Diabetic periodontitis is a chronic inflammatory disease commonly observed in diabetic patients, characterized by progressive destruction of periodontal tissues.1 This condition not only affects oral health but may also negatively impact the overall control of diabetes, creating a vicious cycle.2 Epidemiological studies indicate that diabetic patients have a significantly higher risk of developing periodontitis compared to non-diabetic individuals. Statistics show that approximately 45% of patients with type 2 diabetes suffer from moderate to severe periodontitis, a proportion far exceeding that of the general population.3
The clinical manifestations of diabetic periodontitis are typically more severe than those in non-diabetic patients. Common symptoms include gingival bleeding, swelling, deepening of periodontal pockets, and accelerated alveolar bone resorption.4 Notably, diabetic periodontitis often progresses more rapidly, exhibits more severe tissue destruction, and responds relatively poorly to conventional treatments.1 These characteristics underscore the necessity of in-depth research into its pathogenic mechanisms to develop more effective prevention and treatment strategies.
The pathological changes of diabetes and their impact on periodontal disease are mainly reflected in the following points: (1) The characteristic of diabetes is chronic hyperglycemia, which has a profound effect on multiple organ systems, including periodontal tissues. Hyperglycemia induces the formation of advanced glycation end products (AGEs), which accumulate in periodontal tissues and bind to their receptors (RAGE).5 This interaction activates inflammatory pathways, including NF-κB, leading to the overproduction of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6.6 (2) Diabetes also impairs the immune system by reducing neutrophil chemotaxis, phagocytosis, and pathogen clearance. This compromised host defense makes individuals with diabetes more susceptible to periodontal infections.7 (3) Endothelial dysfunction caused by diabetes leads to reduced bioavailability of nitric oxide, thereby impairing vascular tone and reducing blood flow to periodontal tissues. This results in delayed wound healing and reduced oxygen delivery to periodontal tissues, exacerbating tissue damage.8 (4) Hyperglycemia increases the production of reactive oxygen species (ROS), leading to oxidative stress and further amplifying the inflammatory response. In periodontal tissues, excessive ROS can damage cellular components such as proteins, lipids, and DNA, ultimately causing cell death and exacerbating periodontal destruction.9,10
Mitochondria, serving as the “power plants” of cells, play a crucial role in maintaining normal cellular functions.11 These organelles are not only responsible for energy production but also participate in various important cellular processes, including calcium ion balance regulation, ROS generation and elimination, and apoptosis regulation.12 The basic structure of mitochondria includes the outer membrane, inner membrane, intermembrane space and matrix.13 In terms of energy metabolism, mitochondria synthesize ATP through the tricarboxylic acid cycle and electron transport chain, providing energy for cells. Additionally, mitochondria play important roles in cellular signal transduction, including the transmission of stress response signals and sensing of metabolic states.14 In recent years, researchers have increasingly recognized the complex communication between mitochondria and cell nucleus, which is crucial for maintaining cellular homeostasis.15
Mitochondrial dysfunction has been confirmed to be closely associated with various chronic diseases, including neurodegenerative diseases, cardiovascular diseases, and metabolic syndrome.16,17 In the context of diabetes, persistent hyperglycemic conditions can lead to significant alterations in mitochondrial function, including mitochondrial DNA (mtDNA) damage, energy metabolism disorders, and increased oxidative stress.18 These changes may play important roles in the occurrence and development of diabetic complications, including diabetic periodontitis.19
In recent years, significant advances have been made in mitochondrial biology research, particularly in the areas of mitochondrial dynamics (fusion and fission), quality control mechanisms, and mitochondria-targeted therapeutic strategies.20,21 This new knowledge provides fresh perspectives for understanding the pathophysiological processes of diabetic periodontitis, while also indicating directions for developing innovative treatment methods.
This review aims to systematically summarize the role and potential mechanisms of mitochondrial dysfunction in diabetic periodontitis. We will first explore the relationship between diabetes and mitochondrial dysfunction, then analyze the specific manifestations of mitochondrial dysfunction in diabetic periodontitis in detail. Next, we will delve into the connections between mitochondrial dysfunction and the pathogenic mechanisms of diabetic periodontitis, including exacerbated inflammatory responses, increased apoptosis, and decreased tissue repair capacity. Finally, we will discuss potential therapeutic targets based on mitochondrial function and propose future research directions. Through this review, we hope not only to provide new insights into understanding the pathogenic mechanisms of diabetic periodontitis but also to provide a theoretical basis for developing targeted prevention and treatment strategies, ultimately improving the oral health and quality of life for diabetic patients.
Diabetes and Mitochondrial DysfunctionDiabetes is a common metabolic disorder characterized by chronic hyperglycemia.22 In recent years, accumulating evidence suggests that mitochondrial dysfunction plays a crucial role in the pathogenesis and progression of diabetes.23–25 This dysfunction not only affects insulin secretion from pancreatic β-cells but also influences insulin sensitivity in peripheral tissues, thereby exacerbating the pathological process of diabetes.
