Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 43(3); 2019 > Article
Sulwon Lecture 2018
Pathophysiology Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation
Chang-Yun Woo1orcid, Jung Eun Jang2, Seung Eun Lee3, Eun Hee Koh1, Ki-Up Lee1orcid
Diabetes & Metabolism Journal 2019;43(3):247-256.
DOI: https://doi.org/10.4093/dmj.2018.0221
Published online: March 27, 2019
  • 8,517 Views
  • 256 Download
  • 69 Web of Science
  • 69 Crossref
  • 74 Scopus

1Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea.

2Department of Internal Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea.

3Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Korea.

Corresponding author: Ki-Up Lee. Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. kulee@amc.seoul.kr
• Received: October 31, 2018   • Accepted: January 19, 2019

Copyright © 2019 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Adipose tissue inflammation is considered a major contributing factor in the development of obesity-associated insulin resistance and cardiovascular diseases. However, the cause of adipose tissue inflammation is presently unclear. The role of mitochondria in white adipocytes has long been neglected because of their low abundance. However, recent evidence suggests that mitochondria are essential for maintaining metabolic homeostasis in white adipocytes. In a series of recent studies, we found that mitochondrial function in white adipocytes is essential to the synthesis of adiponectin, which is the most abundant adipokine synthesized from adipocytes, with many favorable effects on metabolism, including improvement of insulin sensitivity and reduction of atherosclerotic processes and systemic inflammation. From these results, we propose a new hypothesis that mitochondrial dysfunction in adipocytes is a primary cause of adipose tissue inflammation and compared this hypothesis with a prevailing concept that “adipose tissue hypoxia” may underlie adipose tissue dysfunction in obesity. Recent studies have emphasized the role of the mitochondrial quality control mechanism in maintaining mitochondrial function. Future studies are warranted to test whether an inadequate mitochondrial quality control mechanism is responsible for mitochondrial dysfunction in adipocytes and adipose tissue inflammation.
The Sulwon Award for Scientific Achievement is the Korean Diabetes Association's highest scientific award and honors an individual who has excellently contributed to the progress in the field of diabetes and metabolism. The Sulwon Award is named after an emeritus professor, Eung Jin Kim, who founded Korean Diabetes Association. Prof. Ki-Up Lee received the 10th Sulwon Award at 31st Spring Congress of Korean Diabetes Association, May 3 to 5, 2018 at Gwangju, Korea.
Low-grade inflammation in adipose tissue is considered a major contributing factor in the development of obesity-associated insulin resistance and cardiovascular disease [1]. Despite the well-established role in causing insulin resistance, the cause of adipose tissue inflammation is presently unsettled. High-fat diet (HFD) feeding was found to change gut microbiota and gut barrier function to initiate endotoxemia and insulin resistance in rodents [23]. From this, it has been suggested that lipopolysaccharide-induced activation of immune cells may be the primary lesion of adipose tissue inflammation and insulin resistance. Apart from this extrinsic immune system activation, however, there is a possibility that intrinsic changes in adipocytes are responsible for cell death, which triggers activation of inflammatory responses in adipose tissue. Thus, more than 90% of macrophages infiltrating the adipose tissue of obese animals and humans are arranged around dead adipocytes, forming characteristic crown-like structures [45].
Mitochondria generate cellular energy in the form of adenosine triphosphate through glucose and lipid metabolism, and produce many biosynthetic intermediates [67]. Conversely, mitochondrial dysfunction leads to oxidative stress, cell death, inflammation, and metabolic dysfunction [89]. Mitochondria are abundant in skeletal muscle, heart, brown adipocytes, and the liver. On the other hand, the role of mitochondria in white adipocytes has long been neglected because of their low abundance. However, recent evidence suggests that mitochondria are essential for maintaining metabolic homeostasis in white adipocytes [101112]. During the past decade, we investigated the roles of mitochondrial function in white adipocytes and found that mitochondrial function is essential for adiponectin synthesis [131415]. In this short review article, we proposed our new hypothesis that mitochondrial dysfunction in adipocytes may be a primary cause of adipose tissue inflammation and compared this with a prevailing concept that “adipose tissue hypoxia” may underlie adipose tissue dysfunction in obesity [161718].
It is now clear that the adipocyte is not a pure energy store but rather an endocrine gland playing an essential role in energy metabolism. Adipocytes secrete bioactive peptides or proteins, collectively named “adipokines.” Adiponectin is the most abundant adipokine synthesized from adipocytes that has many favorable effects on metabolism, including improvement of insulin sensitivity and reduction of atherosclerotic processes and systemic inflammation [19]. Interestingly, unlike other adipokines, plasma adiponectin levels are paradoxically lower in obese individuals than normal-weight individuals [20]. However, the cause of this paradox has remained unclear.
Adipocytes undergo two stages of maturation: differentiation and hypertrophy [21]. During the early stage of maturation (differentiation), adipocytes have higher levels of metabolic activities and adiponectin secretion, whereas hypertrophic cells lose most of their functional activities and adiponectin secretion [21]. Mitochondrial biogenesis is increased during adipocyte differentiation [22]. Therefore, we conducted a series of studies with the assumption that mitochondrial function is linked to adiponectin synthesis.
Peroxisome proliferator-activated receptor γ (PPAR-γ) agonist (thiazolidinediones [TZDs]) increases insulin sensitivity and plasma adiponectin levels [2324]. Interestingly, this action of TZDs was associated with an increase in the mitochondrial content in adipocytes [1325]. Nuclear respiratory factor 1 (NRF-1) is a key transcription factor that regulates mitochondrial biogenesis [26]. NRF-1 activates mitochondrial gene expression by up-regulating mitochondrial transcription factor A (mtTFA), which is essential for the replication and transcription of mitochondrial DNA (mtDNA). TZDs increased the expression levels of NRF-1 and mtTFA in 3T3L1 adipocytes, and siRNA against mtTFA reversed the TZD-induced increase in adiponectin synthesis and mtDNA content [13]. Adenovirus-mediated overexpression of NRF-1 in cultured 3T3L1 adipocytes increased mtTFA expression and mtDNA content, and these changes were accompanied by increased levels of adiponectin synthesis. Impaired mitochondrial function increased endoplasmic reticulum (ER) stress. Also, agents causing mitochondrial dysfunction or ER stress reduced adiponectin transcription via activation of c-Jun NH2-terminal kinase (JNK) and consequent induction of activating transcription factor 3 (ATF3). This study was the first to show that mitochondrial function is linked to adiponectin synthesis in adipocytes [13].
Among the known nitric oxide (NO) synthases, endothelial NO synthase (eNOS) plays an important role in the regulation of vascular tone and blood pressure [2728]. However, eNOS is also essential for mitochondrial biogenesis [29]. In our study, eNOS inactivation decreased adiponectin secretion in cultured adipocytes, and plasma adiponectin level and mitochondrial biogenesis in white adipose tissue were decreased in adult eNOS knockout (K/O) mice. Moreover, NO supply increased adiponectin synthesis and mitochondrial biogenesis in cultured adipocytes and eNOS K/O mice. These results suggested that proper function of eNOS in adipocytes is necessary for mitochondrial biogenesis and adiponectin synthesis [14].
Cushing's syndrome is a prototype of metabolic syndrome, characterized by central obesity, hypertension, insulin resistance, and glucose intolerance. Elevated levels of either endogenous or exogenous glucocorticoids cause this phenomenon. However, circulating cortisol levels are not always increased in subjects with metabolic syndrome, suggesting an intracellular mechanism that links glucocorticoid metabolism and metabolic syndrome [3031]. 11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) converts inactive glucocorticoids into active glucocorticoids [32], and 11β-HSD1 activity is increased in adipose tissues of leptin-resistant Zucker obese rats [33]. Also, adipocyte-specific overexpression of 11β-HSD1 in transgenic mice produced typical features of metabolic syndrome [34]. In our study, 11β-HSD1 mRNA expression was increased, and mitochondrial respiration was decreased during prolonged culture of 3T3L1 adipocytes. Treatment with AZD 6925, a selective 11β-HSD1 inhibitor, increased mitochondrial biogenesis in cultured adipocytes. It also increased plasma adiponectin levels in db/db mice. This study may explain how hypertrophic adipocytes of obese subjects having decreased mitochondrial function produce a lower amount of adiponectin.
Taken together, it is suggested that mitochondrial function is essential for the production of adiponectin in adipocytes.
Consistent with our studies, other groups also reported that mitochondrial dysfunction in adipocytes may cause obesity or insulin resistance. Cellular levels of mitochondrial proteins, mitochondrial DNA contents, and mitochondrial function were decreased in obese (ob/ob) and diabetic (db/db) mice [1325]. More recently, an inverse regulation of inflammation and mitochondrial function was found in adipose tissue of morbidly obese patients [35]. Haplodeficiency of Crif1, a protein required for the intramitochondrial production of mtDNA-encoded oxidative phosphorylation (OXPHOS) subunits, reduced adipose OXPHOS capacity and triggered inflammation and insulin resistance in mice [36]. Mice deficient in mtTFA in adipocytes showed impaired mitochondrial biogenesis in adipose tissue, lipodystrophic syndrome with insulin resistance, hepatosteatosis, and cardiovascular complications [37]. In humans, mtDNA contents, mtDNA-encoded transcripts, and mitochondrial OXPHOS protein levels in adipose tissue were downregulated in the obese compared with the lean co-twins [38]. Pathway analysis indicated downshifting of fatty acid oxidation, ketone body production and breakdown, and the tricarboxylic acid cycle, which was inversely correlated with adiposity, insulin resistance, and inflammatory cytokines [38].
