Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2413 word(s) 2413 2021-11-02 03:35:07 |
2 format correct Meta information modification 2413 2021-11-24 01:56:23 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Stanek, A. Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis. Encyclopedia. Available online: https://encyclopedia.pub/entry/16319 (accessed on 20 April 2024).
Stanek A. Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis. Encyclopedia. Available at: https://encyclopedia.pub/entry/16319. Accessed April 20, 2024.
Stanek, Agata. "Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis" Encyclopedia, https://encyclopedia.pub/entry/16319 (accessed April 20, 2024).
Stanek, A. (2021, November 23). Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis. In Encyclopedia. https://encyclopedia.pub/entry/16319
Stanek, Agata. "Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis." Encyclopedia. Web. 23 November, 2021.
Obesity-Induced Perivascular Adipose Tissue Dysfunction in Vascular Homeostasis
Edit

Perivascular adipose tissue (PVAT) is an additional special type of adipose tissue surrounding blood vessels. Under physiological conditions, PVAT plays a significant role in regulation of vascular tone, intravascular thermoregulation, and vascular smooth muscle cell (VSMC) proliferation. PVAT is responsible for releasing adipocytes-derived relaxing factors (ADRF) and perivascular-derived relaxing factors (PDRF), which have anticontractile properties.

obesity perivascular adipose tissue exercise endothelial dysfunction

1. Introduction

Today, an increasing prevalence of obesity is observed in many countries, since a third of the worldwide population is described as obese or overweight [1]. National survey data from 2000 to 2018 in the USA reported that obesity prevalence increased to over 42% among adults, and the prevalence of severe obesity (BMI ≥ 40 kg/m 2) doubled to 9.2% over the study period [2]. Weight problems and obesity are increasing at a rapid rate in most of the EU Member States, with estimates of 52.7% of the EU’s population being overweight in 2019 [3]. According to the Global Burden of Disease study, 4.7 million people died prematurely in 2017 as a result of obesity [4]. Obesity is a risk factor for developing many disorders such as diabetes mellitus, hypertension, cardiovascular events, obstructive sleep apnea syndrome, certain cancers, and musculoskeletal diseases [5]. Obesity also has a negative impact on quality of life and increases the costs of healthcare [6][7].

Adipose tissue is known as an endocrine organ. By producing adipokines, it regulates various metabolism pathways and processes such as insulin sensitivity, energy metabolism, blood flow, and even inflammatory stage [8][9]. Adipose tissue is divided into two main subtypes: white (WAT) and brown (BAT), according to their characteristic and different properties. WAT is responsible for storage of the excess of energy as fatty acids, while BAT mostly specializes in thermogenesis [10]. There is also a third type of adipocyte, termed the “beige” adipocyte. It is a brown adipocyte that arises within white adipose depots and also has thermogenic capacity [11].

Perivascular adipose tissue (PVAT) is an additional special type of adipose tissue surrounding blood vessels. PVAT is located around the aorta, coronary arteries, small and resistance vessels, and vasculature of the musculoskeletal system [12][13][14] On the contrary, PVAT is absent among cerebral vessels [15]. It consists of stem cells, adipocytes, mast cells, and nerves [16][17]. PVAT lies outside the adventitia, with no laminar structures or any organized barrier separating them from each other. Although PVAT characteristics resemble both brown and white adipose tissues, recent evidence suggests that PVAT develops from its own distinct precursors, implying a closer link between PVAT and the vascular system [18]. PVAT at different anatomical locations presents different phenotypes. PVAT demonstrates WAT, BAT, and mixed phenotypes, depending on their anatomical placement [19]. In the abdominal PVAT, white adipocytes are more abundant, whereas thoracic PVAT contains more brown adipocytes. These regional differences in PVAT could explain the higher susceptibility of the abdominal aorta to atherosclerosis compared to the thoracic aorta [20][21][22]. Moreover, gender influences differences in PVAT. After menopause in women, there is an increase in perivascular and pericardial adipose tissue, and additionally, the volume of aortic PVAT positively correlates with the reduction in estradiol [23][24]. Additionally, in obesity experimental models, the PVAT mass and adipocyte size are increased [25]. PVAT, similar to every other adipose tissue, secretes cytokines, hormones, growth factors, and adipokines. It plays a beneficial role as long as adipokine levels with opposing properties remain in equilibrium. In obesity, PVAT becomes dysfunctional and exerts detrimental effects on vascular homeostasis [26].

