#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Plasma mitochondrial DNA is elevated in obese type 2 diabetes mellitus patients and correlates positively with insulin resistance


Authors: Larysa V. Yuzefovych aff001;  Viktor M. Pastukh aff001;  Mykhaylo V. Ruchko aff001;  Jon D. Simmons aff001;  William O. Richards aff002;  Lyudmila I. Rachek aff001
Authors place of work: Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama, United States of America aff001;  Department of Surgery, College of Medicine, University of South Alabama, Mobile, Alabama, United States of America aff002
Published in the journal: PLoS ONE 14(10)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0222278

Summary

Cells damaged by mechanical or infectious injury release proinflammatory mitochondrial DNA (mtDNA) fragments into the circulation. We evaluated the relation between plasma levels of mtDNA fragments in obese type 2 diabetes mellitus (T2DM) patients and measures of chronic inflammation and insulin resistance. In 10 obese T2DM patients and 12 healthy control (HC) subjects, we measured levels of plasma cell-free mtDNA with quantitative real-time polymerase chain reaction, and mtDNA damage in skeletal muscle with quantitative alkaline Southern blot. Also, markers of systemic inflammation and oxidative stress in skeletal muscle were measured. Plasma levels of mtDNA fragments, mtDNA damage in skeletal muscle and plasma tumor necrosis factor α levels were greater in obese T2DM patients than HC subjects. Also, the abundance of plasma mtDNA fragments in obese T2DM patients levels positively correlated with insulin resistance. To the best of our knowledge, this is the first published evidence that elevated level of plasma mtDNA fragments is associated with mtDNA damage and oxidative stress in skeletal muscle and correlates with insulin resistance in obese T2DM patients. Plasma mtDNA may be a useful biomarker for predicting and monitoring insulin resistance in obese patients.

Keywords:

inflammation – blood plasma – mitochondria – Mitochondrial DNA – obesity – Skeletal muscles – oxidative stress

Introduction

Insulin resistance in obese patients and the associated disease cluster of type 2 diabetes mellitus (T2DM), hyperlipidemia, and hypertension are major global health problems. Obesity is associated with chronic, low-grade inflammation, known as metabolic inflammation or metaflammation [1], which is considered a pivotal point in the initiation and progression of insulin resistance and T2DM. Mitochondrial dysfunction induced by oxidative stress contributes to obesity-related insulin resistance [24], but the relationship between mitochondrial dysfunction and the pathogenesis of insulin resistance is unknown. Damage to mitochondrial DNA (mtDNA) may disrupt transcription of proteins encoded by mtDNA that are essential for energy metabolism, initiate apoptotic cell death, and alter mitochondrial redox signaling [59]. In support of the concept that oxidative mtDNA damage contributes to T2DM, we previously showed that damage to mtDNA increases mitochondrial oxidative stress and insulin resistance in skeletal muscle cell [10,11]. Moreover, in a mouse model of insulin resistance induced by a high-fat diet, we showed that mtDNA damage is associated with mitochondrial dysfunction and increased oxidative stress in skeletal muscle and liver [12]. Fragments of mtDNA known as mtDNA damage-associated molecular patterns (DAMPs) may be intercellular mediators of inflammation [13,14]. Such mtDNA fragments are released into the circulation after injury or sepsis and are believed to propagate damage from the initial site of injury or infection to distant organs [15,16]. Inflammation may be propagated by mtDNA DAMPs via activation of one or more pro-inflammatory nucleic acid receptors, including the toll-like receptor 9 (TLR9), NLRP3 inflammasome, and cyclic guanosine monophosphate–adenosine monophosphate synthase–stimulator of interferon genes (cGAS-STING) [1316].

Since obesity is associated with metainflammation the major goal of the current study was to determine whether obese T2DM patients display elevated contents of plasma mtDNA and whether plasma mtDNA correlates with insulin resistance. Our results comprise the first preliminary evidence in a small group of obese, predominantly women patients, that increased levels of plasma mtDNA fragments correlate with the degree of insulin resistance in obese T2DM patients. Furthermore, obese T2DM patients have significantly increased mtDNA damage and oxidative stress markers in skeletal muscle, which was accompanied with increased systemic inflammation. This study suggests there may be novel therapeutic strategies for reducing insulin resistance and for the design of new biomarkers to measure insulin resistance in humans.