Taken together, these results suggested that mitochondrial dysfunction is found in adipocytes of human and animals with obesity, and that this is related to insulin resistance, adipose tissue inflammation, and/or lipodystrophy.
Increasing evidence suggests that mitochondrial function is a potential central regulator of the aging process [39]. Mitochondrial dysfunction contributes to cellular senescence, chronic inflammation, and the age-dependent decline in stem cell activity [40]. A recent study reported that mitochondrial complex IV dysfunction is pivotal in aging-associated obesity [41]. Plasma adiponectin levels are decreased in apparently healthy elderly men and women [42], and aging is associated with circulating inflammatory markers independent of body mass index [43]. Taken together, these results suggest that mitochondrial dysfunction in adipocytes is related to adipose tissue inflammation in aging as well as obesity or insulin resistance.
Immune cell infiltration is a prominent feature of dysfunctional adipose tissue, and these immune cells include M1 and M2 macrophages, effector and memory T-cells, interleukin 10 producing FoxP3+ T regulatory cells, natural killer and natural killer T (NKT) cells, and granulocytes [44]. Of these various immune cells, adipose tissue macrophages play a central role in the genesis of adipose tissue inflammation [45]. At least two distinct populations of macrophages infiltrate the adipose tissue: proinflammatory (M1) and anti-inflammatory (M2). Among the M1 and M2 macrophage markers, inducible nitric oxide synthase (iNOS) and arginase are considered the most important molecules responsible for M1 and M2 activities, respectively [45]. iNOS metabolizes arginine to NO and citrulline. On the other hand, arginase expressed in M2 macrophages hydrolyzes arginine to ornithine and urea.
Macrophages have been shown to aggregate, forming crown-like structures surrounding necrotic adipocytes in obesity [446]. Therefore, it was proposed that necrosis of adipocytes is a prominent phagocytic stimulus that activates adipose tissue macrophage infiltration [4]. Mitochondria play a central role in cell death [4748]. Thus, mitochondrial dysfunction in adipocytes may trigger cell death in adipocytes to induce adipose tissue inflammation.
Monocyte chemoattractant protein-1 (MCP-1) is produced predominantly by macrophages and endothelial cells and is a potent chemotactic factor for monocytes. However, it is now well recognized that adipocytes are an important source of MCP-1 production [49]. mRNA expression in adipose tissue and the plasma concentration of MCP-1 were increased in obese mice [50]. Recently, mitochondrial dysfunction was shown to increase MCP-1 expression in renal tubular epithelial cells causing inflammation [51]. Thus, there is a possibility that mitochondrial dysfunction in adipocytes causes increased expression of MCP-1 in adipocytes to increase chemotaxis of macrophages into adipose tissue.
Finally, adiponectin is well-known to activate AMP-activated protein kinase (AMPK) in diverse tissues [52], and adiponectin activates AMPK in macrophages [53]. AMPK shifts M1 to M2 macrophage polarization [5455], and adiponectin was shown to prime human monocytes into anti-inflammatory M2 macrophages [56]. Therefore, we propose that failure to produce adiponectin synthesis due to mitochondrial dysfunction in adipocytes may lead to decreased M2 macrophage polarization and increased adipose tissue inflammation (Fig. 1).
As described above, mitochondrial dysfunction in adipocytes may be the primary mechanism that triggers adipose tissue inflammation and insulin resistance. However, proinflammatory cytokines were shown to regulate adipocyte mitochondrial metabolism [57]. We thus examined the other possibility that proinflammatory response of macrophages induces mitochondrial dysfunction in adipocytes.
In addition to inflammation, adipose tissue fibrosis is exacerbated in obese human subjects and HFD-fed rodents. Fibrosis limits the expandability of adipose tissue and contributes to ectopic fat accumulation and the development of insulin resistance [45]. Classically activated (M1) macrophages have increased iNOS expression and contribute to adipose tissue inflammation. We examined the relationship between NO produced by macrophages and adipose tissue fibrosis. The hypoxia-inducible factor 1α (HIF-1α) protein level was increased in adipose tissue of wild-type (WT) mice fed an HFD for a prolonged period but not in iNOS K/O mice. The expression of mitochondrial biogenesis factors was decreased in HFD-fed WT mice but not in iNOS K/O mice. In studies with cultured cells, macrophage-derived NO decreased the expression of mitochondrial biogenesis factors and increased DNA damage and phosphorylated p53 in preadipocytes. By activating p53 signaling, NO suppressed expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), which induced mitochondrial dysfunction and inhibited preadipocyte differentiation to adipocytes. The effects of NO were blocked by rosiglitazone. These findings suggest that NO produced by macrophages induces mitochondrial dysfunction in preadipocytes by activating p53 signaling, which in turn increases HIF-1α protein levels and promotes a profibrogenic response in preadipocytes that results in adipose tissue fibrosis [45].
These results suggest that, in addition to primary mitochondrial dysfunction in adipocytes, there may be additional mechanisms that promote and sustain mitochondrial dysfunction in adipocytes and/or adipose tissue inflammation/fibrosis (Fig. 2).
To ensure adequate function of mitochondria, quality control mechanisms are necessary to eliminate damaged mitochondrial proteins or parts of the mitochondrial network by mitophagy and renew components by adding protein and lipids through biogenesis [58]. When dysfunctional mitochondria are not cleared adequately by mitophagy, this may lead to aging-associated diseases [59].
In mammals, mitophagy is mediated either by the PTEN-induced putative kinase 1 (PINK1)-Parkin signaling pathway or the mitophagic receptors Bnip3 and Nix [60]. Even though the role of mitophagy in adipocytes has not been studied adequately, a recent study found that PINK1-Parkin alleviates metabolic stress induced by obesity in adipose tissue and 3T3-L1 preadipocytes [61].
Then, what would be the clinical implication of these findings? Exercise training has long been known to increase mitochondrial biogenesis in skeletal muscle [62]. In addition, recent studies found that exercise training increases mitochondrial biogenesis in white adipose tissue [6364]. Thus, the beneficial effect of exercise training may, at least in part, be due to its effect on mitochondria in adipose tissue.
TZDs are known to increase mitochondrial biogenesis in adipocytes [65] and are currently used in clinical medicine to treat diabetes mellitus and insulin resistance. There may be many other chemical or natural compounds that improve mitochondrial function [66] in adipocytes through increase of mitochondrial biogenesis and/or mitophagy, but their role in clinical medicine remains to be determined.
Finally, we would like to compare our hypothesis with a prevailing concept that “adipose tissue hypoxia” may underlie adipose tissue dysfunction in obesity. In the year 2007, two groups of researchers proposed that adipose tissue of obese mice is hypoxic, and that local adipose tissue hypoxia dysregulates the production of adiponectin [1667]. Since then, this has been regarded as the main mechanism of insulin resistance by many investigators [6869]. However, we found that mitochondrial dysfunction in preadipocytes of HFD-fed mice induced pseudo-hypoxic induction of HIF-1α protein levels, which led to adipose tissue fibrosis [45].
In fact, cellular response to hypoxia and pseudo-hypoxic response by mitochondrial dysfunction is very similar in that both conditions are characterized by decreased mitochondrial respiration and increased glycolysis. “Adipose tissue hypoxia” theory explains that there is hypoxia in enlarged adipocytes distant from the vasculature [70]. The only method that could determine whether adipose tissue hypoxia is present in adipose tissue of obesity is the measurement of oxygen tension. However, the results are quite controversial. Most studies performed in rodents reported that adipose tissue oxygen tension is decreased in obese animals [166771]. On the other hand, a study performed in humans reported increased adipose tissue oxygen tension despite lower adipose tissue blood flow in obese compared with lean individuals [72]. This was accompanied by insulin resistance, lower adipose tissue capillarization, lower adipose tissue expression of genes encoding proteins involved in mitochondrial biogenesis and function, and higher macrophage infiltration and inflammatory markers.
In this article, we proposed that mitochondrial dysfunction in adipocytes may be a primary cause of adipose tissue inflammation (Fig. 1). Despite widespread belief on adipose tissue hypoxia theory, we hypothesize that mitochondrial dysfunction in adipocytes is the primary cause of insulin resistance and adipose tissue inflammation (Fig. 3). Defective mitochondrial function and decreased fatty acid oxidation in adipocytes would increase triglyceride accumulation to cause adipocyte enlargement and adipose tissue hypoxia. In addition, mitochondrial dysfunction in adipocytes may exert a “pseudohypoxic response” to promote adipose tissue fibrosis, making it difficult to distinguish these two theories from each other. Even though adipose tissue hypoxia has been commonly observed in mouse obesity, human data reported increased adipose tissue oxygen tension [60]. In addition, mitochondrial dysfunction in adipose tissue was found in human obesity [3761], supporting our hypothesis that mitochondrial dysfunction in adipocytes is a primary cause of insulin resistance in obesity. The cause of mitochondrial dysfunction in adipocytes in obesity is presently unknown, but future studies are warranted to test whether a defective mitochondrial quality control mechanism is responsible for mitochondrial dysfunction in hypertrophic adipocytes.
Acknowledgements
This work was supported by Korean Diabetes Association Grant (2018 Sulwon Award for Scientific Achievement).