2. The Influence of PVAT-Derived Factors on Vascular Function

Under physiological conditions, PVAT plays a significant role in the regulation of vascular tone, intravascular thermoregulation, and vascular smooth muscle cell (VSMC) proliferation ( Figure 1 ) [27][28][29]. PVAT exhibits an anticontractile effect as a response to several factors such as endothelin-1, phenylephrine, angiotensin II, and serotonin [30][31][32]. PVAT anticontractile factors are divided into adipocytes-derived relaxing factors (ADRF) and perivascular-derived relaxing factors (PDRF) [30][31][32][33]. On the other hand, PVAT induces vasoconstriction by releasing angiotensin II [34] and the superoxide anion [35]. These factors affect vascular tone via endocrine and paracrine mechanisms. Moreover, VSMCs play a significant role in maintaining the balance between vasoconstriction and vasodilator signals. However, PVAT, as a special adipose tissue, is not only a mechanical support for the vasculature but plays a vital role in the homeostasis of the vascular system, sharing a status no less important than that of the endothelium [36].

Hydrogen sulfide (H 2S) is a gaseous factor, which is produced by PVAT, endothelial cells, and VSMCs, controlling the vascular tone. H 2S-induced vasodilation is caused by activation of BK channels in VSMCs, which leads towards cell membrane hyperpolarization, inactivation of voltage-dependent l-type Ca 2+ channels, and a decrease in intracellular Ca 2+ concentration [37]. In addition, H 2S leads to a dose-dependent decrease in intracellular pH, which causes the vasodilation. It is suggested that the Cl − /HCO 3− ionic exchanger is engaged in this process [38]. The shortage of H 2S is important in the development of various cardiovascular diseases, such as hypertension, atherosclerosis, and heart failure [39].

Components of the renin–angiotensin–aldosterone system (RAAS) are present in the aortic and mesenteric PVAT, except renin [40]. The effect of factors on vascular tone is different. Angiotensin 1–7 induce vasodilation by endothelium-dependent mechanisms. After the activation of the Mas receptors, located in the endothelium, the synthesis of NO is increased, which leads to vasodilation by the activation of BK channels [41]. On the contrary, angiotensin II, which is also produced by PVAT, induces vasoconstriction. There are regional differences in angiotensin II synthesis by PVAT, while it is greater in mesenteric adipose tissue than in the periaortic adipose tissue [35]. Moreover, angiotensin II in increased concentration activates immune cells, which can produce cytokines and proinflammatory mediators [42].

Nitric oxide (NO) is a well-known endogenous gas with vasodilative properties, which is produced in almost all human cells. There are three isoforms of NOS: neuronal NOS (nNOS), inducible NOS (iNOS), and endothelial NOS (eNOS) [43]. Each of them is characterized by different attributes. nNOS is present in cells of the central and peripheral nervous system, where produced NO acts as a neurotransmitter and plays a role in the central regulation of blood pressure [44]. iNOS is activated by inflammatory cytokines and plays a role in inflammation; additionally, iNOS is Ca 2+ independent, unlike other isoforms [43]. eNOS, which is located in endothelial cells, regulates blood pressure locally and has an antiatherosclerotic effect [44][45]. PVAT is responsible for increased production of NO by a direct mechanism, while eNOS isoform is also present in PVAT, where NO is directly produced and released affecting vasculature [46]. In addition, NO produced in PVAT positively regulates adiponectin release by PVAT [47]. On the other hand, PVAT-derived factors, mentioned in previous sections, increase NO production, which is responsible for activations of BK channels and stimulating cGMP synthesis endothelium and smooth muscle cells.

3. The Role of Inflammation, Oxidative Stress, and Hypoxia in Obesity

Obesity is characterized by an excessive level of triglycerides and lipids, which are stored in adipocytes. It leads to their hyperplasia and hypertrophy, where hyperplasia is a well-tolerated complication. In contrast, it is suggested that the capacity of lipid storage and subsequent growth in adipocyte size is limited, and exceeding this threshold induces serious molecular changes and induces cellular dysfunction and death of adipocytes [48]. Moreover, enlarged adipocytes induce elevation of IL-6, IL-8, and leptin and decrease the level of adiponectin, which leads to consequent accumulation of inflammatory factors in PVAT [49][50]. Cytokines, fatty acids, and cell-free DNA, which are excreted after adipocytes apoptosis, induce migration of macrophages to the adipose tissue. Adipose tissue macrophages are divided into two subgroups, which differ from each other by type of secreted cytokines and cell markers: M1 with an inflammatory profile and M2 with an immunosuppressive feature [51]. The M1 subclass secretes cytokines such as TNF-α, IL-6, and IL-1β and plays a significant role in inducing an inflammatory state in adipose tissue, which is important in the development of vascular disorders. Obesity is accompanied by a chronic low-grade inflammatory state, which is confirmed by an elevated level of inflammatory markers, especially C-reactive protein and IL-6, which are significantly higher among obese nonmorbid patients and positively correlates with BMI [52][53].