Methods

Subjects

We recruited 10 obese (body mass index >35 kg/m2) T2DM patients who had hemoglobin A1C levels > 6.5% and a diagnosis of T2DM based on fasting plasma glucose level > 126 mg/dL or current treatment with any oral hypoglycemic drug. De-identified obese diabetic patients were participants in an ongoing research project conducted by WOR in the Department of Surgery, University of South Alabama College of Medicine. We recruited 12 volunteer healthy control (HC) subjects without obesity (body mass index < 30 kg/m2) or T2DM from the general community. All subjects were sedentary. All human studies including the source study for recruited T2DM patients were conducted according to the principles of the Declaration of Helsinki and approved by the Institutional Review Board (protocols #10–131, 11–150) of the University of South Alabama. All human subjects gave informed written consent.

Metabolic parameters and muscle biopsy

Each subject had a medical history, physical examination including measurement of blood pressure and waist circumference, and blood sampling for screening laboratory tests. On the day of the blood sampling and muscle biopsy, subjects reported to the laboratory after an overnight fast (12 h). Peripheral blood (16 mL) was collected into two sterile density gradient tubes (Vacutainer with Ficoll-Hypaque solution, Becton Dickinson, Franklin Lakes, New Jersey). Blood was fractionated by centrifugation at 1,500g for 30 min at 21°C with a swinging bucket rotor. The plasma (upper) fraction was carefully transferred into 15 ml conical tube, frozen immediately, and stored at -80°C until analyzed. Plasma glucose concentration was determined by the glucose oxidase reaction (Glucose Oxidase Analyzer, Beckman, Fullerton, California), and plasma insulin concentration was measured by radioimmunoassay (Coat-A-Count, Diagnostic Products Corp., Los Angeles, California). Homeostasis model assessment for insulin resistance index (HOMA-IR) was calculated using the formula: HOMA-IR = [glucose (nmol/L)X insulin (μU/mL)/22.5], using fasting values. Hemoglobin A1C percentage was measured using a standard kit. Tumor necrosis factor alpha (TNF-α) concentration (Human TNF alpha PicoKine ELISA Kit, RayBiotech, Norcross, Georgia) and lipid profiles were measured using commercially available methods (VAP Cholesterol Test, Atherotech Diagnostics Lab, Birmingham, Alabama).

Samples of skeletal muscle (100–150 mg) were obtained from the vastus lateralis by percutaneous needle biopsy under sterile conditions with local anesthesia (lidocaine, 1%). The muscle samples were immediately dissected free of any adipose and connective tissue under low-magnification microscopy. Muscle samples were snap frozen in liquid nitrogen for DNA and protein analysis.

Analysis of plasma mtDNA

One T2DM patient and one volunteer from HC were excluded from the analysis of plasma mtDNA since the plasma sample was missing or it was not a sufficient amount of plasma for these patients, accordingly. Thus, we isolated plasma DNA fractions from 9 T2DM patients and 11 volunteers from the HC group (presented in Table 1) and performed quantitative real-time polymerase chain reaction (qRT-PCR) as described previously [17], with minor modifications. Plasma DNA was isolated using a kit (DNeasy, Qiagen, Hilden, Germany) from aliquots of plasma samples (200 μL) that had been frozen at -80°C. DNA was eluted with deionized water (100 μL), and equal amounts of eluant (5 μL) were used for qRT-PCR (VeriQuest Fast SYBR Green qPCR Kit, Affymetrix, Santa Clara, California), according to the manufacturer’s protocol. All primers were designed by Beacon program (Beacon Designer, Premier Biosoft, Palo Alto, California) to amplify 100–200 bp sequences within the specific sequences of the experimental mitochondrial genes. MtDNA sequences quantified included the displacement loop (D-loop) transcriptional regulatory region, which is the transcriptional regulatory of the genome and is particularly prone to oxidative injury [18] as well as regions encoding cytochrome C oxidase subunit I (Cox1), and NADH dehydrogenase subunit 4 (ND4). ND4 lie within the mtDNA “common deletion” sequence, a 4,977-bp region of the mitochondrial genome whose deletion is frequently associated with various pathologies [19], whereas Cox1 and D-loop flank either side of the deleted sequence. The abundance of nuclear DNA (nDNA) in each sample was quantified using sequences of the housekeeping gene 18S rRNA. Sequences of primers are provided in Table 1. The qRT-PCR results were normalized by the mean value of 1 HC sample set to 1 and presented as arbitrary units (AU).

Tab. 1. Primers for qPCR detection of plasma mtDNA and nDNA sequences.
Primers for qPCR detection of plasma mtDNA and nDNA sequences.

