharmacol. 2017, 174, 1771–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Pinti, M.V.; Fink, G.K.; Hathaway, Q.A.; Durr, A.J.; Kunovac, A.; Hollander, J.M. Mitochondrial dysfunction in type 2 diabetes mellitus: An organ-based analysis. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E268–E285. [Google Scholar] [CrossRef] [PubMed]
  6. Wesselink, E.; Koekkoek, W.A.C.; Grefte, S.; Witkamp, R.F.; van Zanten, A.R.H. Feeding mitochondria: Potential role of nutritional components to improve critical illness convalescence. Clin. Nutr. 2019, 38, 982–995. [Google Scholar] [CrossRef] [Green Version]
  7. Pagano, G.; Talamanca Aiello, A.; Castello, G.; Cordero, M.D.; d’Ischia, M.; Gadaleta, M.N.; Pallardó, F.V.; Petrović, S.; Tiano, L.; Zatterale, A. Current experience in testing mitochondrial nutrients in disorders featuring oxidative stress and mitochondrial dysfunction: Rational design of chemoprevention trials. Int. J. Mol. Sci. 2014, 15, 20169–20208. [Google Scholar] [CrossRef]
  8. Tarnopolsky, M.A. The mitochondrial cocktail: Rationale for combined nutraceutical therapy in mitochondrial cytopathies. Adv. Drug Deliv. Rev. 2008, 60, 1561–1567. [Google Scholar] [CrossRef]
  9. Palade, G.E. The organization of living matter. Proc. Natl. Acad. Sci. USA 1964, 52, 613–634. [Google Scholar] [CrossRef] [Green Version]
  10. Haghighatdoost, F.; Hariri, M. The effect of alpha-lipoic acid on inflammatory mediators: A systematic review and meta-analysis on randomized clinical trials. Eur. J. Pharmacol. 2019, 849, 115–123. [Google Scholar] [CrossRef]
  11. Mantle, D.; Hargreaves, I. Coenzyme Q10 and degenerative disorders affecting longevity: An overview. Antioxidants 2019, 8, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sizova, Z.M.; Shikh, E.V.; Makhova, A.A. Significance of L-carnitine in internal medicine. Ter. Arkhiv 2019, 91, 114–128. [Google Scholar] [CrossRef] [PubMed]
  13. Baynes, J.W. Role of oxidative stress in development of complications in diabetes. Diabetes 1991, 40, 405–412. [Google Scholar] [CrossRef] [PubMed]
  14. Wolff, S.P.; Jiang, Z.Y.; Hunt, J.V. Protein glycation and oxidative stress in diabetes mellitus and ageing. Free Radic. Biol. Med. 1991, 10, 339–352. [Google Scholar] [CrossRef]
  15. Bandeira Sde, M.; Guedes Gda, S.; da Fonseca, L.J.; Pires, A.S.; Gelain, D.P.; Moreira, J.C.; Rabelo, L.A.; Vasconcelos, S.L.; Goulart, M.O.F. Characterization of blood oxidative stress in type 2 diabetes mellitus patients: Increase in lipid peroxidation and SOD activity. Oxid. Med. Cell. Longev. 2012, 2012, 819310. [Google Scholar] [CrossRef]
  16. Yaribeygi, H.; Atkin, S.L.; Sahebkar, A. Mitochondrial dysfunction in diabetes and the regulatory roles of antidiabetic agents on the mitochondrial function. J. Cell. Physiol. 2019, 234, 8402–8410. [Google Scholar] [CrossRef]
  17. Darroudi, S.; Fereydouni, N.; Tayefi, M.; Ahmadnezhad, M.; Zamani, P.; Tayefi, B.; Kharazmi, J.; Tavalaie, S.; Heidari-Bakavoli, A.; Azarpajouh, M.R.; et al. Oxidative stress and inflammation, two features associated with a high percentage body fat, and that may lead to diabetes mellitus and metabolic syndrome. Biofactors 2018, 45, 35–42. [Google Scholar] [CrossRef]
  18. Robertson, R.P.; Harmon, J.; Tran, P.O.; Poitout, V. Beta-cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes 2004, 53, S119–S124. [Google Scholar] [CrossRef] [Green Version]
  19. Borghetti, G.; von Lewinski, D.; Eaton, D.M.; Sourij, H.; Houser, S.R.; Wallner, M. Diabetic cardiomyopathy: Current and future therapies. beyond glycemic control. Front. Physiol. 2018, 9, 1514. [Google Scholar] [CrossRef]
  20. Wu, M.Y.; Yiang, G.T.; Lai, T.T.; Li, C.J. The oxidative stress and mitochondrial dysfunction during the pathogenesis of diabetic retinopathy. Oxid. Med. Cell. Longev. 2018, 2018, 3420187. [Google Scholar] [CrossRef]
  21. Santiago, A.R.; Boia, R.; Aires, I.D.; Ambrósio, A.F.; Fernandes, R. Sweet stress: Coping with vascular dysfunction in diabetic retinopathy. Front. Physiol. 2018, 9, 820. [Google Scholar] [CrossRef] [PubMed]
  22. Björntorp, P.; Scherstén, T.; Fagerberg, S.E. Respiration and phosphorylation of mitochondria isolated from the skeletal muscle of diabetic and normal subjects. Diabetologia 1967, 3, 346–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Choi, S.C.; Hall, J.C. A study of respiration and oxidative phosphorylation of mitochondria from regenerating liver of normal and diabetic rats. Cancer Res. 1974, 34, 2351–2357. [Google Scholar] [PubMed]