CONFLICTS OF INTEREST: No potential conflict of interest relevant to this article was reported.

  • 1. Lackey DE, Olefsky JM. Regulation of metabolism by the innate immune system. Nat Rev Endocrinol 2016;12:15-28. ArticlePubMedPDF
  • 2. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, Waget A, Delmee E, Cousin B, Sulpice T, Chamontin B, Ferrieres J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, Burcelin R. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56:1761-1772. ArticlePubMedPDF
  • 3. Luck H, Tsai S, Chung J, Clemente-Casares X, Ghazarian M, Revelo XS, Lei H, Luk CT, Shi SY, Surendra A, Copeland JK, Ahn J, Prescott D, Rasmussen BA, Chng MH, Engleman EG, Girardin SE, Lam TK, Croitoru K, Dunn S, Philpott DJ, Guttman DS, Woo M, Winer S, Winer DA. Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab 2015;21:527-542. ArticlePubMed
  • 4. Murano I, Barbatelli G, Parisani V, Latini C, Muzzonigro G, Castellucci M, Cinti S. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res 2008;49:1562-1568. ArticlePubMed
  • 5. Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, Wang S, Fortier M, Greenberg AS, Obin MS. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 2005;46:2347-2355. ArticlePubMed
  • 6. Kwak SH, Park KS, Lee KU, Lee HK. Mitochondrial metabolism and diabetes. J Diabetes Investig 2010;1:161-169.ArticlePubMedPMC
  • 7. Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell 2012;148:1145-1159. ArticlePubMedPMC
  • 8. Hock MB, Kralli A. Transcriptional control of mitochondrial biogenesis and function. Annu Rev Physiol 2009;71:177-203. ArticlePubMed
  • 9. Kim MJ, Koo YD, Kim M, Lim S, Park YJ, Chung SS, Jang HC, Park KS. Rg3 improves mitochondrial function and the expression of key genes involved in mitochondrial biogenesis in C2C12 myotubes. Diabetes Metab J 2016;40:406-413. ArticlePubMedPMCPDF
  • 10. Kusminski CM, Scherer PE. Mitochondrial dysfunction in white adipose tissue. Trends Endocrinol Metab 2012;23:435-443. ArticlePubMedPMC
  • 11. Flachs P, Rossmeisl M, Kuda O, Kopecky J. Stimulation of mitochondrial oxidative capacity in white fat independent of UCP1: a key to lean phenotype. Biochim Biophys Acta 2013;1831:986-1003. ArticlePubMed
  • 12. Boudina S, Graham TE. Mitochondrial function/dysfunction in white adipose tissue. Exp Physiol 2014;99:1168-1178. ArticlePubMed
  • 13. Koh EH, Park JY, Park HS, Jeon MJ, Ryu JW, Kim M, Kim SY, Kim MS, Kim SW, Park IS, Youn JH, Lee KU. Essential role of mitochondrial function in adiponectin synthesis in adipocytes. Diabetes 2007;56:2973-2981. ArticlePubMedPDF
  • 14. Koh EH, Kim M, Ranjan KC, Kim HS, Park HS, Oh KS, Park IS, Lee WJ, Kim MS, Park JY, Youn JH, Lee KU. eNOS plays a major role in adiponectin synthesis in adipocytes. Am J Physiol Endocrinol Metab 2010;298:E846-E853. ArticlePubMed
  • 15. Koh EH, Kim AR, Kim H, Kim JH, Park HS, Ko MS, Kim MO, Kim HJ, Kim BJ, Yoo HJ, Kim SJ, Oh JS, Woo CY, Jang JE, Leem J, Cho MH, Lee KU. 11β-HSD1 reduces metabolic efficacy and adiponectin synthesis in hypertrophic adipocytes. J Endocrinol 2015;225:147-158. ArticlePubMed
  • 16. Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, Furukawa S, Tochino Y, Komuro R, Matsuda M, Shimomura I. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 2007;56:901-911. ArticlePubMedPDF
  • 17. Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab 2013;18:470-477. ArticlePubMedPMC
  • 18. Trayhurn P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol Rev 2013;93:1-21. ArticlePubMed
  • 19. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest 2006;116:1784-1792. ArticlePubMedPMC
  • 20. Hoffstedt J, Arvidsson E, Sjolin E, Wahlen K, Arner P. Adipose tissue adiponectin production and adiponectin serum concentration in human obesity and insulin resistance. J Clin Endocrinol Metab 2004;89:1391-1396. ArticlePubMedPDF
  • 21. Yu YH, Zhu H. Chronological changes in metabolism and functions of cultured adipocytes: a hypothesis for cell aging in mature adipocytes. Am J Physiol Endocrinol Metab 2004;286:E402-E410. ArticlePubMed
  • 22. Wilson-Fritch L, Burkart A, Bell G, Mendelson K, Leszyk J, Nicoloro S, Czech M, Corvera S. Mitochondrial biogenesis and remodeling during adipogenesis and in response to the insulin sensitizer rosiglitazone. Mol Cell Biol 2003;23:1085-1094. ArticlePubMedPMCPDF
  • 23. Olefsky JM, Saltiel AR. PPAR gamma and the treatment of insulin resistance. Trends Endocrinol Metab 2000;11:362-368. PubMed
  • 24. Maeda N, Takahashi M, Funahashi T, Kihara S, Nishizawa H, Kishida K, Nagaretani H, Matsuda M, Komuro R, Ouchi N, Kuriyama H, Hotta K, Nakamura T, Shimomura I, Matsuzawa Y. PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein. Diabetes 2001;50:2094-2099. PubMed
  • 25. Choo HJ, Kim JH, Kwon OB, Lee CS, Mun JY, Han SS, Yoon YS, Yoon G, Choi KM, Ko YG. Mitochondria are impaired in the adipocytes of type 2 diabetic mice. Diabetologia 2006;49:784-791. ArticlePubMedPDF
  • 26. Villena JA. New insights into PGC-1 coactivators: redefining their role in the regulation of mitochondrial function and beyond. FEBS J 2015;282:647-672. ArticlePubMedPDF
  • 27. Duplain H, Burcelin R, Sartori C, Cook S, Egli M, Lepori M, Vollenweider P, Pedrazzini T, Nicod P, Thorens B, Scherrer U. Insulin resistance, hyperlipidemia, and hypertension in mice lacking endothelial nitric oxide synthase. Circulation 2001;104:342-345. ArticlePubMed
  • 28. Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discov 2015;14:623-641. ArticlePubMedPDF
  • 29. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, Bracale R, Valerio A, Francolini M, Moncada S, Carruba MO. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science 2003;299:896-899. ArticlePubMed
  • 30. Kumari M, Chandola T, Brunner E, Kivimaki M. A nonlinear relationship of generalized and central obesity with diurnal cortisol secretion in the Whitehall II study. J Clin Endocrinol Metab 2010;95:4415-4423. ArticlePubMedPMCPDF
  • 31. Hackett RA, Steptoe A, Kumari M. Association of diurnal patterns in salivary cortisol with type 2 diabetes in the Whitehall II study. J Clin Endocrinol Metab 2014;99:4625-4631. ArticlePubMedPMC
  • 32. Dube S, Norby BJ, Pattan V, Carter RE, Basu A, Basu R. 11β-Hydroxysteroid dehydrogenase types 1 and 2 activity in subcutaneous adipose tissue in humans: implications in obesity and diabetes. J Clin Endocrinol Metab 2015;100:E70-E76. ArticlePubMed
  • 33. Livingstone DE, Kenyon CJ, Walker BR. Mechanisms of dysregulation of 11 beta-hydroxysteroid dehydrogenase type 1 in obese Zucker rats. J Endocrinol 2000;167:533-539. ArticlePubMed
  • 34. Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, Seckl JR, Flier JS. A transgenic model of visceral obesity and the metabolic syndrome. Science 2001;294:2166-2170. ArticlePubMed
  • 35. Qatanani M, Tan Y, Dobrin R, Greenawalt DM, Hu G, Zhao W, Olefsky JM, Sears DD, Kaplan LM, Kemp DM. Inverse regulation of inflammation and mitochondrial function in adipose tissue defines extreme insulin sensitivity in morbidly obese patients. Diabetes 2013;62:855-863. ArticlePubMedPMCPDF
  • 36. Ryu MJ, Kim SJ, Kim YK, Choi MJ, Tadi S, Lee MH, Lee SE, Chung HK, Jung SB, Kim HJ, Jo YS, Kim KS, Lee SH, Kim JM, Kweon GR, Park KC, Lee JU, Kong YY, Lee CH, Chung J, Shong M. Crif1 deficiency reduces adipose OXPHOS capacity and triggers inflammation and insulin resistance in mice. PLoS Genet 2013;9:e1003356. ArticlePubMedPMC