Mitochondrial DNA damage analysis

Damage to mtDNA was assessed as described previously [12], with minor modifications. Skeletal muscle was homogenized in liquid nitrogen, and total DNA was isolated with a kit (DNeasy, Qiagen). DNA was digested with BamHI and quantified. Quantitative alkaline Southern blot was performed to evaluate changes in the density of mtDNA lesions using human mtDNA specific probe (cytochrome c oxidase, subunit I) as previously described [7]. Hybridization images were scanned and band intensities were normalized by the mean value of 1 HC sample set to 1 and presented as AU.

Protein isolation, Western blot, and oxidative protein carbonylation

For protein isolation from total cellular fractions, frozen skeletal muscle were homogenized in liquid nitrogen and incubated in the lysis buffer (Cell Lysis Buffer, Cell Signaling, Beverly, Massachusetts) supplemented with 0.1 mg PMSF and a 1:100 dilution of protease and phosphatase inhibitor cocktails (Sigma, St. Louis, Missouri). Homogenization was repeated, samples were centrifuged for 10 min at 14 000 g, and the supernatants were used for Western blot as described previously [1012] with the antibodies against PTEN-induced putative kinase 1 (PINK1) (Abcam, Cambridge, Massachusetts) and actin (Sigma, St. Louis, MO). The complexes that formed were detected with horseradish peroxidase conjugated anti-mouse or anti-rabbit IgG antibodies (Promega, Madison, Wisconsin) using chemiluminescent reagents (SuperSignal, Pierce, Rockford, Illinois). An oxidative protein carbonylation assay was performed as described previously [12]. Where indicated, the resultant band images were scanned and analyzed (Fujifilm Image Gauge Version 2.2 software, Fuji, Tokyo, Japan). For protein carbonylation, summarized band intensities for 2 major bands (~50 kDa and ~40 kDa) were normalized by the mean value of 1 HC sample set to 1 and presented as AU. For PINK1 protein content, data were normalized to the density of actin bands and presented as AU.

Statistical analysis

Data are reported as mean ± standard error of the mean (SEM) and analyzed using statistical software (GraphPad Prism version 7.02 for Windows, GraphPad Software, La Jolla, California). For parametric analysis, differences between groups were assessed using unpaired 2-tailed t test. For indicated sets of data, groups were compared using nonparametric analysis (Mann-Whitney test). Correlation analysis (Spearman test) was used to evaluate the relations between mtDNA fragments in plasma and insulin resistance (HOMA-IR). Statistical significance was defined by P <0.05.

Results

Metabolic parameters

In contrast to HC subjects, the T2DM patients were markedly insulin resistant (high HOMA-IR index) and had markedly increased fasting plasma glucose, hemoglobin A1C, insulin, and triglyceride levels (Table 2).

Tab. 2. Clinical and metabolic data of patients*.
Clinical and metabolic data of patients<em class="ref">*</em>.

Plasma mtDNA fragments and systemic inflammation are increased in obese T2DM patients

First, plasma levels of the Cox1, ND4, and D-loop fragments from mtDNA regions were significantly greater in obese T2DM patients compared to HC subjects (Table 3). This suggested selective release of mtDNA and not cell death because nDNA levels in plasma (18S rRNA) were similar between both groups. There was a significant positive correlation between levels of Cox1, ND4, and D-loop plasma mtDNA sequences and insulin resistance quantified by HOMA-IR, despite the high variability in T2DM samples (Fig 1A and 1B). The strongest correlation was observed for the ND4 sequence (Fig 1A). Importantly, plasma TNF-α levels were significantly greater in obese T2DM patients than HC subjects (Fig 2).

Tab. 3. Levels of plasma DNA fragments in patients*.
Levels of plasma DNA fragments in patients<em class="ref">*</em>.
Relationship between plasma mtDNA and HOMA-IR.
Fig. 1. Relationship between plasma mtDNA and HOMA-IR.
Significant positive correlation was observed between plasma levels of ND4 (A), D-loop (B) and Cox1 (C) sequences and HOMA-IR (n = 9–11 subjects per group).
Increased <i>s</i>ystemic inflammation in obese T2DM patients.
Fig. 2. Increased systemic inflammation in obese T2DM patients.
Level of plasma tumor necrosis factor alpha (TNF-α) was significantly greater in obese T2DM patients than HC (mean ± SEM; n = 5–8 subjects per group). *P <0.05, Mann-Whitney U-test.