  24. Wang, C.H.; Wei, Y.H. Role of mitochondrial dysfunction and dysregulation of Ca2+ homeostasis in the pathophysiology of insulin resistance and type 2 diabetes. J. Biomed. Sci. 2017, 24, 70. [Google Scholar] [CrossRef]
  25. Kähler, W.; Kuklinski, B.; Rühlmann, C.; Plötz, C. Diabetes mellitus—A free radical-associated disease. Results of adjuvant antioxidant supplementation. Z. Gesamte Inn. Med. 1993, 48, 223–232. [Google Scholar]
  26. Pieralice, S.; Vari, R.; Minutolo, A.; Maurizi, A.R.; Fioriti, E.; Napoli, N.; Pozzilli, P.; Manfrini, S.; Maddaloni, E. Biomarkers of response to alpha-lipoic acid ± palmitoiletanolamide treatment in patients with diabetes and symptoms of peripheral neuropathy. Endocrine 2019, 66, 178–184. [Google Scholar] [CrossRef]
  27. Meng, B.; Li, J.; Cao, H. Antioxidant and antiinflammatory activities of curcumin on diabetes mellitus and its complications. Curr. Pharm. Des. 2013, 19, 2101–2113. [Google Scholar]
  28. Rochette, L.; Zeller, M.; Cottin, Y.; Vergely, C. Diabetes, oxidative stress and therapeutic strategies. Biochim. Biophys. Acta 2014, 1840, 2709–2729. [Google Scholar] [CrossRef]
  29. Montero, D.; Walther, G.; Stehouwer, C.D.; Houben, A.J.; Beckman, J.A.; Vinet, A. Effect of antioxidant vitamin supplementation on endothelial function in type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Obes. Rev. 2014, 15, 107–116. [Google Scholar] [CrossRef]
  30. Panahi, Y.; Khalili, N.; Sahebi, E.; Namazi, S.; Karimian, M.S.; Majeed, M.; Sahebkar, A. Antioxidant effects of curcuminoids in patients with type 2 diabetes mellitus: A randomized controlled trial. Inflammopharmacology 2017, 25, 25–31. [Google Scholar] [CrossRef]
  31. Bi, X.; Lim, J.; Henry, C.J. Spices in the management of diabetes mellitus. Food Chem. 2017, 281–293. [Google Scholar] [CrossRef] [PubMed]
  32. Park, J.K.; Shim, J.Y.; Cho, A.R.; Cho, M.R.; Lee, Y.J. Korean red ginseng protects against mitochondrial damage and intracellular inflammation in an animal model of type 2 diabetes mellitus. J. Med. Food 2018, 21, 544–550. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, J.; You, Y.J. Regulation of organelle function by metformin. IUBMB Life 2017, 69, 459–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ziegler, D.; Hanefeld, M.; Ruhnau, K.J.; Meissner, H.P.; Lobisch, M.; Schütte, K.; Gries, F.A.; The ALADIN Study Group. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia 1995, 38, 1425–1433. [Google Scholar] [CrossRef]
  35. Ziegler, D.; Hanefeld, M.; Ruhnau, K.J.; Hasche, H.; Lobisch, M.; Schütte, K.; Kerum, G.; Malessa, R. Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: A 7-month multicenter randomized controlled trial (ALADIN III Study). ALADIN III Study Group. Alpha-lipoic acid in diabetic neuropathy. Diabetes Care 1999, 22, 1296–1301. [Google Scholar] [CrossRef]
  36. Jacob, S.; Ruus, P.R.; Hermann, R.; Tritschler, H.J.; Maerker, E.; Renn, W.; Augustin, H.J.; Dietze, G.J.; Rett, K. Oral administration of RAC-alpha-lipoic acid modulates insulin sensitivity in patients with type-2 diabetes mellitus: A placebo-controlled pilot trial. Free Radic. Biol. Med. 1999, 27, 309–314. [Google Scholar] [CrossRef]
  37. Androne, L.; Gavan, N.A.; Veresiu, I.A.; Orasan, R. In Vivo effect of lipoic acid on lipid peroxidation in patients with diabetic neuropathy. In Vivo 2000, 14, 327–330. [Google Scholar]
  38. Liu, F.; Zhang, Y.; Yang, M.; Liu, B.; Shen, Y.D.; Jia, W.P.; Xiang, K.S. Curative effect of alpha-lipoic acid on peripheral neuropathy in type 2 diabetes: A clinical study. Zhonghua Yi Xue Za Zhi 2007, 87, 2706–2709. [Google Scholar] [PubMed]
  39. Xiang, G.D.; Sun, H.L.; Zhao, L.S.; Hou, J.; Yue, L.; Xu, L. The antioxidant alpha-lipoic acid improves endothelial dysfunction induced by acute hyperglycaemia during OGTT in impaired glucose tolerance. Clin. Endocrinol. (Oxf.) 2008, 68, 716–723. [Google Scholar] [CrossRef] [PubMed]
  40. Heinisch, B.B.; Francesconi, M.; Mittermayer, F.; Schaller, G.; Gouya, G.; Wolzt, M.; Pleiner, J. Alpha-lipoic acid improves vascular endothelial function in patients with type 2 diabetes: A placebo-controlled randomized trial. Eur. J. Clin. Investig. 2010, 40, 148–154. [Google Scholar] [CrossRef]