The excess of carbohydrates, fatty acids, and hyper nutrition induce oxidative stress activation by various pathways such as glycoxidation, oxidative phosphorylation in mitochondria, and NADPH oxidase (NOX) activation with consequent reactive oxygen species (ROS) production [54][55]. The increase in NOX activity leads to excessive production of the superoxide anion (O 2− ), which can react with DNA, lipids, and proteins leading to their destruction [56]. Moreover, O 2− leads to the alteration of NO activity and consequent endothelial dysfunction and cardiovascular events among obese populations [57]. In addition, an elevated level of ROS induces VSMC proliferation and remodeling, which contribute to hypertension development and increased risk of cardiovascular events [58][59].

Moreover, hypertrophy of adipocytes does not proceed hand in hand with angiogenesis, and the demand of tissues for oxygen is greater than the supply. As a result, hypoxia and consequent necrosis and inflammation occur [60]. Hypoxia-inducible factor (HIF-1α), which is increased in adipose tissue among obese individuals, plays the role of mediator in hypoxia. HIF-1α induces the elevation of IL-6 and TNF-α activity and reduces adiponectin concentration [61].

4. The Potential Influence of Diet on PVAT-Derived Factors among Obese Patients

Besides exercise, an adequate diet is another beneficial intervention in weight loss and obesity treatment. Nowadays, there are a wide variety of diet strategies, which differ from one another in terms of the percentage content of macronutrients, such as carbohydrates, proteins, and fats. However, a reduction in daily calorie intake is a universal rule and a recommended strategy in weight loss [62]. Some data that present the influence of dietary intervention directly on PVAT are available. Nevertheless, the association between diets and PVAT are not clearly understood. Reports mainly refer to animal models, in which high-carbohydrate (HC) diets induce obesity and the consequent loss of the anticontractile effect of PVAT by an imbalance in PVAT-derived factor secretion [63]. In contrast, Costa et al. have shown that consuming an HC diet for 4 weeks enhanced the release of vasodilatory factors from PVAT, suggesting that this could be a compensatory adaptive characteristic in order to preserve the vascular function during the initial stages of obesity [64]. Additionally, it is suggested that imbalanced diets can cause PVAT inflammation and dysfunction as well as impaired vascular function. The recent published study has showed that a high-fat (HF) and a high-sucrose (HS) diet affected PVAT at different sites. Sasoh et al. have presented characteristic differences in the effects of HF and HS diets on PVAT and aortae [65]. A HF diet induced an increased number of large-sized lipid droplets and increased cluster of differentiation (CD) 68+ macrophage- and monocyte chemotactic protein (MCP)-1-positive areas in the abdominal aortic PVAT (aPVAT). Furthermore, a HF diet caused a decreased collagen fiber-positive area and increased CD68+ macrophage- and MCP-1-positive areas in the abdominal aorta. In contrast, a HS diet induced an increased number of large-sized lipid droplets, increased CD68+ macrophage- and MCP-1-positive areas, and decreased UCP-1 positive area in the thoracic aortic PVAT (tPVAT). Moreover, a HS diet caused a decreased collagen fiber-positive area and increased CD68+ macrophage- and MCP-1-positive areas in the thoracic aorta. However, there were some factors that did not follow the trend to this variation. For example, angiotensinogen levels were increased in both tPVAT and aPVAT of the HF group. The authors concluded that the potential mechanisms underlying these effects may be related to the different adipocyte species that comprise tPVAT and aPVAT [65]. Victorio et al. reported that the effect of HF and HS diets on PVAT differs depending on sex [66]. The anti-contractile effect of PVAT was measured by comparing the phenylephrine-induced contraction in mesenteric arteries after 3 and 5 months of HF or HF+HS diet among male and female mice. The results showed that anticontractile function was impaired after 3 months of both obesogenic diets among females, while among males, the anti-contractile effect remained comparable during the experiment. Moreover, the assessment of PVAT-derived endothelial function after acetylcholine administration likewise demonstrated differences between sexes, while obesogenic diet among females induces endothelial dysfunction after 3 months and only after 5 months among males.

However, there are many reports that relate the positive impact of different diets on inflammatory state, oxidative stress, NO, adiponectin, or leptin concentration. Thus, one could conclude that similar changes could be observed in PVAT; however, further studies should be conducted.

The Mediterranean diet (MD) is the most popular diet and is commonly known as a healthy, balanced diet with proven efficiency in reducing the cardiovascular risk among high-risk patients and reducing overall mortality [67][68]. A typical MD contains 55–60% carbohydrates, mainly complex ones, 25–30% polyunsaturated and monounsaturated fats, and 15–20% proteins, and meals are generally based on fish, nuts, olive oil, and plant-based foods [69]. Luisi et al. reported that the implementation of an MD for 3 months among overweight/obese patients, with high-quality extra virgin olive oil, induced weight loss and the significant elevation of adiponectin levels [70]. Interestingly, among normal weight controls, the MD has no impact on weight, and the increase in adiponectin concentration was not as considerable as that found among overweight/obese patients. It can be concluded that weight loss and the consequent reduction in adipose tissue contribute to a size reduction in adipocytes and an improvement in adiponectin synthesis and release. Among both groups, the concentration of IL-6 significantly decreased after dietary intervention, which provides proof of the anti-inflammatory properties of MD [70]. Moreover, it is suggested that the higher the amount of fiber in one’s diet, the greater the adiponectin concentration in one’s blood [71].