Mitochondrial DNA damage, protein carbonylation, and PINK1 expression

Next, we showed that in skeletal muscle, T2DM patients had significantly greater mtDNA damage (Fig 3A and 3B) and protein carbonylation level (Fig 3C) than HC subjects. Since inhibition of mitophagy is a plausible mechanism driving mtDNA fragments elevation in the circulation (vide infra), we next compared the content of phosphatase and tensin homologue-induced kinase 1 (PINK1), an established marker of mitophagy [20] in skeletal muscle isolated from both groups. Contrary to our expectation, there is a trend toward increase in abundance of PINKI in skeletal muscle biopsies from obese T2DM subjects (P = 0.1508, Mann-Whitney U-test, Fig 4B), suggesting increased mitophagy in skeletal muscle in T2DM.

Mitochondrial DNA damage and oxidative protein carbonylation in obese T2DM patients.
Fig. 3. Mitochondrial DNA damage and oxidative protein carbonylation in obese T2DM patients.
(A) Representatives of Quantitative Alkaline Southern blot showing that mtDNA damage was greater in obese T2DM patients than HC. In T2DM patients, there was diminished intensity in the 16.5-kb major restricted band (arrow), indicating that DNA breaks caused smaller fragments that migrated further down the gel, with fewer residual full-size restriction fragments. Representatives from different films are presented. (B) Quantitative alkaline Southern blot densitometry showing lower hybridization intensity, indicative of greater mtDNA damage, in T2DM than HC subjects (mean ± SEM; n = 4–5 subjects per group). *P <0.05, Mann-Whitney U-test. (C) Left side-representatives of oxidative protein carbonylation in skeletal muscle from T2DM and HC. Representatives from one film are presented. Right side represents the graph after densitometric quantitation of data. The average results ± SEM are shown. n = 5–6 per group, *P < 0.05, Mann-Whitney U-test.
Levels of PINK1 in skeletal muscle in obese T2DM patients.
Fig. 4. Levels of PINK1 in skeletal muscle in obese T2DM patients.
(A) Western blot shows PINK1 and actin bands in 5 T2DM patients and 5 HC subjects. (B) Densitometric quantitation of data normalized to the density of actin bands and presented as AU. The average results ± SEM are shown, (n = 5 per group). P = 0.1508, Mann-Whitney U-test.

Discussion

This is the first observational report to provide evidence that increased levels of mtDNA fragments in plasma may be associated with insulin resistance in obese human subjects. We showed that plasma levels of mtDNA are elevated in a small group of obese predominantly women T2DM patients, and that there is a significant positive correlation between elevated levels of plasma mtDNA fragments and insulin resistance in humans. Furthermore, mtDNA damage and oxidative stress in skeletal muscle, as well as systemic inflammation were increased in obese T2DM patients.

A recent study showed that obesity-induced DNA released from adipocytes stimulated chronic adipose tissue inflammation and insulin resistance [21]. Although this study did not specify the origin (nDNA or mtDNA) of cell-free DNA. Also, previous studies showed that mtDNA is present in the circulation of healthy human subjects and patients with T2DM, but no quantitative analysis was provided for comparison of plasma levels of mtDNA between T2DM and healthy patients [22]. In the present study, we performed quantitative analysis of plasma mtDNA sequences by using qRT-PCR. The previous study also showed the presence of the mt3243 mutation in the circulatory mtDNA samples only from T2DM patients [22]. Further quantitative studies are justified to evaluate whether there may be a preference for mutated mtDNA sequences entering the circulation.

While a complete understanding of obesity and its metabolic consequences has yet to be achieved, increased oxidative stress in skeletal muscle has recently been proposed as a unifying mechanism promoting mitochondrial dysfunction, lipid accumulation, and insulin resistance [24]. Complementing this concept, it has also been suggested that mitochondrial superoxide production is a central component of insulin resistance, including high fat diet-induced insulin resistance in skeletal muscle [4]. Although limited information is available about the molecular triggers for these events, the present results suggest that damage to mtDNA may be an important trigger for insulin resistance, given that mtDNA is more sensitive to damage than nDNA [23]. In addition, we showed that increased mtDNA damage was associated with increased protein carbonylation, a marker for oxidative stress.