  41. Gu, X.M.; Zhang, S.S.; Wu, J.C.; Tang, Z.Y.; Lu, Z.Q.; Li, H.; Liu, C.; Chen, L.; Ning, G. Efficacy and safety of high-dose α-lipoic acid in the treatment of diabetic polyneuropathy. Zhonghua Yi Xue Za Zhi 2010, 90, 2473–2476. [Google Scholar] [PubMed]
  42. Xiang, G.; Pu, J.; Yue, L.; Hou, J.; Sun, H. α-lipoic acid can improve endothelial dysfunction in subjects with impaired fasting glucose. Metabolism 2011, 60, 480–485. [Google Scholar] [CrossRef] [PubMed]
  43. de Oliveira, A.M.; Rondó, P.H.; Luzia, L.A.; D’Abronzo, F.H.; Illison, V.K. The effects of lipoic acid and α-tocopherol supplementation on the lipid profile and insulin sensitivity of patients with type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled trial. Diabetes Res. Clin. Pract. 2011, 92, 253–260. [Google Scholar] [CrossRef] [PubMed]
  44. Rieusset, J. Role of endoplasmic reticulum-mitochondria communication in type 2 diabetes. Adv. Exp. Med. Biol. 2017, 997, 171–186. [Google Scholar] [PubMed]
  45. Thivolet, C.; Vial, G.; Cassel, R.; Rieusset, J.; Madec, A.M. Reduction of endoplasmic reticulum-mitochondria interactions in beta cells from patients with type 2 diabetes. PLoS ONE 2017, 12, e0182027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Gonzalez-Franquesa, A.; Patti, M.E. Insulin resistance and mitochondrial dysfunction. Adv. Exp. Med. Biol. 2017, 982, 465–520. [Google Scholar]
  47. Amara, F.; Hafez, S.; Orabi, A.; El Etriby, A.; Abdel Rahim, A.A.; Zakaria, E.; Koura, F.; Talaat, F.M.; Gawish, H.; Attia, I.; et al. Review of diabetic polyneuropathy: Pathogenesis, diagnosis and management according to the consensus of Egyptian experts. Curr. Diabetes Rev. 2019, 15, 340–345. [Google Scholar] [CrossRef]
  48. Nguyen, N.; Takemoto, J.K. A case for alpha-lipoic acid as an alternative treatment for diabetic polyneuropathy. J. Pharm. Pharm. Sci. 2018, 21, 177s–191s. [Google Scholar] [CrossRef]
  49. German Drug Index. 1994. Available online: http://www.gbe-bund.de (accessed on 17 December 2019).
  50. Watts, G.F.; Playford, D.A.; Croft, K.D.; Ward, N.C.; Mori, T.A.; Burke, V. Coenzyme Q(10) improves endothelial dysfunction of the brachial artery in Type II diabetes mellitus. Diabetologia 2002, 45, 420–426. [Google Scholar] [CrossRef] [Green Version]
  51. Hodgson, J.M.; Watts, G.F.; Playford, D.A.; Burke, V.; Croft, K.D. Coenzyme Q10 improves blood pressure and glycaemic control: A controlled trial in subjects with type 2 diabetes. Eur. J. Clin. Nutr. 2002, 56, 1137–1142. [Google Scholar] [CrossRef] [Green Version]
  52. Chew, G.T.; Watts, G.F.; Davis, T.M.; Stuckey, B.G.; Beilin, L.J. Hemodynamic effects of fenofibrate and coenzyme Q10 in type 2 diabetic subjects with left ventricular diastolic dysfunction. Diabetes Care 2008, 31, 1502–1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Hamilton, S.J.; Chew, G.T.; Watts, G.F. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care 2009, 32, 810–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kolahdouz Mohammadi, R.; Hosseinzadeh-Attar, M.J.; Eshraghian, M.R.; Nakhjavani, M.; Khorami, E.; Esteghamati, A. The effect of coenzyme Q10 supplementation on metabolic status of type 2 diabetic patients. Minerva Gastroenterol. Dietol. 2013, 59, 231–236. [Google Scholar] [PubMed]
  55. Di Meo, S.; Iossa, S.; Venditti, P. Skeletal muscle insulin resistance: Role of mitochondria and other ROS sources. J. Endocrinol. 2017, 233, R15–R42. [Google Scholar] [CrossRef] [Green Version]
  56. Mingrone, G.; Greco, A.V.; Capristo, E.; Benedetti, G.; Giancaterini, A.; De Gaetano, A.; Gasbarrini, G. L-carnitine improves glucose disposal in type 2 diabetic patients. J. Am. Coll. Nutr. 1999, 18, 77–82. [Google Scholar] [CrossRef]
  57. De Grandis, D.; Minardi, C. Acetyl-L-carnitine (levacecarnine) in the treatment of diabetic neuropathy. A long-term, randomised, double-blind, placebo-controlled study. Drugs R D 2002, 3, 223–231. [Google Scholar] [CrossRef]
  58. Derosa, G.; Cicero, A.F.; Gaddi, A.; Mugellini, A.; Ciccarelli, L.; Fogari, R. The effect of L-carnitine on plasma lipoprotein(a) levels in hypercholesterolemic patients with type 2 diabetes mellitus. Clin. Ther. 2003, 25, 1429–1439. [Google Scholar] [CrossRef]
  59. Ragozzino, G.; Mattera, E.; Madrid, E.; Salomone, P.; Fasano, C.; Gioia, F.; Acerra, G.; del Guercio, R.; Federico, P. Effects of propionyl-carnitine in patients with type 2 diabetes and peripheral vascular disease: Results of a pilot trial. Drugs R D 2004, 5, 185–190. [Google Scholar] [CrossRef]