Recently, the ketogenic diet has become very popular due to its therapeutic properties in relation to different diseases. It has been widely used in drug-resistant epilepsy with good outcomes and is increasingly being used in metabolic disorders such as obesity or diabetes mellitus [72][73]. The ketogenic diet is characterized by low carbohydrates and high fat, inducing changes in the metabolism of energy substrates, with a switch from glucose to fatty acids [72]. A very low-calorie ketogenic diet (VLCKD) is a special type of caloric reduction diet characterized by a very low or extremely low daily food energy intake, circa 800 kcal per day [74]. It provides 30–50 g of carbohydrates, about 30–40 g of fats, and 0.8–1.5 g/kg of ideal body weight (IBW) of proteins [75]. Monda et al. reported that obese patients who consumed a VLCKD diet for 8 weeks presented with a significant body mass reduction, a decreased concentration of inflammatory markers such as IL-6, TNF-α, and CRP, and a significant elevation in the level of adiponectin in their blood [74]. This relatively short period of intervention induced a significant multifactorial improvement; however, the main limitation of the aforementioned study is the small sample size. In other reports, the ketogenic diet has also been proven to have anti-inflammatory properties [76][77].

References

  1. Global Burden of Disease Study 2015 (GBD 2015) Obesity and Overweight Prevalence 1980–2015 | GHDx. Available online: http://ghdx.healthdata.org/record/ihme-data/gbd-2015-obesity-and-overweight-prevalence-1980-2015 (accessed on 3 August 2021).
  2. Hales, C.M. Prevalence of Obesity and Severe Obesity Among Adults: United States, 2017–2018. NCHS Data Brief 2020, 360, 8.
  3. Overweight and Obesity-BMI Statistics. Available online: https://ec.europa.eu/eurostat/statistics-explained/index.php?title=Overweight_and_obesity_-_BMI_statistics (accessed on 6 September 2021).
  4. Dai, H.; Alsalhe, T.A.; Chalghaf, N.; Riccò, M.; Bragazzi, N.L.; Wu, J. The Global Burden of Disease Attributable to High Body Mass Index in 195 Countries and Territories, 1990–2017: An Analysis of the Global Burden of Disease Study. PLoS Med. 2020, 17, e1003198.
  5. Singh, G.M.; Danaei, G.; Farzadfar, F.; Stevens, G.A.; Woodward, M.; Wormser, D.; Kaptoge, S.; Whitlock, G.; Qiao, Q.; Lewington, S.; et al. The Age-Specific Quantitative Effects of Metabolic Risk Factors on Cardiovascular Diseases and Diabetes: A Pooled Analysis. PLoS ONE 2013, 8, e65174.
  6. Kolotkin, R.L.; Andersen, J.R. A Systematic Review of Reviews: Exploring the Relationship between Obesity, Weight Loss and Health-related Quality of Life. Clin. Obes. 2017, 7, 273–289.
  7. Tremmel, M.; Gerdtham, U.-G.; Nilsson, P.M.; Saha, S. Economic Burden of Obesity: A Systematic Literature Review. Int. J. Environ. Res. Public Health 2017, 14, 435.
  8. Fernández-Alfonso, M.S.; Gil-Ortega, M.; García-Prieto, C.F.; Aranguez, I.; Ruiz-Gayo, M.; Somoza, B. Mechanisms of Perivascular Adipose Tissue Dysfunction in Obesity. Int. J. Endocrinol. 2013, 2013, 402053.
  9. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223.
  10. Saely, C.H.; Geiger, K.; Drexel, H. Brown versus White Adipose Tissue: A Mini-Review. Gerontology 2012, 58, 15–23.
  11. Kiefer, F.W.; Cohen, P.; Plutzky, J. Fifty Shades of Brown: Perivascular Fat, Thermogenesis, and Atherosclerosis. Circulation 2012, 126, 1012–1015.