Mitochondrial Metabolic DysfunctionPersistent hyperglycemia exerts multiple adverse effects on intracellular mitochondria. Firstly, high glucose levels lead to over-activation of the mitochondrial electron transport chain, increasing the production of ROS. Excess ROS not only directly damage mtDNA, proteins, and lipids but also trigger a series of inflammatory responses, further exacerbating cellular damage.26 Secondly, hyperglycemia affects mitochondrial energy metabolism function. Studies have found that in diabetic conditions, mitochondrial oxidative phosphorylation efficiency decreases, resulting in reduced ATP production. This energy metabolic disorder may be caused by decreased activity of electron transport chain complexes and impaired mitochondrial inner membrane integrity caused by hyperglycemia.27,28 Additionally, hyperglycemia interferes with mitochondrial calcium ion balance. Mitochondria are important intracellular calcium ion storage organelles, and calcium ion balance is crucial for maintaining mitochondrial function.23 Under diabetic conditions, mitochondrial calcium overload is common, which may lead to mitochondrial membrane potential collapse and trigger cell apoptosis.29
Diabetes-Related MtDNA DamageMtDNA is more susceptible to oxidative stress damage than nuclear DNA due to its unique structure and repair mechanisms.30 In diabetic patients, researchers have found significantly increased rates of mtDNA mutations and deletions.31 These damages may originate from hyperglycemia-induced oxidative stress and the accumulation of AGEs.32
mtDNA damage directly affects the synthesis of mitochondrial proteins, leading to respiratory chain dysfunction and forming a vicious cycle.33 For example, a study on patients with type 2 diabetes found a significant decrease in mtDNA copy number in peripheral blood, which negatively correlated with patients’ blood glucose levels.34 Furthermore, mtDNA damage may exacerbate systemic inflammatory responses by activating inflammatory signaling pathways, such as the NLRP3 inflammasome, which plays an important role in the development of diabetic complications.35
Mitochondrial Dynamics AbnormalitiesMitochondria are highly dynamic organelles, their morphology and function finely regulated by fusion and fission processes.36 This dynamic balance is crucial for maintaining the integrity and function of the mitochondrial network. However, under diabetic conditions, this balance is often disrupted.37 Studies have found that hyperglycemic environments promote the activation of mitochondrial fission proteins (such as Drp1) while inhibiting the expression of fusion proteins (such as Mfn1, Mfn2, and OPA1).38,39 This imbalance leads to fragmentation of the mitochondrial network, affecting mitochondrial function and cellular metabolic state. For instance, in a diabetic nephropathy model, researchers observed significant fragmentation of mitochondria in glomerular podocytes, accompanied by mitochondrial dysfunction and increased cell apoptosis.40,41
Manifestations of Mitochondrial Dysfunction in Diabetic PeriodontitisDiabetic periodontitis is a common oral complication of diabetes, with a complex pathogenesis involving multiple cellular and molecular level alterations.17 In recent years, increasing research has focused on the crucial role of mitochondrial dysfunction in diabetic periodontitis.42 This dysfunction not only affects the metabolic state of periodontal tissues but also participates in the regulation of inflammatory responses and tissue damage processes (Figure 1).43
Figure 1 The main manifestations of mitochondrial dysfunction in diabetic periodontitis.
Morphological Changes of Mitochondria in Periodontal TissuesSignificant morphological changes in mitochondria have been observed in the periodontal tissues of patients with diabetic periodontitis.44 Electron microscopy studies have shown that compared with healthy controls, mitochondria in gingival fibroblasts and periodontal ligament cells of diabetic patients exhibit characteristics of swelling, disorganized cristae structure, and even fragmentation.45 These morphological changes are generally considered to be visual manifestations of mitochondrial dysfunction.46 A study on experimental diabetic rats found that as diabetes progressed, the number of mitochondria decreased while their volume increased.47 This change may reflect the attempt of mitochondria to compensate for functional decline by increasing volume, ultimately leading to the destruction of the mitochondrial network. Furthermore, a significant decrease in mtDNA copy number was observed in the diabetes.48 The reduction in mtDNA copy number is typically closely associated with mitochondrial dysfunction and may lead to decreased mitochondrial protein synthesis and reduced energy production efficiency.49
Mitochondrial Energy Metabolic DisordersMitochondria are the center of cellular energy metabolism, and their dysfunction directly affects cellular energy supply.50 Energy metabolic disorders not only affect normal cellular function but may also trigger adaptive responses. For instance, studies have found increased activity of AMP-activated protein kinase (AMPK) in the periodontal tissues of patients with diabetic.51 AMPK is a cellular energy sensor, and its increased activity may be a compensatory response to mitochondrial dysfunction.52 The tricarboxylic acid (TCA) cycle, a central pathway in mitochondrial energy metabolism, exhibits significant perturbations in diabetic periodontitis. A metabolomic analysis of gingival tissues from patients with diabetic periodontitis revealed substantial alterations in TCA cycle-associated metabolites.53 Notably, elevated levels of citrate and α-ketoglutarate were observed, concurrent with decreased succinate concentrations. These metabolic shifts suggest inhibition of key enzymes of TCA cycle, potentially compromising energy metabolism efficiency.54 The etiology of these metabolic disruptions is postulated to be linked to persistent hyperglycemia-induced enzyme glycation and oxidative stress.55 Moreover, fatty acid β-oxidation, another crucial mitochondrial energy pathway, is similarly impaired in diabetic periodontitis. Under hyperglycemic conditions, periodontal tissue cells demonstrate reduced fatty acid oxidation capacity.56 These alterations may precipitate intracellular lipid accumulation, potentially exacerbating mitochondrial dysfunction and inflammatory responses.57 Such metabolic dysregulation underscores the complex interplay between diabetes and periodontal pathology, highlighting potential therapeutic targets for intervention.