  • 37. Vernochet C, Damilano F, Mourier A, Bezy O, Mori MA, Smyth G, Rosenzweig A, Larsson NG, Kahn CR. Adipose tissue mitochondrial dysfunction triggers a lipodystrophic syndrome with insulin resistance, hepatosteatosis, and cardiovascular complications. FASEB J 2014;28:4408-4419. ArticlePubMedPMCPDF
  • 38. Heinonen S, Buzkova J, Muniandy M, Kaksonen R, Ollikainen M, Ismail K, Hakkarainen A, Lundbom J, Lundbom N, Vuolteenaho K, Moilanen E, Kaprio J, Rissanen A, Suomalainen A, Pietilainen KH. Impaired mitochondrial biogenesis in adipose tissue in acquired obesity. Diabetes 2015;64:3135-3145. ArticlePubMedPDF
  • 39. Jang JY, Blum A, Liu J, Finkel T. The role of mitochondria in aging. J Clin Invest 2018;128:3662-3670. ArticlePubMedPMC
  • 40. Sun N, Youle RJ, Finkel T. The mitochondrial basis of aging. Mol Cell 2016;61:654-666. ArticlePubMedPMC
  • 41. Soro-Arnaiz I, Li QOY, Torres-Capelli M, Melendez-Rodriguez F, Veiga S, Veys K, Sebastian D, Elorza A, Tello D, Hernansanz-Agustin P, Cogliati S, Moreno-Navarrete JM, Balsa E, Fuertes E, Romanos E, Martinez-Ruiz A, Enriquez JA, Fernandez-Real JM, Zorzano A, De Bock K, Aragones J. Role of mitochondrial complex IV in age-dependent obesity. Cell Rep 2016;16:2991-3002. ArticlePubMed
  • 42. Adamczak M, Rzepka E, Chudek J, Wiecek A. Ageing and plasma adiponectin concentration in apparently healthy males and females. Clin Endocrinol (Oxf) 2005;62:114-118. ArticlePubMed
  • 43. Miles EA, Rees D, Banerjee T, Cazzola R, Lewis S, Wood R, Oates R, Tallant A, Cestaro B, Yaqoob P, Wahle KW, Calder PC. Age-related increases in circulating inflammatory markers in men are independent of BMI, blood pressure and blood lipid concentrations. Atherosclerosis 2008;196:298-305. ArticlePubMed
  • 44. Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2017;113:1009-1023. ArticlePubMedPMC
  • 45. Jang JE, Ko MS, Yun JY, Kim MO, Kim JH, Park HS, Kim AR, Kim HJ, Kim BJ, Ahn YE, Oh JS, Lee WJ, Harris RA, Koh EH, Lee KU. Nitric oxide produced by macrophages inhibits adipocyte differentiation and promotes profibrogenic responses in preadipocytes to induce adipose tissue fibrosis. Diabetes 2016;65:2516-2528. ArticlePubMedPDF
  • 46. Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW 2nd, DeFuria J, Jick Z, Greenberg AS, Obin MS. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes 2007;56:2910-2918. ArticlePubMedPDF
  • 47. Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 2012;13:780-788. ArticlePubMedPDF
  • 48. Green DR, Galluzzi L, Kroemer G. Cell biology. Metabolic control of cell death. Science 2014;345:1250256. PubMedPMC
  • 49. Omar A, Chatterjee TK, Tang Y, Hui DY, Weintraub NL. Proinflammatory phenotype of perivascular adipocytes. Arterioscler Thromb Vasc Biol 2014;34:1631-1636. ArticlePubMedPMC
  • 50. Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K, Kasuga M. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest 2006;116:1494-1505. ArticlePubMedPMC
  • 51. Zhao H, Liu YJ, Liu ZR, Tang DD, Chen XW, Chen YH, Zhou RN, Chen SQ, Niu HX. Role of mitochondrial dysfunction in renal fibrosis promoted by hypochlorite-modified albumin in a remnant kidney model and protective effects of antioxidant peptide SS-31. Eur J Pharmacol 2017;804:57-67. ArticlePubMed
  • 52. Long YC, Zierath JR. AMP-activated protein kinase signaling in metabolic regulation. J Clin Invest 2006;116:1776-1783. ArticlePubMedPMC
  • 53. Kim MJ, Kim EH, Pun NT, Chang JH, Kim JA, Jeong JH, Choi DY, Kim SH, Park PH. Globular adiponectin inhibits lipopolysaccharide-primed inflammasomes activation in macrophages via autophagy induction: the critical role of AMPK signaling. Int J Mol Sci 2017;18:E1275. Article
  • 54. Griffiths HR, Gao D, Pararasa C. Redox regulation in metabolic programming and inflammation. Redox Biol 2017;12:50-57. ArticlePubMedPMC
  • 55. Chan KL, Pillon NJ, Sivaloganathan DM, Costford SR, Liu Z, Theret M, Chazaud B, Klip A. Palmitoleate reverses high fat-induced proinflammatory macrophage polarization via AMP-activated protein kinase (AMPK). J Biol Chem 2015;290:16979-16988. ArticlePubMedPMC
  • 56. Lovren F, Pan Y, Quan A, Szmitko PE, Singh KK, Shukla PC, Gupta M, Chan L, Al-Omran M, Teoh H, Verma S. Adiponectin primes human monocytes into alternative anti-inflammatory M2 macrophages. Am J Physiol Heart Circ Physiol 2010;299:H656-H663. ArticlePubMedPMC
  • 57. Hahn WS, Kuzmicic J, Burrill JS, Donoghue MA, Foncea R, Jensen MD, Lavandero S, Arriaga EA, Bernlohr DA. Proinflammatory cytokines differentially regulate adipocyte mitochondrial metabolism, oxidative stress, and dynamics. Am J Physiol Endocrinol Metab 2014;306:E1033-E1045. ArticlePubMedPMC
  • 58. Pickles S, Vigie P, Youle RJ. Mitophagy and quality control mechanisms in mitochondrial maintenance. Curr Biol 2018;28:R170-R185. ArticlePubMedPMC
  • 59. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science 2011;333:1109-1112. ArticlePubMedPMC
  • 60. Novak I. Mitophagy: a complex mechanism of mitochondrial removal. Antioxid Redox Signal 2012;17:794-802. ArticlePubMed
  • 61. Cui C, Chen S, Qiao J, Qing L, Wang L, He T, Wang C, Liu F, Gong L, Chen L, Hou X. PINK1-Parkin alleviates metabolic stress induced by obesity in adipose tissue and in 3T3-L1 preadipocytes. Biochem Biophys Res Commun 2018;498:445-452. ArticlePubMed
  • 62. Heo JW, No MH, Park DH, Kang JH, Seo DY, Han J, Neufer PD, Kwak HB. Effects of exercise on obesity-induced mitochondrial dysfunction in skeletal muscle. Korean J Physiol Pharmacol 2017;21:567-577. ArticlePubMedPMCPDF
  • 63. Trevellin E, Scorzeto M, Olivieri M, Granzotto M, Valerio A, Tedesco L, Fabris R, Serra R, Quarta M, Reggiani C, Nisoli E, Vettor R. Exercise training induces mitochondrial biogenesis and glucose uptake in subcutaneous adipose tissue through eNOS-dependent mechanisms. Diabetes 2014;63:2800-2811. ArticlePubMedPDF
  • 64. Stanford KI, Middelbeek RJ, Goodyear LJ. Exercise effects on white adipose tissue: beiging and metabolic adaptations. Diabetes 2015;64:2361-2368. ArticlePubMedPMCPDF
  • 65. Bogacka I, Xie H, Bray GA, Smith SR. Pioglitazone induces mitochondrial biogenesis in human subcutaneous adipose tissue in vivo. Diabetes 2005;54:1392-1399. ArticlePubMedPDF
  • 66. Teixeira J, Chavarria D, Borges F, Wojtczak L, Wieckowski MR, Karkucinska-Wieckowska A, Oliveira PJ. Dietary polyphenols and mitochondrial function: role in health and disease. Curr Med Chem 2017 5 28 [Epub].Article
  • 67. Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 2007;293:E1118-E1128. ArticlePubMed
  • 68. McLaughlin T, Ackerman SE, Shen L, Engleman E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J Clin Invest 2017;127:5-13. ArticlePubMedPMC
  • 69. Rasouli N. Adipose tissue hypoxia and insulin resistance. J Investig Med 2016;64:830-832.ArticlePubMedPDF
  • 70. Trayhurn P, Wang B, Wood IS. Hypoxia in adipose tissue: a basis for the dysregulation of tissue function in obesity? Br J Nutr 2008;100:227-235. ArticlePubMed
  • 71. Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond) 2008;32:451-463. ArticlePubMedPDF
  • 72. Goossens GH, Bizzarri A, Venteclef N, Essers Y, Cleutjens JP, Konings E, Jocken JW, Cajlakovic M, Ribitsch V, Clement K, Blaak EE. Increased adipose tissue oxygen tension in obese compared with lean men is accompanied by insulin resistance, impaired adipose tissue capillarization, and inflammation. Circulation 2011;124:67-76. ArticlePubMed
Fig. 1