  12. Police, S.B.; Thatcher, S.E.; Charnigo, R.; Daugherty, A.; Cassis, L.A. Obesity Promotes Inflammation in Periaortic Adipose Tissue and Angiotensin II-Induced Abdominal Aortic Aneurysm Formation. Arter. Thromb. Vasc. Biol. 2009, 29, 1458–1464.
  13. Verlohren, S.; Dubrovska, G.; Tsang, S.-Y.; Essin, K.; Luft, F.C.; Huang, Y.; Gollasch, M. Visceral Periadventitial Adipose Tissue Regulates Arterial Tone of Mesenteric Arteries. Hypertension 2004, 44, 271–276.
  14. Yudkin, J.S.; Eringa, E.; Stehouwer, C.D.A. “Vasocrine” Signalling from Perivascular Fat: A Mechanism Linking Insulin Resistance to Vascular Disease. Lancet 2005, 365, 1817–1820.
  15. Gao, Y.-J. Dual Modulation of Vascular Function by Perivascular Adipose Tissue and Its Potential Correlation with Adiposity/Lipoatrophy-Related Vascular Dysfunction. Curr. Pharm. Des. 2007, 13, 2185–2192.
  16. Takaoka, M.; Suzuki, H.; Shioda, S.; Sekikawa, K.; Saito, Y.; Nagai, R.; Sata, M. Endovascular Injury Induces Rapid Phenotypic Changes in Perivascular Adipose Tissue. Arter. Thromb. Vasc. Biol. 2010, 30, 1576–1582.
  17. Hu, Y.; Zhang, Z.; Torsney, E.; Afzal, A.R.; Davison, F.; Metzler, B.; Xu, Q. Abundant Progenitor Cells in the Adventitia Contribute to Atherosclerosis of Vein Grafts in ApoE-Deficient Mice. J. Clin. Investig. 2004, 113, 1258–1265.
  18. Qi, X.-Y.; Qu, S.-L.; Xiong, W.-H.; Rom, O.; Chang, L.; Jiang, Z.-S. Perivascular Adipose Tissue (PVAT) in Atherosclerosis: A Double-Edged Sword. Cardiovasc. Diabetol. 2018, 17, 134.
  19. Gil-Ortega, M.; Somoza, B.; Huang, Y.; Gollasch, M.; Fernández-Alfonso, M.S. Regional Differences in Perivascular Adipose Tissue Impacting Vascular Homeostasis. Trends Endocrinol. Metab. 2015, 26, 367–375.
  20. Lasar, D.; Julius, A.; Fromme, T.; Klingenspor, M. Browning Attenuates Murine White Adipose Tissue Expansion during Postnatal Development. Biochim. Biophys Acta 2013, 1831, 960–968.
  21. Lu, D.; Wang, W.; Xia, L.; Xia, P.; Yan, Y. Gene Expression Profiling Reveals Heterogeneity of Perivascular Adipose Tissues Surrounding Coronary and Internal Thoracic Arteries. Acta Biochim. Biophys. Sin. 2017, 49, 1075–1082.
  22. Britton, K.A.; Pedley, A.; Massaro, J.M.; Corsini, E.M.; Murabito, J.M.; Hoffmann, U.; Fox, C.S. Prevalence, Distribution, and Risk Factor Correlates of High Thoracic Periaortic Fat in the Framingham Heart Study. J. Am. Heart Assoc. 2012, 1, e004200.
  23. El Khoudary, S.R.; Shields, K.J.; Janssen, I.; Hanley, C.; Budoff, M.J.; Barinas-Mitchell, E.; Everson-Rose, S.A.; Powell, L.H.; Matthews, K.A. Cardiovascular Fat, Menopause, and Sex Hormones in Women: The SWAN Cardiovascular Fat Ancillary Study. J. Clin. Endocrinol. Metab. 2015, 100, 3304–3312.
  24. Victorio, J.A.; da Costa, R.M.; Tostes, R.C.; Davel, A.P. Modulation of Vascular Function by Perivascular Adipose Tissue: Sex Differences. Curr. Pharm. Des. 2020, 26, 3768–3777.
  25. Marchesi, C.; Ebrahimian, T.; Angulo, O.; Paradis, P.; Schiffrin, E.L. Endothelial Nitric Oxide Synthase Uncoupling and Perivascular Adipose Oxidative Stress and Inflammation Contribute to Vascular Dysfunction in a Rodent Model of Metabolic Syndrome. Hypertension 2009, 54, 1384–1392.
  26. Sowka, A.; Dobrzyn, P. Role of Perivascular Adipose Tissue-Derived Adiponectin in Vascular Homeostasis. Cells 2021, 10, 1485.
  27. Chang, L.; Villacorta, L.; Li, R.; Hamblin, M.; Xu, W.; Dou, C.; Zhang, J.; Wu, J.; Zeng, R.; Chen, Y.E. Loss of Perivascular Adipose Tissue on Peroxisome Proliferator-Activated Receptor-γ Deletion in Smooth Muscle Cells Impairs Intravascular Thermoregulation and Enhances Atherosclerosis. Circulation 2012, 126, 1067–1078.