  60. Sima, A.A.; Calvani, M.; Mehra, M.; Amato, A. Acetyl-L-Carnitine Study Group. Acetyl-L-carnitine improves pain, nerve regeneration, and vibratory perception in patients with chronic diabetic neuropathy: An analysis of two randomized placebo-controlled trials. Diabetes Care 2005, 28, 89–94. [Google Scholar] [CrossRef] [Green Version]
  61. Rahbar, A.R.; Shakerhosseini, R.; Saadat, N.; Taleban, F.; Pordal, A.; Gollestan, B. Effect of L-carnitine on plasma glycemic and lipidemic profile in patients with type II diabetes mellitus. Eur. J. Clin. Nutr. 2005, 59, 592–596. [Google Scholar] [CrossRef] [Green Version]
  62. Solfrizzi, V.; Capurso, C.; Colacicco, A.M.; D’Introno, A.; Fontana, C.; Capurso, S.A.; Torres, F.; Gadaleta, A.M.; Koverech, A.; Capurso, A.; et al. Efficacy and tolerability of combined treatment with L-carnitine and simvastatin in lowering lipoprotein (a) serum levels in patients with type 2 diabetes mellitus. Atherosclerosis 2006, 188, 455–461. [Google Scholar] [CrossRef] [PubMed]
  63. Malaguarnera, M.; Vacante, M.; Avitabile, T.; Malaguarnera, M.; Cammalleri, L.; Motta, M. L-Carnitine supplementation reduces oxidized LDL cholesterol in patients with diabetes. Am. J. Clin. Nutr. 2009, 89, 71–76. [Google Scholar] [CrossRef] [PubMed]
  64. Ruggenenti, P.; Cattaneo, D.; Loriga, G.; Ledda, F.; Motterlini, N.; Gherardi, G.; Orisio, S.; Remuzzi, G. Ameliorating hypertension and insulin resistance in subjects at increased cardiovascular risk: Effects of acetyl-L-carnitine therapy. Hypertension 2009, 54, 567–574. [Google Scholar] [CrossRef] [PubMed]
  65. Molfino, A.; Cascino, A.; Conte, C.; Ramaccini, C.; Rossi Fanelli, F.; Laviano, A. Caloric restriction and L-carnitine administration improves insulin sensitivity in patients with impaired glucose metabolism. J. Parenter. Enter. Nutr. 2010, 34, 295–299. [Google Scholar] [CrossRef]
  66. Palacka, P.; Kucharska, J.; Murin, J.; Dostalova, K.; Okkelova, A.; Cizova, M.; Waczulikova, I.; Moricova, S.; Gvozdjakova, A. Complementary therapy in diabetic patients with chronic complications: A pilot study. Bratisl. Lek. Listy 2010, 111, 205–211. [Google Scholar]
  67. Liu, J.; Ames, B.N. Reducing mitochondrial decay with mitochondrial nutrients to delay and treat cognitive dysfunction, Alzheimer’s disease, and Parkinson’s disease. Nutr. Neurosci. 2005, 8, 67–89. [Google Scholar] [CrossRef]
  68. Ames, B.N. Optimal micronutrients delay mitochondrial decay and age-associated diseases. Mech. Ageing Dev. 2010, 131, 473–479. [Google Scholar] [CrossRef]
  69. Shen, W.; Liu, K.; Tian, C.; Yang, L.; Li, X.; Ren, J.; Packer, L.; Head, E.; Sharman, E.; Liu, J. Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J. Cell. Biochem. 2008, 104, 1232–1243. [Google Scholar] [CrossRef]
  70. Shen, W.; Liu, K.; Tian, C.; Yang, L.; Li, X.; Ren, J.; Packer, L.; Cotman, C.W.; Liu, J. R-alpha-lipoic acid and acetyl-L-carnitine complementarily promote mitochondrial biogenesis in murine 3T3-L1 adipocytes. Diabetologia 2008, 51, 165–174. [Google Scholar] [CrossRef]
  71. Liu, J.; Shen, W.; Zhao, B.; Wang, Y.; Wertz, K.; Weber, P.; Zhang, P. Targeting mitochondrial biogenesis for preventing and treating insulin resistance in diabetes and obesity: Hope from natural mitochondrial nutrients. Adv. Drug Deliv. Rev. 2009, 61, 1343–1352. [Google Scholar] [CrossRef]
  72. Hao, J.; Shen, W.; Tian, C.; Liu, Z.; Ren, J.; Luo, C.; Long, J.; Sharman, E.; Liu, J. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J. Cell. Mol. Med. 2009, 13, 701–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hao, J.; Shen, W.; Sun, L.; Long, J.; Sharman, E.; Shi, X.; Liu, J. Mitochondrial dysfunction in the liver of type 2 diabetic Goto-Kakizaki rats: Improvement by a combination of nutrients. Br. J. Nutr. 2011, 106, 648–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bagby, G. Recent advances in understanding hematopoiesis in Fanconi Anemia. F1000Res. 2018, 7, 105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wajnrajch, M.P.; Gertner, J.M.; Huma, Z.; Popovic, J.; Lin, K.; Verlander, P.C.; Batish, S.D.; Giampietro, P.F.; Davis, J.G.; New, M.I. Evaluation of growth and hormonal status in patients referred to the International Fanconi Anemia Registry. Pediatrics 2001, 107, 744–754. [Google Scholar] [CrossRef]