Hypothesis: mitochondrial dysfunction in adipocytes may be a primary cause of adipose tissue inflammation. Hypertrophic adipocytes in obesity or cellular aging are associated with increased expression levels of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), and these cells show reduced mitochondrial respiration and increased glycolysis. Mitochondrial dysfunction may trigger necroptosis in adipocytes or increase monocyte chemoattractant protein-1 (MCP-1) production, which initiates adipose tissue inflammation by attracting macrophages into adipose tissue. In addition, mitochondrial dysfunction in adipocytes decreases synthesis of adiponectin, a well-known AMP-activated protein kinase (AMPK) activator. Reduced AMPK activation in macrophages may increase M1/M2 macrophage polarization. Therefore, mitochondrial dysfunction in adipocytes may increase adipose tissue inflammation with increased M1/M2 polarization through a decrease in adiponectin synthesis.

dmj-43-247-g001.jpg
Fig. 2

Proinflammatory response of macrophages may exacerbate mitochondrial dysfunction in adipocytes. In addition to the primary mitochondrial dysfunction resulting from possible intrinsic defect in mitochondrial quality control in adipocytes, nitric oxide (NO) produced by inducible nitric oxide synthase in activated macrophages may exacerbate mitochondrial dysfunction in preadipocytes. Mitochondrial dysfunction in preadipocytes increases pseudohypoxic response, which leads to a defective differentiation of preadipocytes to mature adipocytes, and adipose tissue fibrosis. PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1α.