  28. Ozen, G.; Daci, A.; Norel, X.; Topal, G. Human perivascular adipose tissue dysfunction as a cause of vascular disease: Focus on vascular tone and wall remodeling. Eur. J. Pharm. 2015, 766, 16–24.
  29. Miao, C.-Y.; Li, Z.-Y. The Role of Perivascular Adipose Tissue in Vascular Smooth Muscle Cell Growth. Br. J. Pharm. 2012, 165, 643–658.
  30. Gollasch, M. Vasodilator Signals from Perivascular Adipose Tissue. Br. J. Pharmacol. 2012, 165, 633.
  31. Szasz, T.; Webb, R.C. Perivascular Adipose Tissue: More than Just Structural Support. Clin. Sci. 2012, 122, 1–12.
  32. Gálvez-Prieto, B.; Somoza, B.; Gil-Ortega, M.; García-Prieto, C.F.; de las Heras, A.I.; González, M.C.; Arribas, S.; Aranguez, I.; Bolbrinker, J.; Kreutz, R.; et al. Anticontractile Effect of Perivascular Adipose Tissue and Leptin Are Reduced in Hypertension. Front. Pharm. 2012, 3, 103.
  33. Lee, Y.-C.; Chang, H.-H.; Chiang, C.-L.; Liu, C.-H.; Yeh, J.-I.; Chen, M.-F.; Chen, P.-Y.; Kuo, J.-S.; Lee, T.J.F. Role of Perivascular Adipose Tissue-Derived Methyl Palmitate in Vascular Tone Regulation and Pathogenesis of Hypertension. Circulation 2011, 124, 1160–1171.
  34. Gálvez-Prieto, B.; Bolbrinker, J.; Stucchi, P.; de Las Heras, A.I.; Merino, B.; Arribas, S.; Ruiz-Gayo, M.; Huber, M.; Wehland, M.; Kreutz, R.; et al. Comparative Expression Analysis of the Renin-Angiotensin System Components between White and Brown Perivascular Adipose Tissue. J. Endocrinol. 2008, 197, 55–64.
  35. Gao, Y.; Takemori, K.; Su, L.; An, W.; Lu, C.; Sharma, A.; Lee, R. Perivascular Adipose Tissue Promotes Vasoconstriction: The Role of Superoxide Anion. Cardiovasc. Res. 2006, 71, 363–373.
  36. Cheng, C.K.; Bakar, H.A.; Gollasch, M.; Huang, Y. Perivascular Adipose Tissue: The Sixth Man of the Cardiovascular System. Cardiovasc. Drugs 2018, 32, 481–502.
  37. Wang, R. Shared Signaling Pathways among Gasotransmitters. Proc. Natl. Acad. Sci. USA 2012, 109, 8801–8802.
  38. Lee, S.W.; Cheng, Y.; Moore, P.K.; Bian, J.-S. Hydrogen Sulphide Regulates Intracellular PH in Vascular Smooth Muscle Cells. Biochem. Biophys. Res. Commun 2007, 358, 1142–1147.
  39. Zhong, X.; Wang, Y.; Wu, J.; Sun, A.; Yang, F.; Zheng, D.; Li, T.; Dong, S.; Zhao, Y.; Yang, G.; et al. Calcium Sensing Receptor Regulating Smooth Muscle Cells Proliferation through Initiating Cystathionine-Gamma-Lyase/Hydrogen Sulfide Pathway in Diabetic Rat. Cell Physiol. Biochem. 2015, 35, 1582–1598.
  40. Xia, N.; Li, H. The Role of Perivascular Adipose Tissue in Obesity-induced Vascular Dysfunction. Br. J. Pharm. 2017, 174, 3425–3442.
  41. Lee, R.M.K.W.; Lu, C.; Su, L.-Y.; Gao, Y.-J. Endothelium-Dependent Relaxation Factor Released by Perivascular Adipose Tissue. J. Hypertens 2009, 27, 782–790.
  42. Kusters, P.J.H.; Lutgens, E.; Seijkens, T.T.P. Exploring Immune Checkpoints as Potential Therapeutic Targets in Atherosclerosis. Cardiovasc. Res. 2018, 114, 368–377.
  43. Bredt, D.S.; Snyder, S.H. Isolation of Nitric Oxide Synthetase, a Calmodulin-Requiring Enzyme. Proc. Natl. Acad. Sci. USA 1990, 87, 682–685.
  44. Förstermann, U.; Closs, E.I.; Pollock, J.S.; Nakane, M.; Schwarz, P.; Gath, I.; Kleinert, H. Nitric Oxide Synthase Isozymes. Characterization, Purification, Molecular Cloning, and Functions. Hypertension 1994, 23 Pt 2, 1121–1131.