  76. Elder, D.A.; D’Alessio, D.A.; Eyal, O.; Mueller, R.; Smith, F.O.; Kansra, A.R.; Rose, S.R. Abnormalities in glucose tolerance are common in children with Fanconi anemia and associated with impaired insulin secretion. Pediatric Blood Cancer 2008, 51, 256–260. [Google Scholar] [CrossRef]
  77. Petryk, A.; Kanakatti Shankar, R.; Giri, N.; Hollenberg, A.N.; Rutter, M.M.; Nathan, B.; Lodish, M.; Alter, B.P.; Stratakis, C.A.; Rose, S.R. Endocrine disorders in Fanconi anemia: Recommendations for screening and treatment. J. Clin. Endocrinol. Metab. 2015, 100, 803–811. [Google Scholar] [CrossRef] [Green Version]
  78. Li, J.; Sipple, J.; Maynard, S.; Mehta, P.A.; Rose, S.R.; Davies, S.M.; Pang, Q. Fanconi anemia links reactive oxygen species to insulin resistance and obesity. Antioxid. Redox Signal. 2012, 17, 1083–1098. [Google Scholar] [CrossRef]
  79. Izzo, A.; Mollo, N.; Nitti, M.; Paladino, S.; Calì, G.; Genesio, R.; Bonfiglio, F.; Cicatiello, R.; Barbato, M.; Sarnataro, V.; et al. Mitochondrial dysfunction in Down syndrome: Molecular mechanisms and therapeutic targets. Mol. Med. 2018, 24, 2. [Google Scholar] [CrossRef] [Green Version]
  80. Nishioka, M.; Kamei, S.; Kinoshita, T.; Sanada, J.; Fushimi, Y.; Irie, S.; Hirata, Y.; Tanabe, A.; Hirukawa, H.; Kimura, T.; et al. Werner syndrome and diabetes mellitus accompanied by adrenal cortex cancer. Intern. Med. 2017, 56, 1987–1992. [Google Scholar] [CrossRef] [Green Version]
  81. Tsikouras, P.; Spyros, L.; Manav, B.; Zervoudis, S.; Poiana, C.; Nikolaos, T.; Petros, P.; Dimitraki, M.; Koukouli, C.; Galazios, G.; et al. Features of polycystic ovary syndrome in adolescence. J. Med. Life 2015, 8, 291–296. [Google Scholar]
  82. Jia, G.; Aroor, A.R.; Sowers, J.R. Estrogen and mitochondria function in cardiorenal metabolic syndrome. Prog. Mol. Biol. Transl. Sci. 2014, 127, 229–249. [Google Scholar] [PubMed] [Green Version]
  83. Espach, Y.; Lochner, A.; Strijdom, H.; Huisamen, B. ATM protein kinase signaling, type 2 diabetes and cardiovascular disease. Cardiovasc. Drugs Ther. 2015, 29, 51–58. [Google Scholar] [CrossRef] [PubMed]
  84. Nordenson, I. Effect of superoxide dismutase and catalase on spontaneously occuring chromosome breaks in patients with Fanconi’s anemia. Hereditas 1977, 86, 147–150. [Google Scholar] [CrossRef] [PubMed]
  85. Joenje, H.; Arwert, F.; Eriksson, A.W.; de Koning, H.; Oostra, A.B. Oxygen-dependence of chromosomal aberrations in Fanconi’s anaemia. Nature 1981, 290, 142–143. [Google Scholar] [CrossRef]
  86. Kruyt, F.A.; Hoshino, T.; Liu, J.M.; Joseph, P.; Jaiswal, A.K.; Youssoufian, H. Abnormal microsomal detoxification implicated in Fanconi anemia group C by interaction of the FAC protein with NADPH cytochrome P450 reductase. Blood 1998, 92, 3050–3056. [Google Scholar] [CrossRef]
  87. Le Page, F.; Randrianarison, V.; Marot, D.; Cabannes, J.; Perricaudet, M.; Feunteun, J.; Sarasin, A. BRCA1 and BRCA2 are necessary for the transcription-coupled repair of the oxidative 8-oxoguanine lesion in human cells. Cancer Res. 2000, 60, 5548–5552. [Google Scholar]
  88. Cumming, R.C.; Lightfoot, J.; Beard, K.; Youssoufian, H.; O’Brien, P.J.; Buchwald, M. Fanconi anemia group C protein prevents apoptosis in hematopoietic cells through redox regulation of GSTP1. Nat. Med. 2001, 7, 814–820. [Google Scholar] [CrossRef]
  89. Futaki, M.; Igarashi, T.; Watanabe, S.; Kajigaya, S.; Tatsuguchi, A.; Wang, J.; Liu, J.M. The FANCG Fanconi anemia protein interacts with CYP2E1: Possible role in protection against oxidative DNA damage. Carcinogenesis 2002, 23, 67–72. [Google Scholar] [CrossRef] [Green Version]
  90. Otsuki, T.; Nagashima, T.; Komatsu, N.; Kirito, K.; Furukawa, Y.; Kobayashi, S.; Liu, J.M.; Ozawa, K. Phosphorylation of Fanconi anemia protein, FANCA, is regulated by Akt kinase. Biochem. Biophys. Res. Commun. 2002, 291, 628–634. [Google Scholar] [CrossRef]