Mitochondrial DNA is highly specialized and encodes for proteins essential for energy metabolism. Previously, it had been demonstrated that depletion of mtDNA caused impaired glucose utilization and insulin resistance in skeletal muscle cells, suggesting a crucial role for mtDNA in the development of insulin resistance [24]. In agreement with the notion of a destructive role for reactive oxygen species (ROS) on mtDNA in insulin resistance and T2DM is the finding that there are increased mtDNA mutations and reduced mtDNA content in skeletal muscle from T2DM patients [25, 26], consistent with the hypothesis that ROS may degrade mtDNA in patients who have insulin resistance and T2DM. Additionally, a recent study showed that intracellular oxidative stress increases in skeletal muscle and is associated with an increase in oxidation of mtDNA and a decrease in cytochrome b transcription in a rat model of T2DM [27]. Oxidized mtDNA might not transcribe enough cytochrome b that impairs the function of complex III of the electron transport chain leading to an increase in ROS production in a vicious cycle [27]. Altogether this vicious cycle of ROS generation might be involved in the development of insulin resistance in skeletal muscle and further T2DM [27]. In agreement with this study [27], our results also show that obese T2DM patients have significantly increased mtDNA damage in skeletal muscle, providing observational evidence linking mtDNA damage to insulin resistance.

Numerous studies reported an increase in circulating mtDNA fragments in various human diseases, aging and in conditions with acute tissue injury, such as trauma, myocardial infarction, sepsis, and intensive care unit [13, 14,17, 2833]. Interestingly, recent work showed an increase in circulating mtDNA fragments in coronary heart disease patients without T2DM compared to healthy patients and a consistent increase of circulating mtDNA fragments in coronary heart disease patients with T2DM compared with those without the diseases [34]. The present results extend these findings, since we showed elevated levels of mtDNA sequences in the circulation of obese T2DM patients compared to healthy nondiabetic subjects without obesity.

The origin of plasma mtDNA and mechanisms involved in the increase in plasma mtDNA abundance are unknown. Possible mechanisms of accumulation of mtDNA in plasma may include the release of mtDNA from cells of varied origin because of depletion of mitochondrial fission or mitochondrial permeability pores [13], cell death in necrosis or apoptosis, or mtDNA release without degradation during autophagy [29]. In addition to passive release of mtDNA from the cells, some cells deliver mtDNA into circulation actively. Thus, mtDNA in plasma may be associated with a different kinds of subcellular or molecular particles, including mitochondria derived vesicles, exosomes and microparticles [35]. Boudreau et al. showed that activated platelets secrete functional intact mitochondria into the extracellular space as free organelles or incorporated into microparticles [36]. They showed that released mitochondria could be hydrolyzed by phospholipase A2-IIA-releasing mtDNA, thus promoting leukocytes activation [36]. Also, neutrophils are spontaneously releasing mtDNA in the circulation as a part of neutrophil extracellular traps in absence of cell death or membrane disruption [37]. Moreover, a recent study demonstrated that red blood cells homeostatically bind and scavenge mtDNA and thus prevent lung injury [38]. Additional studies are needed to further characterize the origins and size of disease-related forms of circulating mtDNA fragments in plasma of obese T2DM patients.

Regarding our study, most likely inhibition of autophagy was not involved in the release of mtDNA, because the level of PINK1 tended to increase in T2DM patients, rather indicating increased mitophagy in skeletal muscle of T2DM patients. Since mtDNA is more sensitive to oxidative stress than nDNA [23], it is possible that formation of plasma mtDNA fragments in obese T2DM patients may be triggered by molecular fragmentation induced by oxidative stress, followed by their release into the circulation [39]. In this regard it is interesting, that the plasma mtDNA fragments within the ND4 sequence have the strongest correlation with insulin resistance; in addition, ND4-mtDNA showed the highest level in plasma from obese T2DM patients. The ND4 gene is located within the mitochondrial “common deletion” which is prone to oxidation and is associated with a variety of diseases [19]. The second strongest correlation was shown for D-Loop transcriptional regulatory region, which is also specifically prone to oxidation [18]. The lowest correlation was observed for Cox1 sequence which flanks the “common deletion” area. Although requiring further investigation, these observations support the idea that abundance of plasma mtDNA in obese T2DM group may represent the increased degradation and release of selective sequences under oxidative stress. Degradation of mtDNA occurs under conditions of increased oxidative stress [40], but the fate of the degraded DNA is poorly understood. In addition, endogenously oxidized mtDNA causes inflammatory responses in vitro and in vivo [41,42]. Mitochondrial ROS and oxidized mtDNA may activate the NLRR3 inflammasome and TLR9 signaling [4143]. Based on the recent findings that NLRP3-associated inflammasome is upregulated in T2DM patients [44] and instigates obesity-induced inflammation in insulin resistance [45, 46], we have started to evaluate whether plasma mtDNA is oxidatively damaged in T2DM patients. Future studies will be required to define the cascade of events that increase plasma mtDNA and the signaling pathways that are involved in the obesity-related inflammatory responses to plasma mtDNA.