  45. Hong, F.; Liang, X.; Liu, W.; Lv, S.; He, S.; Kuang, H.; Yang, S. Roles of ENOS in Atherosclerosis Treatment. Inflamm. Res. 2019, 68, 429–441.
  46. Xia, N.; Horke, S.; Habermeier, A.; Closs, E.I.; Reifenberg, G.; Gericke, A.; Mikhed, Y.; Münzel, T.; Daiber, A.; Förstermann, U.; et al. Uncoupling of Endothelial Nitric Oxide Synthase in Perivascular Adipose Tissue of Diet-Induced Obese Mice. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 78–85.
  47. Weston, A.H.; Egner, I.; Dong, Y.; Porter, E.L.; Heagerty, A.M.; Edwards, G. Stimulated Release of a Hyperpolarizing Factor (ADHF) from Mesenteric Artery Perivascular Adipose Tissue: Involvement of Myocyte BKCa Channels and Adiponectin. Br. J. Pharmacol. 2013, 169, 1500–1509.
  48. Cotillard, A.; Poitou, C.; Torcivia, A.; Bouillot, J.-L.; Dietrich, A.; Klöting, N.; Grégoire, C.; Lolmede, K.; Blüher, M.; Clément, K. Adipocyte Size Threshold Matters: Link with Risk of Type 2 Diabetes and Improved Insulin Resistance After Gastric Bypass. J. Clin. Endocrinol. Metab. 2014, 99, E1466–E1470.
  49. Chandalia, M.; Lin, P.; Seenivasan, T.; Livingston, E.H.; Snell, P.G.; Grundy, S.M.; Abate, N. Insulin Resistance and Body Fat Distribution in South Asian Men Compared to Caucasian Men. PLoS ONE 2007, 2, e812.
  50. Skurk, T.; Alberti-Huber, C.; Herder, C.; Hauner, H. Relationship between Adipocyte Size and Adipokine Expression and Secretion. J. Clin. Endocrinol. Metab. 2007, 92, 1023–1033.
  51. Martinez, F.O.; Gordon, S. The M1 and M2 Paradigm of Macrophage Activation: Time for Reassessment. F1000Prime Rep. 2014, 6, 13.
  52. Sudhakar, M.; Silambanan, S.; Chandran, A.S.; Prabhakaran, A.A.; Ramakrishnan, R. C-Reactive Protein (CRP) and Leptin Receptor in Obesity: Binding of Monomeric CRP to Leptin Receptor. Front. Immunol. 2018, 9, 1167.
  53. Gnacińska, M.; Małgorzewicz, S.; Guzek, M.; Lysiak-Szydłowska, W.; Sworczak, K. Adipose Tissue Activity in Relation to Overweight or Obesity. Endokrynol. Pol. 2010, 61, 160–168.
  54. Savini, I.; Catani, M.V.; Evangelista, D.; Gasperi, V.; Avigliano, L. Obesity-Associated Oxidative Stress: Strategies Finalized to Improve Redox State. Int. J. Mol. Sci. 2013, 14, 10497–10538.
  55. Codoñer-Franch, P.; Tavárez-Alonso, S.; Murria-Estal, R.; Tortajada-Girbés, M.; Simó-Jordá, R.; Alonso-Iglesias, E. Elevated Advanced Oxidation Protein Products (AOPPs) Indicate Metabolic Risk in Severely Obese Children. Nutr. Metab. Cardiovasc. Dis. 2012, 22, 237–243.
  56. Jiang, F.; Lim, H.K.; Morris, M.J.; Prior, L.; Velkoska, E.; Wu, X.; Dusting, G.J. Systemic Upregulation of NADPH Oxidase in Diet-Induced Obesity in Rats. Redox Rep. 2011, 16, 223–229.
  57. Beckman, J.S.; Koppenol, W.H. Nitric Oxide, Superoxide, and Peroxynitrite: The Good, the Bad, and the Ugly. Am. J. Physiol.-Cell Physiol. 1996, 271, C1424–C1437.
  58. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C.S. Oxidative Stress and Metabolic Disorders: Pathogenesis and Therapeutic Strategies. Life Sci. 2016, 148, 183–193.
  59. Mulvany, M.J. Small Artery Remodelling in Hypertension. Basic Clin. Pharm. Toxicol. 2012, 110, 49–55.
  60. Goossens, G.H.; Bizzarri, A.; Venteclef, N.; Essers, Y.; Cleutjens, J.P.; Konings, E.; Jocken, J.W.E.; Cajlakovic, M.; Ribitsch, V.; Clément, K.; et al. 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.
  61. Cancello, R.; Henegar, C.; Viguerie, N.; Taleb, S.; Poitou, C.; Rouault, C.; Coupaye, M.; Pelloux, V.; Hugol, D.; Bouillot, J.-L.; et al. Reduction of Macrophage Infiltration and Chemoattractant Gene Expression Changes in White Adipose Tissue of Morbidly Obese Subjects after Surgery-Induced Weight Loss. Diabetes 2005, 54, 2277–2286.