Increased Mitochondria-Related Oxidative StressOxidative stress is a key factor in the pathogenesis of diabetic periodontitis, and mitochondria are the main source of ROS in cells.58 In diabetic periodontitis, mitochondrial dysfunction leads to increased ROS production, while the antioxidant defense system is impaired, ultimately resulting in exacerbated oxidative stress.54 Studies have found that in the gingival tissues of patients with diabetic periodontitis, the activity of mitochondrial superoxide dismutase (MnSOD) is significantly reduced, while lipid peroxidation levels are elevated.59 This disruption of the oxidative-antioxidative balance not only directly damages cellular components but may also activate inflammatory signaling pathways, such as the NF-κB pathway, further exacerbating the inflammatory response.60 Moreover, the increase in mitochondria-derived ROS may lead to oxidative damage of mtDNA.61 A study on diabetic mice showed significantly elevated levels of 8-hydroxy-2’-deoxyguanosine (8-OHdG), a marker of DNA oxidative damage, in periodontal tissues.62 Damage to mtDNA may further exacerbate mitochondrial dysfunction, forming a vicious cycle.63
Increased Mitochondria-Mediated Cell ApoptosisMitochondria play a central role in regulating the apoptosis process.64 In diabetes, researchers have observed a significant increase in apoptosis in periodontal tissue cells, which may be closely related to mitochondrial dysfunction.65 Specifically, in the periodontal tissue of rats with diabetes, the expression of pro-apoptotic protein Bax is increased, while the expression of anti-apoptotic protein Bcl-2 is decreased.66 These changes lead to increased mitochondrial membrane permeability, promoting the release of cytochrome c, which in turn activates downstream caspase cascades, ultimately resulting in cell apoptosis.67 Furthermore, mitochondrial dysfunction may promote cell apoptosis through other mechanisms. For example, mitochondrial calcium ion homeostasis imbalance may activate calcium-dependent proteases, triggering the apoptosis process.29 Additionally, the increase in mitochondria-derived ROS may directly damage DNA, activating p53-mediated apoptotic pathways.68
Mitochondrial Dysfunction and Pathogenic Mechanisms of Diabetic PeriodontitisMitochondrial dysfunction plays a central role in the pathogenic mechanisms of diabetic periodontitis, influencing disease progression through multiple pathways.69 These mechanisms interact to form a complex network, ultimately leading to periodontal tissue destruction and functional loss. A thorough understanding of these mechanisms is crucial for developing new prevention and treatment strategies (Figure 2).
Figure 2 Pathogenic Mechanisms and Potential Therapeutic Targets for Mitochondrial Dysfunction in Diabetic Periodontitis.
Exacerbation of Inflammatory ResponsesMitochondrial dysfunction is a significant factor contributing to the exacerbation of inflammatory responses in diabetic periodontitis.54 Firstly, excessive production of mitochondria-derived ROS can activate various inflammatory signaling pathways, such as NF-κB and the NLRP3 inflammasome.70 A study on diabetic rat periodontal tissues found that ROS increase due to mitochondrial dysfunction positively correlated with NF-κB activation, subsequently promoting the expression of inflammatory factors IL-1β, TNF-α, and IL-6.71
Secondly, the release of mtDNA can act as damage-associated molecular patterns (DAMPs), triggering innate immune responses.72 In the gingival tissues of patients with chronic periodontitis, researchers detected significantly elevated levels of circulating mtDNA, which positively correlated with inflammatory marker levels.73 This mtDNA release may be caused by impaired mitochondrial membrane integrity. Furthermore, mitochondrial dysfunction may regulate inflammatory responses by influencing the metabolic reprogramming of immune cells.74 For instance, under diabetic conditions, impaired mitochondrial oxidative phosphorylation in macrophages leads to a preference for glycolytic metabolism, which is associated with a pro-inflammatory phenotype.75 An in vitro study demonstrated that targeted improvement of macrophage mitochondrial function could significantly alleviate high glucose-induced inflammatory responses.76
Decreased Periodontal Tissue Repair CapacityMitochondrial dysfunction also affects the repair capacity of periodontal tissues, playing a crucial role in the sustained progression of diabetic periodontitis.77 Firstly, mitochondrial dysfunction impacts the self-renewal and differentiation abilities of periodontal ligament stem cells (PDLSCs).78 A study found that PDLSCs derived from diabetic patients exhibited significant mitochondrial dysfunction, including decreased mitochondrial membrane potential and reduced ATP production, which was closely related to their diminished osteogenic differentiation capacity.79 Secondly, mitochondrial dysfunction affects the balance between synthesis and degradation of the extracellular matrix (ECM).80 This imbalance ultimately results in excessive ECM degradation and reduced periodontal tissue support capacity.81 Additionally, mitochondrial dysfunction interferes with the blood supply to periodontal tissues by affecting vascular endothelial cell function.82 Studies have shown that mitochondrial dysfunction in vascular endothelial cells under hyperglycemic conditions leads to decreased expression of angiogenic factors (such as VEGF), ultimately affecting vascular regeneration during tissue repair processes.83
Autophagy DysregulationAutophagy is an important mechanism for cellular stress response, with mitophagy being a key process for clearing damaged mitochondria.84 In diabetic periodontitis, there is a complex interaction between mitochondrial dysfunction and autophagy dysregulation.85 On one hand, persistent mitochondrial dysfunction can lead to overactivation of the autophagy pathway, potentially resulting in autophagic cell death.86 In the periodontal tissues of diabetic rats, researchers observed significantly increased expression of autophagy markers LC3-II and p62, which positively correlated with the degree of tissue damage.87 On the other hand, long-term hyperglycemic environments may lead to impaired autophagy function, especially in the mitophagy process.88 This decline in autophagy function results in the accumulation of damaged mitochondria, further exacerbating oxidative stress and inflammatory responses.89 An in vitro study showed that pharmacological activation of PINK1/Parkin-mediated mitophagy could significantly improve mitochondrial function and cell viability in periodontal ligament cells under high glucose conditions.90
Potential Therapeutic Targets for Mitochondrial Dysfunction in Diabetic PeriodontitisAs our understanding of the role of mitochondrial dysfunction in diabetic periodontitis deepens, mitochondria-targeted therapeutic strategies have gradually become a research hotspot. These strategies primarily focus on reducing oxidative stress, improving mitochondrial energy metabolism, regulating mitochondrial dynamics, and promoting mitochondrial biogenesis.56,91
Antioxidant StrategiesGiven the crucial role of oxidative stress in diabetic periodontitis, mitochondria-targeted antioxidant strategies have become an important therapeutic direction.92
Mitochondria-Targeted AntioxidantsTraditional antioxidants have shown limited efficacy in treating diabetic periodontitis, partly due to their difficulty in effectively entering mitochondria.93 Consequently, researchers have developed a series of mitochondria-targeted antioxidants, such as MitoQ, SkQ1, and SS-31.94 These compounds can specifically accumulate in mitochondria, effectively scavenging excess ROS.95
Nrf2 ActivatorsNuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor regulating the expression of antioxidant genes.96 In diabetic periodontitis, Nrf2 activity is often suppressed.97 Therefore, activating Nrf2 has become a potential therapeutic strategy.