  91. Pang, Q.; Christianson, T.A.; Keeble, W.; Koretsky, T.; Bagby, G.C. The anti-apoptotic function of Hsp70 in the interferon-inducible double-stranded RNA-dependent protein kinase-mediated death signaling pathway requires the Fanconi anemia protein, FANCC. J. Biol. Chem. 2002, 277, 49638–49643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zanier, R.; Briot, D.; Dugas du Villard, J.A.; Sarasin, A.; Rosselli, F. Fanconi anemia C gene product regulates expression of genes involved in differentiation and inflammation. Oncogene 2004, 23, 5004–5013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Park, S.J.; Ciccone, S.L.M.; Beck, B.D.; Hwang, B.; Freie, B.; Clapp, D.W.; Lee, S.H. Oxidative stress/damage induces multimerization and interaction of Fanconi anemia proteins. J. Biol. Chem. 2004, 279, 30053–30059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ho, G.P.; Margossian, S.; Taniguchi, T.; D’Andrea, A.D. Phosphorylation of FANCD2 on two novel sites is required for mitomycin C resistance. Mol. Cell. Biol. 2006, 26, 7005–7015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Pagano, G.; Talamanca Aiello, A.; Castello, G.; d’Ischia, M.; Pallardó, F.V.; Petrović, S.; Porto, B.; Tiano, L.; Zatterale, A. Bone marrow cell transcripts from Fanconi anaemia patients reveal In Vivo alterations in mitochondrial, redox and DNA repair pathways. Eur. J. Haematol. 2013, 91, 141–151. [Google Scholar] [CrossRef] [PubMed]
  96. Schultz, J.C.; Shahidi, N.T. Tumor necrosis factor-α overproduction in Fanconi’s anemia. Am. J. Hematol. 1993, 42, 196–201. [Google Scholar] [CrossRef] [PubMed]
  97. Takeuchi, T.; Morimoto, K. Increased formation of 8-hydroxydeoxy-guanosine, an oxidative DNA damage, in lymphoblasts from Fanconi’s anemia patients due to possible catalase deficiency. Carcinogenesis 1993, 14, 1115–1120. [Google Scholar] [CrossRef]
  98. Clarke, A.A.; Philpott, N.J.; Gordon-Smith, E.C.; Rutherford, T.R. The sensitivity of Fanconi anaemia group C cells to apoptosis induced by mitomycin C is due to oxygen radical generation, not DNA crosslinking. Br. J. Haematol. 1997, 96, 240–247. [Google Scholar] [CrossRef]
  99. Pagano, G.; Korkina, L.G.; Degan, P.; Del Principe, D.; Lindau-Shepard, B.; Zatterale, A.; Franceschi, C. In Vitro hypersensitivity to oxygen of Fanconi anemia (FA) cells is linked to ex vivo evidence for oxidative stress in FA homozygotes and heterozygotes. Blood 1997, 89, 1111–1112. [Google Scholar] [CrossRef]
  100. Raj, A.S.; Heddle, J.A. The effect of superoxide dismutase, catalase and L-cysteine on spontaneous and mitomycin C induced chromosomal breakage in Fanconi’s anemia and normal fibroblasts as measured by the micronucleus method. Mutat. Res. 1980, 78, 59–66. [Google Scholar]
  101. Ruppitsch, W.; Meisslitzer, C.; Hirsch-Kauffmann, M.; Schweiger, M. Overexpression of thioredoxin in Fanconi anemia fibroblasts prevents the cytotoxic and DNA damaging effect of mitomycin C and diepoxybutane. FEBS Lett. 1998, 422, 99–102. [Google Scholar] [CrossRef] [Green Version]
  102. Ponte, F.; Carvalho, F.; Porto, B. Protective effect of acetyl-l-carnitine and α-lipoic acid against the acute toxicity of diepoxybutane to human lymphocytes. Toxicology 2011, 289, 52–58. [Google Scholar] [CrossRef]
  103. Ponte, F.; Sousa, R.; Fernandes, A.P.; Gonçalves, C.; Barbot, J.; Carvalho, F.; Porto, B. Improvement of genetic stability in lymphocytes from Fanconi anemia patients through the combined effect of α-lipoic acid and N-acetylcysteine. Orphanet J. Rare Dis. 2012, 7, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Degan, P.; Bonassi, S.; De Caterina, M.; Korkina, L.G.; Pinto, L.; Scopacasa, F.; Zatterale, A.; Calzone, R.; Pagano, G. In Vivo accumulation of 8-hydroxy-2′-deoxyguanosine in DNA correlates with release of reactive oxygen species in Fanconi’s anaemia families. Carcinogenesis 1995, 16, 735–742. [Google Scholar] [CrossRef] [PubMed]
  105. Pagano, G.; Degan, P.; d’Ischia, M.; Kelly, F.J.; Pallardó, F.V.; Zatterale, A.; Anak, S.S.; Akişık, E.E.; Beneduce, G.; Calzone, R.; et al. Gender- and age-related distinctions for the In Vivo prooxidant state in Fanconi anaemia patients. Carcinogenesis 2004, 25, 1899–1909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lloret, A.; Calzone, R.; Dunster, C.; Manini, P.; d’Ischia, M.; Degan, P.; Kelly, F.J.; Pallardó, F.V.; Zatterale, A.; Pagano, G. Different patterns of In Vivo prooxidant states in a set of cancer- or ageing-related genetic diseases. Free Radic. Biol. Med. 2008, 44, 495–503. [Google Scholar] [CrossRef] [PubMed]
  107. Mukhopadhyay, S.S.; Leung, K.S.; Hicks, M.J.; Hastings, P.J.; Youssoufian, H.; Plon, S.E. Defective mitochondrial peroxiredoxin-3 results in sensitivity to oxidative stress in Fanconi anemia. J. Cell Biol. 2006, 175, 225–235. [Google Scholar] [CrossRef] [Green Version]
  108. Lyakhovich, A. Damaged mitochondria and overproduction of ROS in Fanconi anemia cells. Rare Dis. 2013, 1, e24048. [Google Scholar] [CrossRef] [Green Version]