dmj-43-247-g002.jpg
Fig. 3

Comparison of our hypothesis and “adipose tissue hypoxia” theory. The hypothesis of “adipose tissue hypoxia” explains that hypoxia occurs in enlarged adipocytes distant from the vasculature and that this local adipose tissue hypoxia decreases the production of adiponectin. On the other hand, we hypothesize that mitochondrial dysfunction in adipocytes is a primary cause of adipocyte enlargement, adipose tissue inflammation, and insulin resistance. Defective mitochondrial function and fatty acid oxidation (FAO) in adipocytes would increase triglyceride accumulation to cause adipocyte enlargement. Further, mitochondrial dysfunction in adipocytes may induce pseudo-hypoxia to increase accumulation of hypoxia-inducible factor 1α (HIF-1α), which promotes adipose tissue inflammation and fibrosis. Even though adipose tissue hypoxia has been commonly observed in mouse obesity, human data reported increased adipose tissue oxygen tension and mitochondrial dysfunction in adipose tissue. TG, triglyceride.

dmj-43-247-g003.jpg

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Prolonged Endurance Exercise Increases Macrophage Content and Mitochondrial Respiration in Adipose Tissue in Trained Men
      Ronni Eg Sahl, Ioanna Patsi, Mikkel Thunestvedt Hansen, Tue Rømer, Jacob Frandsen, Hanne Kruuse Rasmusen, Arthur Ingersen, Steen Seier Poulsen, Flemming Dela, Steen Larsen, Jørn Wulff Helge
      The Journal of Clinical Endocrinology & Metabolism.2024; 109(2): e799.     CrossRef
    • Diabetes Mellitus, Energy Metabolism, and COVID-19
      Caterina Conte, Elisa Cipponeri, Michael Roden
      Endocrine Reviews.2024; 45(2): 281.     CrossRef
    • The Role of Ion-Transporting Proteins in Human Disease
      Yoshinori Marunaka
      International Journal of Molecular Sciences.2024; 25(3): 1726.     CrossRef
    • The Role of Obesity in Type 2 Diabetes Mellitus—An Overview
      Preethi Chandrasekaran, Ralf Weiskirchen
      International Journal of Molecular Sciences.2024; 25(3): 1882.     CrossRef
    • The Metabolic Syndrome, a Human Disease
      Marià Alemany
      International Journal of Molecular Sciences.2024; 25(4): 2251.     CrossRef
    • Inflammation‐mediated metabolic regulation in adipose tissue
      Shujie Xu, Feng Lu, Jianhua Gao, Yi Yuan
      Obesity Reviews.2024;[Epub]     CrossRef
    • Could very low-calorie ketogenic diets turn off low grade inflammation in obesity? Emerging evidence
      Luigi Barrea, Massimiliano Caprio, Mikiko Watanabe, Giuseppe Cammarata, Alessandra Feraco, Giovanna Muscogiuri, Ludovica Verde, Annamaria Colao, Silvia Savastano
      Critical Reviews in Food Science and Nutrition.2023; 63(26): 8320.     CrossRef
    • The emergent role of mitochondrial RNA modifications in metabolic alterations
      Hatim Boughanem, Yvonne Böttcher, João Tomé‐Carneiro, María‐Carmen López de las Hazas, Alberto Dávalos, Akin Cayir, Manuel Macias‐González
      WIREs RNA.2023;[Epub]     CrossRef
    • Age‐associated adipose tissue inflammation promotes monocyte chemotaxis and enhances atherosclerosis
      Jianrui Song, Diana Farris, Paola Ariza, Smriti Moorjani, Mita Varghese, Muriel Blin, Judy Chen, Daniel Tyrrell, Min Zhang, Kanakadurga Singer, Morgan Salmon, Daniel R. Goldstein
      Aging Cell.2023;[Epub]     CrossRef
    • Obesity, diabetes mellitus, and cardiometabolic risk: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2023
      Harold Edward Bays, Shagun Bindlish, Tiffany Lowe Clayton
      Obesity Pillars.2023; 5: 100056.     CrossRef
    • A role of STING signaling in obesity-induced lung inflammation
      Yong Qi, Zhuhua Wu, Dan Chen, Li Zhu, Yunlei Yang
      International Journal of Obesity.2023; 47(4): 325.     CrossRef
    • Estrogens in Adipose Tissue Physiology and Obesity-Related Dysfunction
      Alina Kuryłowicz
      Biomedicines.2023; 11(3): 690.     CrossRef
    • White Adipose Tissue Dysfunction: Pathophysiology and Emergent Measurements
      Natalia Santillana, Camila Astudillo-Guerrero, Amanda D’Espessailles, Gonzalo Cruz
      Nutrients.2023; 15(7): 1722.     CrossRef
    • Pleiotropic and multi-systemic actions of physical exercise on PGC-1α signaling during the aging process
      Ivo Vieira de Sousa Neto, Ana Paula Pinto, Vitor Rosetto Muñoz, Rita de Cássia Marqueti, José Rodrigo Pauli, Eduardo Rochete Ropelle, Adelino Sanchez Ramos da Silva
      Ageing Research Reviews.2023; 87: 101935.     CrossRef
    • The impact of metabolic endotoxaemia on the browning process in human adipocytes
      Farah Omran, Alice M. Murphy, Awais Z. Younis, Ioannis Kyrou, Jana Vrbikova, Vojtech Hainer, Petra Sramkova, Martin Fried, Graham Ball, Gyanendra Tripathi, Sudhesh Kumar, Philip G. McTernan, Mark Christian
      BMC Medicine.2023;[Epub]     CrossRef
    • Molecular Mechanisms of Obesity-Induced Development of Insulin Resistance and Promotion of Amyloid-β Accumulation: Dietary Therapy Using Weak Organic Acids via Improvement of Lowered Interstitial Fluid pH
      Yoshinori Marunaka
      Biomolecules.2023; 13(5): 779.     CrossRef
    • From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs
      Gina Cavaliere, Fabiano Cimmino, Giovanna Trinchese, Angela Catapano, Lidia Petrella, Margherita D’Angelo, Lucio Lucchin, Maria Pina Mollica
      Antioxidants.2023; 12(6): 1172.     CrossRef
    • Receptor for the Advanced Glycation End Products (RAGE) Pathway in Adipose Tissue Metabolism
      Klaudia Gutowska, Krzysztof Czajkowski, Alina Kuryłowicz
      International Journal of Molecular Sciences.2023; 24(13): 10982.     CrossRef
    • Aerobic and Resistance Training Attenuate Differently Knee Joint Damage Caused by a High-Fat–High-Sucrose Diet in a Rat Model
      Nada Abughazaleh, Kevin Boldt, Jaqueline Lourdes Rios, Stela Marcia Mattiello, Kelsey H. Collins, Ruth-Anne Seerattan, Walter Herzog
      CARTILAGE.2023;[Epub]     CrossRef
    • Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction
      Dandan Jia, Zhenjun Tian, Ru Wang
      Ageing Research Reviews.2023; 91: 102087.     CrossRef
    • Mitochondrial dynamics and metabolism across skin cells: implications for skin homeostasis and aging
      Ines Martic, Federica Papaccio, Barbara Bellei, Maria Cavinato
      Frontiers in Physiology.2023;[Epub]     CrossRef
    • Influence of Breastfeeding on the State of Meta-Inflammation in Obesity—A Narrative Review
      Dominika Mazur, Małgorzata Satora, Anna K. Rekowska, Zuzanna Kabała, Aleksandra Łomża, Żaneta Kimber-Trojnar, Bożena Leszczyńska-Gorzelak