Research has found that some natural compounds such as curcumin and resveratrol can improve diabetic periodontitis by activating the Nrf2 pathway.98 For example, a clinical trial showed that oral curcumin supplementation could significantly reduce gingival inflammation index and periodontal pocket depth in diabetic patients.99
Mitochondria-Targeted DrugsIn addition to antioxidant strategies, drugs directly improving mitochondrial function have also shown therapeutic potential.100
Mitochondrial Metabolism RegulatorsMitochondrial energy metabolism disorder is an important feature of diabetic periodontitis.101 Thus, regulating mitochondrial metabolism has become a potential therapeutic target. Metformin, a widely used antidiabetic drug, has recently been found to improve mitochondrial function.102 It has been shown that metformin improved mitochondrial function by activating the AMPK pathway, thereby alleviating inflammatory responses.103 Another potential metabolic regulator is pyrroloquinoline quinone (PQQ), which can promote mitochondrial biogenesis.104
Mitochondrial Dynamics RegulatorsGiven the role of mitochondrial dynamics abnormalities in diabetic periodontitis, regulating mitochondrial fusion and fission processes has become a new therapeutic direction.92 For example, the mitochondrial fission inhibitor Mdivi-1 induces mitochondrial fragmentation induced by P. gingivalis infection, increased the mtROS levels, and decreased the MMP and ATP concentration in vascular endothelial cells.105 On the other hand, promoting mitochondrial fusion may also have therapeutic effects. Research has found that overexpression of the mitochondrial fusion protein Mfn2 can improve mitochondrial function and inflammatory status in the periodontal tissues of diabetic mice.106
Autophagy RegulatorsConsidering the important role of autophagy in maintaining mitochondrial function, regulating the autophagy process has also become a potential therapeutic strategy.107
Rapamycin and Its AnaloguesRapamycin is a classic autophagy activator that promotes autophagy by inhibiting the mTOR pathway.108 Research has found that low-dose rapamycin can improve the periodontal condition of diabetic mice, which may be related to its role in promoting mitophagy and clearing damaged mitochondria.109,110
SIRT1 ActivatorsSIRT1 is an important metabolic sensor involved in regulating autophagy and mitochondrial function.111 Based on existing studies, it is hypothesized that SIRT1 activators such as resveratrol may alleviate symptoms of diabetic periodontitis by promoting autophagy and improving mitochondrial function.112,113
Conclusions and Future DirectionsThis review highlights the pivotal role of mitochondrial dysfunction in diabetic periodontitis. Mitochondrial impairment induced by diabetes, including DNA damage, disrupted energy metabolism, increased oxidative stress, and abnormal dynamics, not only compromises periodontal tissue function but also triggers inflammatory cascades, accelerating tissue destruction. We have identified several promising therapeutic strategies targeting mitochondrial function, such as antioxidants, metabolism regulators, and autophagy modulators. These approaches offer new possibilities for managing diabetic periodontitis. However, further research is needed to validate these treatments in clinical settings and to explore personalized approaches based on mitochondrial status.
In summary, understanding mitochondrial dysfunction in diabetic periodontitis provides valuable insights into the mechanisms of diseases. Future research endeavors should prioritize the translation of basic scientific findings into clinical practice, the development of more efficacious and well-tolerated mitochondria-targeted therapeutic modalities, and the exploration of potential links between mitochondrial dysfunction and other diabetic complications. These efforts aim to formulate more comprehensive and integrated treatment strategies for individuals with diabetes mellitus and its associated periodontal manifestations.
Author ContributionsAll authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
FundingThis work was supported by the Young and Middle-Aged Scientific Research Fund of Wannan medical college (WYRCQD2023028), Anhui Province Engineering Research Center for dental materials and application (Grant No: 2024AMCD06), Program for Excellent Sci-tech innovation Teams of Universities in Anhui Province (Grant No: 2023AH010073).
DisclosureThe authors declare that they have no competing interest.
References1. Zhao M, Xie Y, Gao W, Li C, Ye Q, Li Y. Diabetes mellitus promotes susceptibility to periodontitis-novel insight into the molecular mechanisms. Front Endocrinol. 2023;14:1192625. doi:10.3389/fendo.2023.1192625
2. Preshaw PM, Alba AL, Herrera D, et al. Periodontitis and diabetes: a two-way relationship. Diabetologia. 2012;55(1):21–31. doi:10.1007/s00125-011-2342-y
3. Chien WC, Fu E, Chung CH, et al. Type 2 diabetes mellitus and periodontitis: bidirectional association in population-based 15-year retrospective cohorts. J Clin Endocrinol Metab. 2023;108(11):e1289–e1297. doi:10.1210/clinem/dgad287
4. Rohani B. Oral manifestations in patients with diabetes mellitus. World J Diabetes. 2019;10(9):485–489. doi:10.4239/wjd.v10.i9.485
5. Kido R, Hiroshima Y, Kido JI, et al. Advanced glycation end-products increase lipocalin 2 expression in human oral epithelial cells. J Periodontal Res. 2020;55(4):539–550. doi:10.1111/jre.12741
6. Nonaka K, Kajiura Y, Bando M, et al. Advanced glycation end-products increase IL-6 and ICAM-1 expression via RAGE, MAPK and NF-κB pathways in human gingival fibroblasts. J Periodontal Res. 2018;53(3):334–344. doi:10.1111/jre.12518
7. Warren KR, Postolache TT, Groer ME, Pinjari O, Kelly DL, Reynolds MA. Role of chronic stress and depression in periodontal diseases. Periodontol. 2014;64(1):127–138. doi:10.1111/prd.12036
8. Wang L, Li X, Zhang Y, Huang Y, Zhang Y, Ma Q. Oxymatrine ameliorates diabetes-induced aortic endothelial dysfunction via the regulation of eNOS and NOX4. J Cell Biochem. 2019;120(5):7323–7332. doi:10.1002/jcb.28006
9. Lu S, Liao Z, Lu X, et al. Hyperglycemia acutely increases cytosolic reactive oxygen species via O-linked GlcNAcylation and CaMKII activation in mouse ventricular myocytes. Circ Res. 2020;126(10):e80–e96. doi:10.1161/CIRCRESAHA.119.316288
10. Lin F, Yang Y, Wei S, et al. Hydrogen sulfide protects against high glucose-induced human umbilical vein endothelial cell injury through activating PI3K/Akt/eNOS pathway. Drug Des Devel Ther. 2020;14:621–633.