  109. Pagano, G.; Shyamsunder, P.; Verma, R.S.; Lyakhovich, A. Damaged mitochondria in Fanconi anemia—An isolated event or a general phenomenon? Oncoscience 2014, 1, 287–295. [Google Scholar] [CrossRef] [Green Version]
  110. Shyamsunder, P.; Esner, M.; Barvalia, M.; Wu, Y.J.; Loja, T.; Boon, H.B.; Lleonart, M.E.; Verma, R.S.; Krejci, L.; Lyakhovich, A. Impaired mitophagy in Fanconi anemia is dependent on mitochondrial fission. Oncotarget 2016, 7, 58065–58074. [Google Scholar] [CrossRef]
  111. Zhang, T.; Du, W.; Wilson, A.F.; Namekawa, S.H.; Andreassen, P.R.; Meetei, A.R.; Pang, Q. Fancd2 In Vivo interaction network reveals a non-canonical role in mitochondrial function. Sci. Rep. 2017, 7, 45626. [Google Scholar] [CrossRef] [Green Version]
  112. Ravera, S.; Dufour, C.; Degan, P.; Cappelli, E. Fanconi anemia: From DNA repair to metabolism. Eur. J. Hum. Genet. 2018, 26, 475–476. [Google Scholar] [CrossRef] [PubMed]
  113. Bottega, R.; Nicchia, E.; Cappelli, E.; Ravera, S.; De Rocco, D.; Faleschini, M.; Corsolini, F.; Pierri, F.; Calvillo, M.; Russo, G.; et al. Hypomorphic FANCA mutations correlate with mild mitochondrial and clinical phenotype in Fanconi anemia. Haematologica 2018, 103, 417–426. [Google Scholar] [CrossRef] [Green Version]
  114. Jayabal, P.; Ma, C.; Nepal, M.; Shen, Y.; Che, R.; Turkson, J.; Fei, P. Involvement of FANCD2 in energy metabolism via ATP5α. Sci. Rep. 2017, 7, 4921. [Google Scholar] [CrossRef] [PubMed]
  115. Perrone, S.; Lotti, F.; Geronzi, U.; Guidoni, E.; Longini, M.; Buonocore, G. Oxidative stress in cancer-prone genetic diseases in pediatric age: The role of mitochondrial dysfunction. Oxid. Med. Cell. Longev. 2016, 2016, 4782426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Jones, W.; Li, X.; Qu, Z.C.; Perriott, L.; Whitesell, R.R.; May, J.M. Uptake, recycling, and antioxidant actions of α-lipoic acid in endothelial cells. Free Radic. Biol. Med. 2002, 33, 83–93. [Google Scholar] [CrossRef]
  117. Mayr, J.A.; Feichtinger, R.G.; Tort, F.; Ribes, A.; Sperl, W. Lipoic acid biosynthesis defects. J. Inherit. Metab. Dis. 2014, 37, 553–563. [Google Scholar] [CrossRef]
  118. Schupke, H.; Hempel, R.; Peter, G.; Hermann, R.; Wessel, K.; Engel, J.; Kronbach, T. New metabolic pathways of alpha-lipoic acid. Drug Metab. Dispos. 2001, 29, 855–862. [Google Scholar]
  119. Teichert, J.; Tuemmers, T.; Achenbach, H.; Preiss, C.; Hermann, R.; Ruus, P.; Preiss, R. Pharmacokinetics of alpha-lipoic acid in subjects with severe kidney damage and end-stage renal disease. J. Clin. Pharmacol. 2005, 45, 313–328. [Google Scholar] [CrossRef]
  120. Emir, D.F.; Ozturan, I.U.; Yilmaz, S. Alpha lipoic acid intoxicatıon: An adult. Am. J. Emerg. Med. 2018, 36, 1125.e3. [Google Scholar] [CrossRef]
  121. Cremer, D.R.; Rabeler, R.; Roberts, A.; Lynch, B. Safety evaluation of alpha-lipoic acid ALA. Regul. Toxicol. Pharmacol. 2006, 46, 29–41. [Google Scholar] [CrossRef]
  122. Suh, S.H.; Lee, K.E.; Kim, I.J.; Kim, O.; Kim, C.S.; Choi, J.S.; Choi, H.-I.; Bae, E.H.; Ma, S.K.; Lee, J.U.; et al. Alpha-lipoic acid attenuates lipopolysaccharide-induced kidney injury. Clin. Exp. Nephrol. 2015, 19, 82–91. [Google Scholar] [CrossRef] [PubMed]
  123. Acosta, M.J.; Vazquez Fonseca, L.; Desbats, M.A.; Cerqua, C.; Zordan, R.; Trevisson, E.; Salviati, L. Coenzyme Q biosynthesis in health and disease. Biochim. Biophys. Acta Bioenerg. 2016, 1857, 1079–1085. [Google Scholar] [CrossRef]
  124. Navas, P.; Manuel Villalba, J.; de Caboc, R. The importance of plasma membrane coenzyme Q in aging and stress responses. Mitochondrion 2007, 7, S34–S40. [Google Scholar] [CrossRef] [PubMed]
  125. Hidaka, T.; Fujii, K.; Funahashi, I.; Fukutomi, N.; Hosoe, K. Safety assessment of coenzyme Q10 (CoQ10). Biofactors 2008, 32, 199–208. [Google Scholar] [CrossRef] [PubMed]
  126. Ikematsu, H.; Nakamura, K.; Harashima, S.; Fujii, K.; Fukutomi, N. Safety assessment of coenzyme Q10 Kaneka Q10 in healthy subjects: A double-blind, randomized, placebo-controlled trial. Regul. Toxicol. Pharmacol. 2006, 44, 212–218. [Google Scholar] [CrossRef] [PubMed]
  127. Alehagen, U.; Aaseth, J.; Johansson, P. Reduced cardiovascular mortality 10 years after supplementation with selenium and coenzyme Q10 for four years: Follow-up results of a prospective randomized double-blind placebo-controlled trial in elderly citizens. PLoS ONE 2015, 10, e0141641. [Google Scholar] [CrossRef] [PubMed]