Limitations of the present study include the sex difference between the groups of T2DM and HC subjects (Table 1), and we cannot exclude the effect of sex on the observed results. In addition, a limited number of subjects were included, and we did not evaluate non obese T2DM patients and nondiabetic obese patients. Future studies with larger groups of age-, sex-, and comorbidity-matched individuals may resolve these uncertainties in the present study.

The results of the present study justify the evaluation of plasma mtDNA as a potential cost-effective biomarker for detection of insulin resistance in patients. Insulin resistance may be subtle in the early stages, and diagnosis may be delayed until symptoms are evident and end organ damage has occurred. Current methods to test insulin sensitivity in humans include the euglycemic hyperinsulinemic glucose clamp, and measurement of steady-state plasma glucose are direct, but time and resource-intensive approaches. Alternative surrogate approaches, including calculations based on fasting plasma insulin and glucose levels, or analyses of glucose and insulin levels after intravenous or oral glucose administration; are useful but provide only modest correlation with the primary analyses [47, 48]. It is important to note, that determination of mtDNA abundance in plasma is less costly, invasive and laborious than above mentioned direct assessments of insulin resistance. Further studies may determine whether the levels of circulating, cell-free mtDNA might be a proxy for insulin resistance severity and a cost-effective, early, and reliable marker for the prognosis and diagnosis of insulin resistance, and whether pharmacologic strategies to lower the circulatory levels of mtDNA may prevent insulin resistance and metabolic syndrome.

Supporting information

S1 Dataset [xls]
Excel sheet of dataset on which the conclusions of this manuscript were made.


Zdroje

1. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol 2011; 29:415–445. doi: 10.1146/annurev-immunol-031210-101322 21219177

2. Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA, Morio B, et al. Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin-resistant mice. J Clin Invest 2008;118(2):789–800. 18188455

3. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J Clin Invest 2009;119(3):573–81. doi: 10.1172/JCI37048 19188683

4. Hoehn KL, Salmon AB, Hohnen-Behrens C, Turner N, Hoy AJ, Maghzal GJ, et al. Insulin resistance is a cellular antioxidant defense mechanism. Proc Natl Acad Sci U S A 2009;106(42):17787–92. doi: 10.1073/pnas.0902380106 19805130

5. Rachek LI, Grishko VI, Musiyenko SI, Kelley MR, LeDoux SP, Wilson GL. Conditional targeting of the DNA repair enzyme hOGG1 into mitochondria. J Biol Chem 2002;277:44932–44937. doi: 10.1074/jbc.M208770200 12244119

6. Rachek LI, Thornley NP, Grishko VI, LeDoux SP, Wilson GL. Protection of INS-1 cells from free fatty acid-induced apoptosis by targeting hOGG1 to mitochondria. Diabetes 2006; 55:1022–1028. doi: 10.2337/diabetes.55.04.06.db05-0865 16567524

7. Rachek LI, Yuzefovych LV, Ledoux SP, Julie NL, Wilson GL. Troglitazone, but not rosiglitazone, damages mitochondrial DNA and induces mitochondrial dysfunction and cell death in human hepatocytes. Toxicol Appl Pharmacol 2009; 240:348–354. doi: 10.1016/j.taap.2009.07.021 19632256

8. Ruchko M, Gorodnya O, LeDoux SP, Alexeyev MF, Al-Mehdi AB, Gillespie MN. Mitochondrial DNA damage triggers mitochondrial dysfunction and apoptosis in oxidant-challenged lung endothelial cells. Am J Physiol Lung Cell Mol Physiol 2005; 288:L530–L535. doi: 10.1152/ajplung.00255.2004 15563690

9. Ruchko MV, Gorodnya OM, Zuleta A, Pastukh VM, Gillespie MN. The DNA glycosylase Ogg1 defends against oxidant-induced mtDNA damage and apoptosis in pulmonary artery endothelial cells. Free Radic Biol Med 2011; 50:1107–1113. doi: 10.1016/j.freeradbiomed.2010.10.692 20969951

10. Yuzefovych LV, Solodushko VA, Wilson GL, Rachek LI. Protection from palmitate-induced mitochondrial DNA damage prevents from mitochondrial oxidative stress, mitochondrial dysfunction, apoptosis, and impaired insulin signaling in rat L6 skeletal muscle cells. Endocrinology 2012; 153:92–100. doi: 10.1210/en.2011-1442 22128025