11. Popov LD. Mitochondria as intracellular signalling organelles. An update. Cell Signal. 2023;109:110794. doi:10.1016/j.cellsig.2023.110794
12. Mailloux RJ. An update on methods and approaches for interrogating mitochondrial reactive oxygen species production. Redox Biol. 2021;45:102044. doi:10.1016/j.redox.2021.102044
13. Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev. 2023;103(4):2349–2422. doi:10.1152/physrev.00058.2021
14. Chan DC. Mitochondrial dynamics and its involvement in disease. Annu Rev Pathol. 2020;15:235–259. doi:10.1146/annurev-pathmechdis-012419-032711
15. Suomalainen A, Nunnari J. Mitochondria at the crossroads of health and disease. Cell. 2024;187(11):2601–2627. doi:10.1016/j.cell.2024.04.037
16. Blagov A, Nedosugova L, Kirichenko T, Sukhorukov V, Melnichenko A, Orekhov A. Mitochondrial dysfunction as a factor of energy metabolism disorders in type 2 diabetes mellitus. Front Biosci. 2024;16(1):5. doi:10.31083/j.fbs1601005
17. Deng Y, Xiao J, Ma L, et al. Mitochondrial dysfunction in periodontitis and associated systemic diseases: implications for pathomechanisms and therapeutic strategies. Int J Mol Sci. 2024;25(2):1024. doi:10.3390/ijms25021024
18. Bergamini C, Bonora E, Moruzzi N. Editorial: mitochondrial bioenergetics impairments in genetic and metabolic diseases. Front Physiol. 2023;14:1228926. doi:10.3389/fphys.2023.1228926
19. Behl T, Makkar R, Anwer MK, et al. Mitochondrial dysfunction: a cellular and molecular hub in pathology of metabolic diseases and infection. J Clin Med. 2023;12(8):2882. doi:10.3390/jcm12082882
20. Chaudhry A, Shi R, Luciani DS. A pipeline for multidimensional confocal analysis of mitochondrial morphology, function, and dynamics in pancreatic β-cells. Am J Physiol Endocrinol Metab. 2020;318(2):E87–e101. doi:10.1152/ajpendo.00457.2019
21. Rönn T, Ofori JK, Perfilyev A, et al. Genes with epigenetic alterations in human pancreatic islets impact mitochondrial function, insulin secretion, and type 2 diabetes. Nat Commun. 2023;14(1):8040. doi:10.1038/s41467-023-43719-9
22. Prasun P. Mitochondrial dysfunction in metabolic syndrome. Biochim Biophys Acta Mol Basis Dis. 2020;1866(10):165838. doi:10.1016/j.bbadis.2020.165838
23. Belosludtsev KN, Belosludtseva NV, Dubinin MV. Diabetes mellitus, mitochondrial dysfunction and Ca(2+)-dependent permeability transition pore. Int J Mol Sci. 2020;21(18):6559. doi:10.3390/ijms21186559
24. Tang W, Yan C, He S, et al. Neuron-targeted overexpression of caveolin-1 alleviates diabetes-associated cognitive dysfunction via regulating mitochondrial fission-mitophagy axis. Cell Commun Signal. 2023;21(1):357. doi:10.1186/s12964-023-01328-5
25. Xu D, Jiang Z, Sun Z, et al. Mitochondrial dysfunction and inhibition of myoblast differentiation in mice with high-fat-diet-induced pre-diabetes. J Cell Physiol. 2019;234(5):7510–7523. doi:10.1002/jcp.27512
26. Zhang Z, Huang Q, Zhao D, Lian F, Li X, Qi W. The impact of oxidative stress-induced mitochondrial dysfunction on diabetic microvascular complications. Front Endocrinol. 2023;14:1112363. doi:10.3389/fendo.2023.1112363
27. Audzeyenka I, Rachubik P, Typiak M, et al. Piwkowska A: hyperglycemia alters mitochondrial respiration efficiency and mitophagy in human podocytes. Exp Cell Res. 2021;407(1):112758. doi:10.1016/j.yexcr.2021.112758
28. Chen W, Zhao H, Li Y. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct Target Ther. 2023;8(1):333. doi:10.1038/s41392-023-01547-9
29. Giorgi C, Marchi S, Pinton P. The machineries, regulation and cellular functions of mitochondrial calcium. Nat Rev Mol Cell Biol. 2018;19(11):713–730. doi:10.1038/s41580-018-0052-8
30. Liao S, Chen L, Song Z, He H. The fate of damaged mitochondrial DNA in the cell. Biochim Biophys Acta Mol Cell Res. 2022;1869(5):119233. doi:10.1016/j.bbamcr.2022.119233
31. Al-Ghamdi BA, Al-Shamrani JM, El-Shehawi AM, Al-Johani I, Al-Otaibi BG. Role of mitochondrial DNA in diabetes mellitus type I and type II. Saudi J Biol Sci. 2022;29(12):103434. doi:10.1016/j.sjbs.2022.103434
32. Stopper H, Schupp N, Bahner U, Sebekova K, Klassen A, Heidland A. Genomic damage in end-stage renal failure: potential involvement of advanced glycation end products and carbonyl stress. Semin Nephrol. 2004;24(5):474–478.