  128. Alehagen, U.; Aaseth, J.; Alexander, J.; Johansson, P. Still reduced cardiovascular mortality 12 years after supplementation with selenium and coenzyme Q10 for four years: A validation of previous 10-year follow-up results of a prospective randomized double-blind placebo-controlled trial in elderly. PLoS ONE 2018, 13, e0193120. [Google Scholar] [CrossRef]
  129. Suksomboon, N.; Poolsup, N.; Juanak, N. Effects of coenzyme Q10 supplementation on metabolic profile in diabetes: A systematic review and meta-analysis. J. Clin. Pharm. Ther. 2015, 40, 413–418. [Google Scholar] [CrossRef]
  130. Sadeghiyan Galeshkalami, N.; Abdollahi, M.; Najafi, R.; Baeeri, M.; Jamshidzade, A.; Falak, R.; Gholami, M.D.; Hassanzadeh, G.; Mokhtari, T.; Hassani, S.; et al. Alpha-lipoic acid and coenzyme Q10 combination ameliorates experimental diabetic neuropathy by modulating oxidative stress and apoptosis. Life Sci. 2019, 216, 101–110. [Google Scholar] [CrossRef]
  131. Rebouche, C.J. Kinetics, pharmacokinetics, and regulation of l-carnitine and acetyl-l-carnitine metabolism. Ann. N. Y. Acad. Sci. 2004, 1033, 30–41. [Google Scholar] [CrossRef]
  132. De Leonardis, V.; Neri, B.; Bacalli, S.; Cinelli, P. Reduction of toxicity of anthracyclines by L-carnitine: Preliminary overview of clinical data. Int. J. Clin. Pharmacol. Res. 1985, 5, 137–142. [Google Scholar] [PubMed]
  133. Wainwright, M.S.; Mannix, M.K.; Brown, J.; Stumpf, D.A. L-Carnitine reduces brain injury after hypoxic-ischemia in newborn rats. Pediatric Res. 2003, 54, 688–695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Song, X.; Qu, H.; Yang, Z.; Rong, J.; Cai, W.; Zhou, H. Efficacy and safety of l-carnitine treatment for chronic heart failure: A meta-analysis of randomized controlled trials. Biomed. Res. Int. 2017, 2017, 6274854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Parikh, S.; Goldstein, A.; Kay Koenig, M.; Scaglia, F.; Enns, G.M.; Saneto, R.; Anselm, I.; Cohen, B.H.; Falk, M.J.; Greene, C.; et al. Diagnosis and management of mitochondrial disease: A consensus statement from the Mitochondrial Medicine Society. Genet. Med. 2015, 17, 689–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. López-Domènech, S.; Bañuls, C.; Díaz-Morales, N.; Escribano-López, I.; Morillas, C.; Veses, S.; Orden, S.; Álvarez, Á.; Víctor, V.M.; Hernández-Mijares, A.; et al. Obesity impairs leukocyte-endothelium cell interactions and oxidative stress in humans. Eur. J. Clin. Investig. 2018, 48, e12985. [Google Scholar] [CrossRef]
  137. Alici, D.; Bulbul, F.; Virit, O.; Unal, A.; Altindag, A.; Alpak, G.; Alici, H.; Ermis, B.; Orkmez, M.; Taysi, S.; et al. Evaluation of oxidative metabolism and oxidative DNA damage in patients with obsessive-compulsive disorder. Psychiatry Clin. Neurosci. 2016, 70, 109–115. [Google Scholar] [CrossRef] [Green Version]
Table 1. Reported clinical trials testing adjuvant administration of mitochondrial nutrients (MNs) in Type 1 and Type 2 diabetic patients (reviewed in [7]).
Table 1. Reported clinical trials testing adjuvant administration of mitochondrial nutrients (MNs) in Type 1 and Type 2 diabetic patients (reviewed in [7]).
Mitochondrial NutrientsNo. Studies [Controlled Studies]No. Treated PatientsSuccess Ratio
α-Lipoic acid42 [30]29800.93
Coenzyme Q109 [7]3700.89
(Acyl-)Carnitine13 [9]18941.00

Share and Cite

MDPI and ACS Style

Pagano, G.; Pallardó, F.V.; Porto, B.; Fittipaldi, M.R.; Lyakhovich, A.; Trifuoggi, M. Mitoprotective Clinical Strategies in Type 2 Diabetes and Fanconi Anemia Patients: Suggestions for Clinical Management of Mitochondrial Dysfunction. Antioxidants 2020, 9, 82. https://doi.org/10.3390/antiox9010082

AMA Style

Pagano G, Pallardó FV, Porto B, Fittipaldi MR, Lyakhovich A, Trifuoggi M. Mitoprotective Clinical Strategies in Type 2 Diabetes and Fanconi Anemia Patients: Suggestions for Clinical Management of Mitochondrial Dysfunction. Antioxidants. 2020; 9(1):82. https://doi.org/10.3390/antiox9010082

Chicago/Turabian Style

Pagano, Giovanni, Federico V. Pallardó, Beatriz Porto, Maria Rosa Fittipaldi, Alex Lyakhovich, and Marco Trifuoggi. 2020. "Mitoprotective Clinical Strategies in Type 2 Diabetes and Fanconi Anemia Patients: Suggestions for Clinical Management of Mitochondrial Dysfunction" Antioxidants 9, no. 1: 82. https://doi.org/10.3390/antiox9010082

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