11. Yuzefovych LV, Schuler AM, Chen J, Alvarez DF, Eide L, Ledoux SP, et al. Alteration of mitochondrial function and insulin sensitivity in primary mouse skeletal muscle cells isolated from transgenic and knockout mice: role of OGG1. Endocrinology 2013; 154:2640–2649. doi: 10.1210/en.2013-1076 23748360

12. Yuzefovych LV, Musiyenko SI, Wilson GL, Rachek LI. Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PLoS One 2013; 8:e54059. doi: 10.1371/journal.pone.0054059 23342074

13. West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol 2017; 17:363–375. doi: 10.1038/nri.2017.21 28393922

14. Boyapati RK, Tamborska A, Dorward DA, Ho GT. Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases. F1000Res 2017; 6:169. doi: 10.12688/f1000research.10397.1 28299196

15. Manfredi AA, Rovere-Querini P. The mitochondrion–a Trojan horse that kicks off inflammation? N Engl J Med 2010; 362:2132–2134. doi: 10.1056/NEJMcibr1003521 20519686

16. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011; 469:221–225. doi: 10.1038/nature09663 21124315

17. Simmons JD, Lee YL, Mulekar S, Kuck JL, Brevard SB, Gonzalez RP, et al. Elevated levels of plasma mitochondrial DNA DAMPs are linked to clinical outcome in severely injured human subjects. Ann Surg 2013; 258(4):591–6; discussion 596–8. doi: 10.1097/SLA.0b013e3182a4ea46 23979273

18. Rothfuss O, Gasser T, Patenge N. Analysis of differential DNA damage in the mitochondrial genome employing a semi-long run real-time PCR approach. Nucleic Acids Res 2010; 38: e24. doi: 10.1093/nar/gkp1082 19966269

19. Meissner C, Bruse P, Mohamed SA, Schulz A, Warnk H, Storm T, et al. The 4977 bp deletion of mitochondrial DNA in human skeletal muscle, heart and different areas of the brain: a useful biomarker or more? Exp Gerontol 2008; 43(7):645–52. doi: 10.1016/j.exger.2008.03.004 18439778

20. Mitsuhashi S, Hatakeyama H, Karahashi M, Koumura T, Nonaka I, Hayashi YK, et al. Muscle choline kinase beta defect causes mitochondrial dysfunction and increased mitophagy. Hum Mol Genet 2011; 20(19):3841–51. doi: 10.1093/hmg/ddr305 21750112

21. Nishimoto S, Fukuda D, Higashikuni Y, Tanaka K, Hirata Y, Murata C, et al. Obesity-induced DNA released from adipocytes stimulates chronic adipose tissue inflammation and insulin resistance. Sci Adv 2016; 2(3):e1501332. doi: 10.1126/sciadv.1501332 27051864

22. Cormio A, Milella F, Marra M, Pala M, Lezza AM, Bonfigli AR, et al. Variations at the H-strand replication origins of mitochondrial DNA and mitochondrial DNA content in the blood of type 2 diabetes patients. Biochim Biophys Acta 2009;1787(5):547–52. doi: 10.1016/j.bbabio.2009.01.008 19344660

23. 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 U S A 1997; 94:514–519. doi: 10.1073/pnas.94.2.514 9012815

24. Park SY, Choi GH, Choi HI, Ryu J, Jung CY, Lee W. Depletion of mitochondrial DNA causes impaired glucose utilization and insulin resistance in L6 GLUT4myc myocytes. J Biol Chem 2005; 280(11):9855–64. doi: 10.1074/jbc.M409399200 15764607

25. Liang P, Hughes V, Fukagawa NK. Increased prevalence of mitochondrial DNA deletions in skeletal muscle of older individuals with impaired glucose tolerance: possible marker of glycemic stress. Diabetes 1997; 46:920–923. doi: 10.2337/diab.46.5.920 9133566

26. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, Kelley DE. Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes 2005;54(1):8–14. doi: 10.2337/diabetes.54.1.8 15616005

27. Dos Santos JM, de Oliveira DS, Moreli ML, Benite-Ribeiro SA. The role of mitochondrial DNA damage at skeletal muscle oxidative stress on the development of type 2 diabetes. Mol Cell Biochem 2018; 449:251–255. doi: 10.1007/s11010-018-3361-5 29679277

28. Grazioli S, Pugin J. Mitochondrial Damage-Associated Molecular Patterns: From Inflammatory Signaling to Human Diseases. Front Immunol 2018; 9:832. doi: 10.3389/fimmu.2018.00832 29780380

29. Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai T, et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature; 2012; 485(7397):251–5. doi: 10.1038/nature10992 22535248