33. Wang Y, Wang J, Tao SY, et al. Mitochondrial damage-associated molecular patterns: a new insight into metabolic inflammation in type 2 diabetes mellitus. Diabetes/Metab Res Rev. 2024;40(2):e3733. doi:10.1002/dmrr.3733
34. Memon AA, Sundquist J, Hedelius A, Palmér K, Wang X, Sundquist K. Association of mitochondrial DNA copy number with prevalent and incident type 2 diabetes in women: a population-based follow-up study. Sci Rep. 2021;11(1):4608. doi:10.1038/s41598-021-84132-w
35. Li W, Li Y, Zhao J, et al. Release of damaged mitochondrial DNA: a novel factor in stimulating inflammatory response. Pathol Res Pract. 2024;258:155330. doi:10.1016/j.prp.2024.155330
36. Vásquez-Trincado C, García-Carvajal I, Pennanen C, et al. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol. 2016;594(3):509–525. doi:10.1113/JP271301
37. Wang S, Zhao H, Lin S, et al. New therapeutic directions in type II diabetes and its complications: mitochondrial dynamics. Front Endocrinol. 2023;14:1230168. doi:10.3389/fendo.2023.1230168
38. Wang W, Wang Y, Long J, et al. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab. 2012;15(2):186–200. doi:10.1016/j.cmet.2012.01.009
39. Kumari S, Anderson L, Farmer S, Mehta SL, Li PA. Hyperglycemia alters mitochondrial fission and fusion proteins in mice subjected to cerebral ischemia and reperfusion. Transl Stroke Res. 2012;3(2):296–304. doi:10.1007/s12975-012-0158-9
40. Aluksanasuwan S, Plumworasawat S, Malaitad T, Chaiyarit S, Thongboonkerd V. High glucose induces phosphorylation and oxidation of mitochondrial proteins in renal tubular cells: a proteomics approach. Sci Rep. 2020;10(1):5843. doi:10.1038/s41598-020-62665-w
41. Audzeyenka I, Rachubik P, Rogacka D, Saleem MA, Piwkowska A. Insulin induces bioenergetic changes and alters mitochondrial dynamics in podocytes. J Endocrinol. 2024;261(3). doi:10.1530/JOE-23-0357
42. Serón C, Olivero P, Flores N, et al. Diabetes, periodontitis, and cardiovascular disease: towards equity in diabetes care. Front Public Health. 2023;11:1270557. doi:10.3389/fpubh.2023.1270557
43. Luo S, Xu T, Zheng Q, et al. Mitochondria: an emerging unavoidable link in the pathogenesis of periodontitis caused by porphyromonas gingivalis. Int J Mol Sci. 2024;25(2):737.
44. Kırmızıgül ÖA, Sabanci A, Dişli F, Yıldız S, Milward MR, Aral K. Evaluation of the role of mitofusin-1 and mitofusin-2 in periodontal disease. J Periodontol. 2024;95(1):64–73. doi:10.1002/JPER.23-0072
45. Govindaraj P, Khan NA, Gopalakrishna P, et al. Mitochondrial dysfunction and genetic heterogeneity in chronic periodontitis. Mitochondrion. 2011;11(3):504–512. doi:10.1016/j.mito.2011.01.009
46. Giacomello M, Pyakurel A, Glytsou C, Scorrano L. The cell biology of mitochondrial membrane dynamics. Nat Rev Mol Cell Biol. 2020;21(4):204–224. doi:10.1038/s41580-020-0210-7
47. Ma Z, Wirström T, Borg LA, et al. Diabetes reduces β-cell mitochondria and induces distinct morphological abnormalities, which are reproducible by high glucose in vitro with attendant dysfunction. Islets. 2012;4(3):233–242. doi:10.4161/isl.20516
48. Song J, Oh JY, Sung YA, Pak YK, Park KS, Lee HK. Peripheral blood mitochondrial DNA content is related to insulin sensitivity in offspring of type 2 diabetic patients. Diabetes Care. 2001;24(5):865–869. doi:10.2337/diacare.24.5.865
49. Malik AN, Czajka A. Is mitochondrial DNA content a potential biomarker of mitochondrial dysfunction? Mitochondrion. 2013;13(5):481–492. doi:10.1016/j.mito.2012.10.011
50. Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell. 2012;148(6):1145–1159. doi:10.1016/j.cell.2012.02.035
51. Guo ZL, Zhou J, Lin XJ, et al. Regulation of the AGEs-induced inflammatory response in human periodontal ligament cells via the AMPK/NF-κB/ NLRP3 signaling pathway. Exp Cell Res. 2024;437(1):113999. doi:10.1016/j.yexcr.2024.113999
52. Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13(4):251–262. doi:10.1038/nrm3311
53. Barnes VM, Kennedy AD, Panagakos F, et al. Global metabolomic analysis of human saliva and plasma from healthy and diabetic subjects, with and without periodontal disease. PLoS One. 2014;9(8):e105181. doi:10.1371/journal.pone.0105181
54. Bullon P, Newman HN, Battino M. Obesity, diabetes mellitus, atherosclerosis and chronic periodontitis: a shared pathology via oxidative stress and mitochondrial dysfunction? Periodontol. 2014;64(1):139–153. doi:10.1111/j.1600-0757.2012.00455.x
55. Lima J, Moreira NCS, Sakamoto-Hojo ET. Mechanisms underlying the pathophysiology of type 2 diabetes: from risk factors to oxidative stress, metabolic dysfunction, and hyperglycemia. Mutat Res. 2022;874-875:503437. doi:10.1016/j.mrgentox.2021.503437