      Current Issues in Molecular Biology.2023; 45(11): 9003.     CrossRef
    • AGER-1 Long Non-Coding RNA Levels Correlate with the Expression of the Advanced Glycosylation End-Product Receptor, a Regulator of the Inflammatory Response in Visceral Adipose Tissue of Women with Obesity and Type 2 Diabetes Mellitus
      Klaudia Gutowska, Krzysztof Koźniewski, Michał Wąsowski, Marta Izabela Jonas, Zbigniew Bartoszewicz, Wojciech Lisik, Maurycy Jonas, Artur Binda, Paweł Jaworski, Wiesław Tarnowski, Bartłomiej Noszczyk, Monika Puzianowska-Kuźnicka, Krzysztof Czajkowski, Ali
      International Journal of Molecular Sciences.2023; 24(24): 17447.     CrossRef
    • Pharmacological treatment with FGF21 strongly improves plasma cholesterol metabolism to reduce atherosclerosis
      Cong Liu, Milena Schönke, Enchen Zhou, Zhuang Li, Sander Kooijman, Mariëtte R Boon, Mikael Larsson, Kristina Wallenius, Niek Dekker, Louise Barlind, Xiao-Rong Peng, Yanan Wang, Patrick C N Rensen
      Cardiovascular Research.2022; 118(2): 489.     CrossRef
    • Obesity-Related Adipose Tissue Remodeling in the Light of Extracellular Mitochondria Transfer
      Simon Lecoutre, Karine Clément, Isabelle Dugail
      International Journal of Molecular Sciences.2022; 23(2): 632.     CrossRef
    • IL-4 polarized human macrophage exosomes control cardiometabolic inflammation and diabetes in obesity
      Tuan Anh Phu, Martin Ng, Ngan K. Vu, Laura Bouchareychas, Robert L. Raffai
      Molecular Therapy.2022; 30(6): 2274.     CrossRef
    • Insulin-inducible THRSP maintains mitochondrial function and regulates sphingolipid metabolism in human adipocytes
      Maria A. Ahonen, Marcus Höring, Van Dien Nguyen, Sami Qadri, Juuso H. Taskinen, Meghana Nagaraj, Martin Wabitsch, Pamela Fischer-Posovszky, You Zhou, Gerhard Liebisch, P. A. Nidhina Haridas, Hannele Yki-Järvinen, Vesa M. Olkkonen
      Molecular Medicine.2022;[Epub]     CrossRef
    • Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
      Chin-Wen Wei, Jennifer Nhieu, Yu-Lung Lin, Li-Na Wei
      International Journal of Obesity.2022; 46(10): 1759.     CrossRef
    • The Role of Adipokines in Pancreatic Cancer
      Qi Wang, Huizhi Wang, Yuntao Ding, Mengtian Wan, Min Xu
      Frontiers in Oncology.2022;[Epub]     CrossRef
    • Epigenetic Reprogramming of the Inflammatory Response in Obesity and Type 2 Diabetes
      Federica Zatterale, Gregory Alexander Raciti, Immacolata Prevenzano, Alessia Leone, Michele Campitelli, Veronica De Rosa, Francesco Beguinot, Luca Parrillo
      Biomolecules.2022; 12(7): 982.     CrossRef
    • Cellular Metabolism and Bioenergetic Function in Human Fibroblasts and Preadipocytes of Type 2 Familial Partial Lipodystrophy
      Cristina Algieri, Chiara Bernardini, Fabiana Trombetti, Elisa Schena, Augusta Zannoni, Monica Forni, Salvatore Nesci
      International Journal of Molecular Sciences.2022; 23(15): 8659.     CrossRef
    • Shared pathobiology identifies AMPK as a therapeutic target for obesity and autosomal dominant polycystic kidney disease
      Ioan-Andrei Iliuta, Xuewen Song, Lauren Pickel, Amirreza Haghighi, Ravi Retnakaran, James Scholey, Hoon-Ki Sung, Gregory R. Steinberg, York Pei
      Frontiers in Molecular Biosciences.2022;[Epub]     CrossRef
    • Hypoxia as a Double-Edged Sword to Combat Obesity and Comorbidities
      Ruwen Wang, Qin Sun, Xianmin Wu, Yiyin Zhang, Xiaorui Xing, Kaiqing Lin, Yue Feng, Mingqi Wang, Yibing Wang, Ru Wang
      Cells.2022; 11(23): 3735.     CrossRef
    • Macrophage and Adipocyte Mitochondrial Dysfunction in Obesity-Induced Metabolic Diseases
      Liwen Wang, Jie Hu, Haiyan Zhou
      The World Journal of Men's Health.2021; 39(4): 606.     CrossRef
    • ESRRA (estrogen related receptor alpha) is a critical regulator of intestinal homeostasis through activation of autophagic flux via gut microbiota
      Sup Kim, June-Young Lee, Seul Gi Shin, Jin Kyung Kim, Prashanta Silwal, Young Jae Kim, Na-Ri Shin, Pil Soo Kim, Minho Won, Sang-Hee Lee, Soo Yeon Kim, Miwa Sasai, Masahiro Yamamoto, Jin-Man Kim, Jin-Woo Bae, Eun-Kyeong Jo
      Autophagy.2021; 17(10): 2856.     CrossRef
    • GDF15 as a central mediator for integrated stress response and a promising therapeutic molecule for metabolic disorders and NASH
      Kook Hwan Kim, Myung-Shik Lee
      Biochimica et Biophysica Acta (BBA) - General Subjects.2021; 1865(3): 129834.     CrossRef
    • The Influence of Obesity and Associated Fatty Acids on Placental Inflammation
      Alison J. Eastman, Rebecca E. Moore, Steven D. Townsend, Jennifer A. Gaddy, David M. Aronoff
      Clinical Therapeutics.2021; 43(2): 265.     CrossRef
    • Targeting the G protein-coupled estrogen receptor (GPER) in obesity and diabetes
      Geetanjali Sharma, Eric R. Prossnitz
      Endocrine and Metabolic Science.2021; 2: 100080.     CrossRef
    • Changes in Body Composition Are Associated with Metabolic Changes and the Risk of Metabolic Syndrome
      Yun Hwan Oh, Seulggie Choi, Gyeongsil Lee, Joung Sik Son, Kyae Hyung Kim, Sang Min Park
      Journal of Clinical Medicine.2021; 10(4): 745.     CrossRef
    • N6-Adenosine Methylation (m6A) RNA Modification: an Emerging Role in Cardiovascular Diseases
      Ye-shi Chen, Xin-ping Ouyang, Xiao-hua Yu, Petr Novák, Le Zhou, Ping-ping He, Kai Yin
      Journal of Cardiovascular Translational Research.2021; 14(5): 857.     CrossRef
    • From Metabolic Syndrome to Neurological Diseases: Role of Autophagy
      Jessica Maiuolo, Micaela Gliozzi, Vincenzo Musolino, Cristina Carresi, Federica Scarano, Saverio Nucera, Miriam Scicchitano, Francesca Bosco, Stefano Ruga, Maria Caterina Zito, Roberta Macri, Rosamaria Bulotta, Carolina Muscoli, Vincenzo Mollace
      Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
    • Absent Exercise-Induced Improvements in Fat Oxidation in Women With Polycystic Ovary Syndrome After High-Intensity Interval Training
      Sofie Lionett, Ida Almenning Kiel, Ragnhild Røsbjørgen, Stian Lydersen, Steen Larsen, Trine Moholdt
      Frontiers in Physiology.2021;[Epub]     CrossRef
    • Roles of interstitial fluid pH and weak organic acids in development and amelioration of insulin resistance
      Yoshinori Marunaka
      Biochemical Society Transactions.2021; 49(2): 715.     CrossRef
    • The Role of Mitochondrial Adaptation and Metabolic Flexibility in the Pathophysiology of Obesity and Insulin Resistance: an Updated Overview
      Dimitrios Tsilingiris, Evangelia Tzeravini, Chrysi Koliaki, Maria Dalamaga, Alexander Kokkinos