30. Pinti M, Cevenini E, Nasi M, De Biasi S, Salvioli S, Monti D, et al. Circulating mitochondrial DNA increases with age and is a familiar trait: Implications for “inflamm-aging”. Eur J Immunol 2014; 44(5):1552–62. doi: 10.1002/eji.201343921 24470107

31. Podlesniy P, Figueiro-Silva J, Llado A, Antonell A, Sanchez-Valle R, Alcolea D, et al. Locerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease. Ann Neurol 2013; 74(5):655–68. doi: 10.1002/ana.23955 23794434

32. Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med 2016; 22:146–53. doi: 10.1038/nm.4027 26779811

33. Boyapati RK, Dorward DA, Tamborska A, Kalla R, Ventham NT, Doherty MK. Mitochondrial DNA Is a Pro-Inflammatory Damage-Associated Molecular Pattern Released During Active IBD. Inflamm Bowel Dis 2018; 24:2113–2122. doi: 10.1093/ibd/izy095 29718255

34. Liu J, Zou Y, Tang Y, Xi M, Xie L, Zhang Q, et al. Circulating cell-free mitochondrial deoxyribonucleic acid is increased in coronary heart disease patients with diabetes mellitus. J Diabetes Investig 2016;7(1):109–14. doi: 10.1111/jdi.12366 26816608

35. Grazioli S, Pugin J. Mitochondrial Damage-Associated Molecular Patterns: From Inflammatory Signaling to Human Diseases 2018; Front Immunol 9:832. doi: 10.3389/fimmu.2018.00832 29780380

36. Boudreau LH, Duchez AC, Cloutier N, Soulet D, Martin N, Bollinger J, et al. Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. Blood 2014; 124:2173–83. doi: 10.1182/blood-2014-05-573543 25082876

37. Caielli S, Athale S, Domic B, Murat E, Chandra M, Banchereau R, et al. Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus. J Exp Med 2016; 213:697–713. doi: 10.1084/jem.20151876 27091841

38. Hotz MJ, Qing D, Shashaty MGS, Zhang P, Faust H, Sondheimer N. Red Blood Cells Homeostatically Bind Mitochondrial DNA through TLR9 to Maintain Quiescence and to Prevent Lung Injury. Am J Respir Crit Care Med 2017; 197:470–480.

39. Kuck JL, Obiako BO, Gorodnya OM, Pastukh VM, Kua J, Simmons JD, et al. Mitochondrial DNA damage-associated molecular patterns mediate a feed-forward cycle of bacteria-induced vascular injury in perfused rat lungs. Am J Physiol Lung Cell Mol Physiol; 2015; 308(10):L1078–85. doi: 10.1152/ajplung.00015.2015 25795724

40. Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, Alexeyev MF. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res 2009; 37:2539–2548. doi: 10.1093/nar/gkp100 19264794

41. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012;36(3):401–14. doi: 10.1016/j.immuni.2012.01.009 22342844

42. Collins LV, Hajizadeh S, Holme E, Jonsson IM, Tarkowski A. Endogenously oxidized mitochondrial DNA induces in vivo and in vitro inflammatory responses. J Leukoc Biol 2004;75(6):995–1000. doi: 10.1189/jlb.0703328 14982943

43. Garcia-Martinez I, Santoro N, Chen Y, Hoque R, Ouyang X, Caprio S, et al. Hepatocyte mitochondrial DNA drives nonalcoholic steatohepatitis by activation of TLR9. J Clin Invest 2016; 126(3):859–64. doi: 10.1172/JCI83885 26808498

44. Lee HM, Kim JJ, Kim HJ, Shong M, Ku BJ, Jo EK. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes 2013;62(1):194–204. doi: 10.2337/db12-0420 23086037

45. Stienstra R, van Diepen JA, Tack CJ, Zaki MH, van de Veerdonk FL, Perera D, et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc Natl Acad Sci U S A 2011; 108(37):15324–9. doi: 10.1073/pnas.1100255108 21876127

46. Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 2011; 17:179–188. doi: 10.1038/nm.2279 21217695

47. Bloomgarden ZT Measures of insulin sensitivity. Clin Lab Med 2006; 26:611–633. doi: 10.1016/j.cll.2006.06.007 16938587

48. Muniyappa R, Lee S, Chen H, Quon MJ. Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage. Am J Physiol Endocrinol Metab 2008; 294:E15–26 doi: 10.1152/ajpendo.00645.2007 17957034


Článek vyšel v časopise

PLOS One


2019 Číslo 10
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#