56. Bhansali S, Bhansali A, Walia R, Saikia UN, Dhawan V. Alterations in mitochondrial oxidative stress and mitophagy in subjects with prediabetes and type 2 diabetes mellitus. Front Endocrinol. 2017;8:347. doi:10.3389/fendo.2017.00347
57. Xiao E, Mattos M, Vieira GHA, et al. Diabetes enhances IL-17 expression and alters the oral microbiome to increase its pathogenicity. Cell Host Microbe. 2017;22(1):120–128.e124. doi:10.1016/j.chom.2017.06.014
58. Sczepanik FSC, Grossi ML, Casati M, et al. Periodontitis is an inflammatory disease of oxidative stress: we should treat it that way. Periodontol. 2020;84(1):45–68. doi:10.1111/prd.12342
59. Wei D, Zhang XL, Wang YZ, Yang CX, Chen G. Lipid peroxidation levels, total oxidant status and superoxide dismutase in serum, saliva and gingival crevicular fluid in chronic periodontitis patients before and after periodontal therapy. Aust Dent J. 2010;55(1):70–78. doi:10.1111/j.1834-7819.2009.01123.x
60. Priya Dharshini LC, Vishnupriya S, Sakthivel KM, Rasmi RR. Oxidative stress responsive transcription factors in cellular signalling transduction mechanisms. Cell Signal. 2020;72:109670. doi:10.1016/j.cellsig.2020.109670
61. Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, Alexeyev MF. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 2009;37(8):2539–2548. doi:10.1093/nar/gkp100
62. Tang L, Li T, Chang Y, et al. Diabetic oxidative stress-induced telomere damage aggravates periodontal bone loss in periodontitis. Biochem Biophys Res Commun. 2022;614:22–28. doi:10.1016/j.bbrc.2022.04.039
63. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci USA. 1997;94(2):514–519. doi:10.1073/pnas.94.2.514
64. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011;333(6046):1109–1112. doi:10.1126/science.1201940
65. Graves DT, Liu R, Alikhani M, Al-Mashat H, Trackman PC. Diabetes-enhanced inflammation and apoptosis--impact on periodontal pathology. J Dent Res. 2006;85(1):15–21. doi:10.1177/154405910608500103
66. Mei Y, Shen X, Wang X, et al. Expression of autophagy and apoptosis-related factors in the periodontal tissue of experimental diabetic rats: a histomorphometric, microtomographic and immunohistochemical study. PeerJ. 2021;9:e11577. doi:10.7717/peerj.11577
67. Circu ML, Aw TY. Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med. 2010;48(6):749–762. doi:10.1016/j.freeradbiomed.2009.12.022
68. Liu B, Chen Y, St Clair DK. ROS and p53: a versatile partnership. Free Radic Biol Med. 2008;44(8):1529–1535. doi:10.1016/j.freeradbiomed.2008.01.011
69. Portes J, Bullón B, Quiles JL, Battino M, Bullón P. Diabetes mellitus and periodontitis share intracellular disorders as the main meeting point. Cells. 2021;10(9):2411. doi:10.3390/cells10092411
70. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469(7329):221–225.
71. Kang J, de Brito Bezerra B, Pacios S, et al. Aggregatibacter actinomycetemcomitans infection enhances apoptosis in vivo through a caspase-3-dependent mechanism in experimental periodontitis. Infect Immun. 2012;80(6):2247–2256. doi:10.1128/IAI.06371-11
72. Lin MM, Liu N, Qin ZH, Wang Y. Mitochondrial-derived damage-associated molecular patterns amplify neuroinflammation in neurodegenerative diseases. Acta Pharmacol Sin. 2022;43(10):2439–2447. doi:10.1038/s41401-022-00879-6
73. Liu J, Wang X, Xue F, Zheng M, Luan Q. Abnormal mitochondrial structure and function are retained in gingival tissues and human gingival fibroblasts from patients with chronic periodontitis. J Periodontal Res. 2022;57(1):94–103. doi:10.1111/jre.12941
74. Van den Bossche J, O’Neill LA, Menon D. Macrophage immunometabolism: where are we (going)? Trends Immunol. 2017;38(6):395–406. doi:10.1016/j.it.2017.03.001
75. Bae YS, Lee JH, Choi SH, et al. Macrophages generate reactive oxygen species in response to minimally oxidized low-density lipoprotein: toll-like receptor 4- and spleen tyrosine kinase-dependent activation of NADPH oxidase 2. Circ Res. 2009;104(2):210–218. doi:10.1161/CIRCRESAHA.108.181040
76. Mills EL, Kelly B, Logan A, et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell. 2016;167(2):457–470.e413. doi:10.1016/j.cell.2016.08.064
77. Liu R, Desta T, He H, Graves DT. Diabetes alters the response to bacteria by enhancing fibroblast apoptosis. Endocrinology. 2004;145(6):2997–3003. doi:10.1210/en.2003-1601
78. Zheng DH, Wang XX, Ma D, Zhang LN, Qiao QF, Zhang J. Erythropoietin enhances osteogenic differentiation of human periodontal ligament stem cells via Wnt/β-catenin signaling pathway. Drug Des Devel Ther. 2019;13:2543–2552. doi:10.2147/DDDT.S214116
79. Kim MW, An BM, Wang L, Lee KY, Bang J, Joo BS. Effects of oxysterols on chondrogenesis of human adipose-derived stem cells. Ann Clin Lab Sci. 2020;50(2):190–198.