      Current Obesity Reports.2021; 10(3): 191.     CrossRef
    • Obesity-Related Inflammation and Endothelial Dysfunction in COVID-19: Impact on Disease Severity
      Andrea De Lorenzo, Vanessa Estato, Hugo C Castro-Faria-Neto, Eduardo Tibirica
      Journal of Inflammation Research.2021; Volume 14: 2267.     CrossRef
    • Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential
      Bruna B. Brandão, Ankita Poojari, Atefeh Rabiee
      International Journal of Molecular Sciences.2021; 22(11): 5906.     CrossRef
    • Metabolic Syndrome in an Aging Society – Role of Oxidant-Antioxidant Imbalance and Inflammation Markers in Disentangling Atherosclerosis
      Sylwia Dziegielewska-Gesiak
      Clinical Interventions in Aging.2021; Volume 16: 1057.     CrossRef
    • Recruitment and remodeling of peridroplet mitochondria in human adipose tissue
      Rebeca Acín-Perez, Anton Petcherski, Michaela Veliova, Ilan Y. Benador, Essam A. Assali, Georgia Colleluori, Saverio Cinti, Alexandra J. Brownstein, Siyouneh Baghdasarian, Masha J. Livhits, Michael W. Yeh, Karthickeyan Chella Krishnan, Laurent Vergnes, Na
      Redox Biology.2021; 46: 102087.     CrossRef
    • New Insights Into Mitochondrial Dysfunction at Disease Susceptibility Loci in the Development of Type 2 Diabetes
      Hannah Maude, Winston Lau, Nikolas Maniatis, Toby Andrew
      Frontiers in Endocrinology.2021;[Epub]     CrossRef
    • Effects of sleeve gastrectomy on bone mass, microstructure of femurs and bone metabolism associated serum factors in obese rats
      Ying Xue, Ran Li, Yong Zhao, Ling Li, Yun Zhou
      BMC Endocrine Disorders.2021;[Epub]     CrossRef
    • The cyclin dependent kinase inhibitor Roscovitine prevents diet-induced metabolic disruption in obese mice
      Nabil Rabhi, Kathleen Desevin, Briana Noel Cortez, Ryan Hekman, Jean Z. Lin, Andrew Emili, Stephen R. Farmer
      Scientific Reports.2021;[Epub]     CrossRef
    • Reliability and variation in mitochondrial respiration in human adipose tissue
      Ronni Eg Sahl, Eva Frederikke Høy Helms, Malte Schmücker, Mathias Flensted-Jensen, Arthur Ingersen, Thomas Morville, Flemming Dela, Jørn Wulff Helge, Steen Larsen
      Adipocyte.2021; 10(1): 605.     CrossRef
    • Inhibition of protein tyrosine phosphatase improves mitochondrial bioenergetics and dynamics, reduces oxidative stress, and enhances adipogenic differentiation potential in metabolically impaired progenitor stem cells
      Katarzyna Kornicka-Garbowska, Lynda Bourebaba, Michael Röcken, Krzysztof Marycz
      Cell Communication and Signaling.2021;[Epub]     CrossRef
    • microRNAs in Human Adipose Tissue Physiology and Dysfunction
      Alina Kurylowicz
      Cells.2021; 10(12): 3342.     CrossRef
    • Aging, obese-insulin resistance, and bone remodeling
      Napatsorn Imerb, Chanisa Thonusin, Nipon Chattipakorn, Siriporn C. Chattipakorn
      Mechanisms of Ageing and Development.2020; 191: 111335.     CrossRef
    • Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes
      Federica Zatterale, Michele Longo, Jamal Naderi, Gregory Alexander Raciti, Antonella Desiderio, Claudia Miele, Francesco Beguinot
      Frontiers in Physiology.2020;[Epub]     CrossRef
    • Is Mitochondrial Dysfunction a Common Root of Noncommunicable Chronic Diseases?
      Alexis Diaz-Vegas, Pablo Sanchez-Aguilera, James R Krycer, Pablo E Morales, Matías Monsalves-Alvarez, Mariana Cifuentes, Beverly A Rothermel, Sergio Lavandero
      Endocrine Reviews.2020;[Epub]     CrossRef
    • Inflammatory Signaling and Brown Fat Activity
      Farah Omran, Mark Christian
      Frontiers in Endocrinology.2020;[Epub]     CrossRef
    • Omega-3 fatty acids as regulators of brown/beige adipose tissue: from mechanisms to therapeutic potential
      Marta Fernández-Galilea, Elisa Félix-Soriano, Ignacio Colón-Mesa, Xavier Escoté, Maria J. Moreno-Aliaga
      Journal of Physiology and Biochemistry.2020; 76(2): 251.     CrossRef
    • Anti-Inflammatory Strategies Targeting Metaflammation in Type 2 Diabetes
      Alina Kuryłowicz, Krzysztof Koźniewski
      Molecules.2020; 25(9): 2224.     CrossRef
    • Obese Adipose Tissue Secretion Induces Inflammation in Preadipocytes: Role of Toll-Like Receptor-4
      Mariana Renovato-Martins, Catharina Moreira-Nunes, Georgia C. Atella, Christina Barja-Fidalgo, João Alfredo de Moraes
      Nutrients.2020; 12(9): 2828.     CrossRef
    • Diabetes and Metabolism Journal in 2020: Good to Great
      In-Kyung Jeong
      Diabetes & Metabolism Journal.2020; 44(1): 1.     CrossRef
    • The Effect of Silibinin on Protein Expression Profile in White Adipose Tissue of Obese Mice
      Fei Wang, Shuchun Chen, Luping Ren, Yichao Wang, Zelin Li, Tiantian Song, He Zhang, Qiwen Yang
      Frontiers in Pharmacology.2020;[Epub]     CrossRef
    • Beneficial Effects of Bariatric Surgery-Induced by Weight Loss on the Proteome of Abdominal Subcutaneous Adipose Tissue
      Bárbara María Varela-Rodríguez, Paula Juiz-Valiña, Luis Varela, Elena Outeiriño-Blanco, Susana Belén Bravo, María Jesús García-Brao, Enrique Mena, José Francisco Noguera, Javier Valero-Gasalla, Fernando Cordido, Susana Sangiao-Alvarellos
      Journal of Clinical Medicine.2020; 9(1): 213.     CrossRef
    • Impact of Skeletal Muscle Mass on Metabolic Health
      Gyuri Kim, Jae Hyeon Kim
      Endocrinology and Metabolism.2020; 35(1): 1.     CrossRef
    • Sea buckthorn (Hippophae rhamnoides L.) oil enhances proliferation, adipocytes differentiation and insulin sensitivity in 3T3-L1 cells
      Ting Zhang, Xuze Qin, Yuxin Cao, Jianxin Zhang, Junxing Zhao
      Food Science and Biotechnology.2020; 29(11): 1511.     CrossRef
    • Adipose tissue secretory profile and cardiometabolic risk in obesity
      Pengcheng Zhang, Daniels Konja, Yu Wang
      Endocrine and Metabolic Science.2020; 1(3-4): 100061.     CrossRef
    • Mitochondrial Dynamics in the Brain Are Associated With Feeding, Glucose Homeostasis, and Whole-Body Metabolism
      Jessica L. Haigh, Lauryn E. New, Beatrice M. Filippi
      Frontiers in Endocrinology.2020;[Epub]     CrossRef
    • Adipogenesis: A Necessary but Harmful Strategy
      Mohammed El Hafidi, Mabel Buelna-Chontal, Fausto Sánchez-Muñoz, Roxana Carbó
      International Journal of Molecular Sciences.2019; 20(15): 3657.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation
      Diabetes Metab J. 2019;43(3):247-256.   Published online March 27, 2019
      Close
    • XML DownloadXML Download
    Figure
    Related articles

    Diabetes Metab J : Diabetes & Metabolism Journal