80. Bullon P, Cordero MD, Quiles JL, Morillo JM, Del Carmen Ramirez-Tortosa M, Battino M. Mitochondrial dysfunction promoted by Porphyromonas gingivalis lipopolysaccharide as a possible link between cardiovascular disease and periodontitis. Free Radic Biol Med. 2011;50(10):1336–1343. doi:10.1016/j.freeradbiomed.2011.02.018
81. Graves DT, Li J, Cochran DL. Inflammation and uncoupling as mechanisms of periodontal bone loss. J Dent Res. 2011;90(2):143–153. doi:10.1177/0022034510385236
82. Sena CM, Pereira AM, Seiça R. Endothelial dysfunction - a major mediator of diabetic vascular disease. BBA. 2013;1832(12):2216–2231. doi:10.1016/j.bbadis.2013.08.006
83. Bitar MS, Al-Mulla F. A defect in Nrf2 signaling constitutes a mechanism for cellular stress hypersensitivity in a genetic rat model of type 2 diabetes. Am J Physiol Endocrinol Metab. 2011;301(6):E1119–1129. doi:10.1152/ajpendo.00047.2011
84. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42. doi:10.1016/j.cell.2007.12.018
85. Bullon P, Cordero MD, Quiles JL, et al. Autophagy in periodontitis patients and gingival fibroblasts: unraveling the link between chronic diseases and inflammation. BMC Med. 2012;10:122. doi:10.1186/1741-7015-10-122
86. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451(7182):1069–1075. doi:10.1038/nature06639
87. An Y, Liu WJ, Xue P, et al. Autophagy promotes MSC-mediated vascularization in cutaneous wound healing via regulation of VEGF secretion. Cell Death Dis. 2018;9(2):58. doi:10.1038/s41419-017-0082-8
88. Xie Z, Lau K, Eby B, et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes. 2011;60(6):1770–1778. doi:10.2337/db10-0351
89. Li Y, Zheng W, Lu Y, et al. BNIP3L/NIX-mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis. 2021;13(1):14. doi:10.1038/s41419-021-04469-y
90. Zhang Y, Wang Y, Xu J, et al. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res. 2019;66(2):e12542. doi:10.1111/jpi.12542
91. Wang Y, Andrukhov O, Rausch-Fan X. Oxidative stress and antioxidant system in periodontitis. Front Physiol. 2017;8:910. doi:10.3389/fphys.2017.00910
92. Liesa M, Shirihai OS. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab. 2013;17(4):491–506. doi:10.1016/j.cmet.2013.03.002
93. Chapple IL, Matthews JB. The role of reactive oxygen and antioxidant species in periodontal tissue destruction. Periodontol. 2007;43:160–232. doi:10.1111/j.1600-0757.2006.00178.x
94. Smith RA, Murphy MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann NY Acad Sci. 2010;1201:96–103. doi:10.1111/j.1749-6632.2010.05627.x
95. Oyewole AO, Birch-Machin MA. Mitochondria-targeted antioxidants. FASEB J. 2015;29(12):4766–4771. doi:10.1096/fj.15-275404
96. Ma Q. Role of nrf2 in oxidative stress and toxicity. Ann Rev Pharmacol Toxicol. 2013;53:401–426. doi:10.1146/annurev-pharmtox-011112-140320
97. Sima C, Aboodi GM, Lakschevitz FS, Sun C, Goldberg MB, Glogauer M. Nuclear factor erythroid 2-related factor 2 down-regulation in oral neutrophils is associated with periodontal oxidative damage and severe chronic periodontitis. Am J Pathol. 2016;186(6):1417–1426. doi:10.1016/j.ajpath.2016.01.013
98. Tamaki N, Cristina Orihuela-Campos R, Inagaki Y, Fukui M, Nagata T, Ito HO. Resveratrol improves oxidative stress and prevents the progression of periodontitis via the activation of the Sirt1/AMPK and the Nrf2/antioxidant defense pathways in a rat periodontitis model. Free Radic Biol Med. 2014;75:222–229. doi:10.1016/j.freeradbiomed.2014.07.034
99. Arunachalam LT, Sudhakar U, Vasanth J, Khumukchum S, Selvam VV. Comparison of anti-plaque and anti-gingivitis effect of curcumin and chlorhexidine mouth rinse in the treatment of gingivitis: a clinical and biochemical study. J Indian Soc Periodontol. 2017;21(6):478–483. doi:10.4103/jisp.jisp_116_17
100. Yamada Y, Hibino M, Sasaki D, Abe J, Harashima H. Power of mitochondrial drug delivery systems to produce innovative nanomedicines. Adv Drug Delivery Rev. 2020;154-155:187–209. doi:10.1016/j.addr.2020.09.010
101. Pan S, Yang L, Zhong W, et al. Integrated analyses revealed the potential role and immune link of mitochondrial dysfunction between periodontitis and type 2 diabetes mellitus. Int immunopharmacol. 2024;130:111796. doi:10.1016/j.intimp.2024.111796
102. Bharath LP, Nikolajczyk BS. The intersection of metformin and inflammation. Am J Physiol Cell Physiol. 2021;320(5):C873–c879. doi:10.1152/ajpcell.00604.2020
103. Guo Y, Jiang H, Wang M, Ma Y, Zhang J, Jing L. Metformin alleviates cerebral ischemia/reperfusion injury aggravated by hyperglycemia via regulating AMPK/ULK1/PINK1/Parkin pathway-mediated mitophagy and apoptosis. Chem Biol Interact. 2023;384:110723. doi:10.1016/j.cbi.2023.110723
104. Harris CB, Chowanadisai W, Mishchuk DO, Satre MA, Slupsky CM, Rucker RB. Dietary pyrroloquinoline quinone (PQQ) alters indicators of inflammation and mitochondrial-related metabolism in human subjects. J Nutr Biochem. 2013;24(12):2076–2084. doi:10.1016/j.jnutbio.2013.07.008
105. Xu T, Dong Q, Luo Y, et al. Porphyromonas gingivalis infection promotes mitochondrial dysfunction through Drp1-dependent mitochondrial fission in endothelial cells. Int J Oral Sci. 2021;13(1):28. doi:10.1038/s41368-021-00134-4
106. Li S, Sun X, Xu L, et al. Baicalin attenuates in vivo and in vitro hyperglycemia-exacerbated ischemia/reperfusion injury by regulating mitochondrial function in a manner dependent on AMPK. Eur J Pharmacol. 2017;815:118–126. doi:10.1016/j.ejphar.2017.07.041
107. Li A, Gao M, Liu B, et al. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022;13(5):444. doi:10.1038/s41419-022-04906-6
108. Notte A, Ninane N, Arnould T. Michiels C: Hypoxia counteracts taxol-induced apoptosis in MDA-MB-231 breast cancer cells: role of autophagy and JNK ac
留言 (0)