  62. Julia, C.; Péneau, S.; Andreeva, V.A.; Méjean, C.; Fezeu, L.; Galan, P.; Hercberg, S. Weight-Loss Strategies Used by the General Population: How Are They Perceived? PLoS ONE 2014, 9, e97834.
  63. Almabrouk, T.A.M.; White, A.D.; Ugusman, A.B.; Skiba, D.S.; Katwan, O.J.; Alganga, H.; Guzik, T.J.; Touyz, R.M.; Salt, I.P.; Kennedy, S. High Fat Diet Attenuates the Anticontractile Activity of Aortic PVAT via a Mechanism Involving AMPK and Reduced Adiponectin Secretion. Front. Physiol. 2018, 9, 51.
  64. dos Reis Costa, D.E.F.; Silveira, A.L.M.; Campos, G.P.; Nóbrega, N.R.C.; de Araújo, N.F.; de Figueiredo Borges, L.; dos Santos Aggum Capettini, L.; Ferreira, A.V.M.; Bonaventura, D. High-Carbohydrate Diet Enhanced the Anticontractile Effect of Perivascular Adipose Tissue Through Activation of Renin-Angiotensin System. Front. Physiol. 2021, 11, 628101.
  65. Sasoh, T.; Kugo, H.; Kondo, Y.; Miyamoto, K.; Minami, M.; Higashihara, M.; Kawamoto, H.; Takeshita, F.; Moriyama, T.; Zaima, N. Different effects of high-fat and high-sucrose diets on the physiology of perivascular adipose tissues of the thoracic and abdominal aorta. Adipocyte 2021, 10, 412–423.
  66. Victorio, J.A.; Guizoni, D.M.; Freitas, I.N.; Araujo, T.R.; Davel, A.P. Effects of High-Fat and High-Fat/High-Sucrose Diet-Induced Obesity on PVAT Modulation of Vascular Function in Male and Female Mice. Front. Pharm. 2021, 12, 720224.
  67. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34.
  68. Sofi, F.; Macchi, C.; Abbate, R.; Gensini, G.F.; Casini, A. Mediterranean Diet and Health Status: An Updated Meta-Analysis and a Proposal for a Literature-Based Adherence Score. Public Health Nutr. 2014, 17, 2769–2782.
  69. Luisi, M.L.E.; Biffi, B.; Gheri, C.F.; Sarli, E.; Rafanelli, E.; Graziano, E.; Vidali, S.; Fattirolli, F.; Gensini, G.F.; Macchi, C. Efficacy of a Nutritional Education Program to Improve Diet in Patients Attending a Cardiac Rehabilitation Program: Outcomes of a One-Year Follow-Up. Intern. Emerg. Med. 2015, 10, 671–676.
  70. Luisi, M.L.E.; Lucarini, L.; Biffi, B.; Rafanelli, E.; Pietramellara, G.; Durante, M.; Vidali, S.; Provensi, G.; Madiai, S.; Gheri, C.F.; et al. Effect of Mediterranean Diet Enriched in High Quality Extra Virgin Olive Oil on Oxidative Stress, Inflammation and Gut Microbiota in Obese and Normal Weight Adult Subjects. Front. Pharm. 2019, 10, 1366.
  71. Mantzoros, C.S.; Williams, C.J.; Manson, J.E.; Meigs, J.B.; Hu, F.B. Adherence to the Mediterranean Dietary Pattern Is Positively Associated with Plasma Adiponectin Concentrations in Diabetic Women. Am. J. Clin. Nutr. 2006, 84, 328–335.
  72. Ułamek-Kozioł, M.; Czuczwar, S.J.; Januszewski, S.; Pluta, R. Ketogenic Diet and Epilepsy. Nutrients 2019, 11, 2510.
  73. Muscogiuri, G.; Barrea, L.; Laudisio, D.; Pugliese, G.; Salzano, C.; Savastano, S.; Colao, A. The Management of Very Low-Calorie Ketogenic Diet in Obesity Outpatient Clinic: A Practical Guide. J. Transl. Med. 2019, 17.
  74. Monda, V.; Polito, R.; Lovino, A.; Finaldi, A.; Valenzano, A.; Nigro, E.; Corso, G.; Sessa, F.; Asmundo, A.; Di Nunno, N.; et al. Short-Term Physiological Effects of a Very Low-Calorie Ketogenic Diet: Effects on Adiponectin Levels and Inflammatory States. Int. J. Mol. Sci. 2020, 21, 3228.
  75. Kirkpatrick, C.F.; Bolick, J.P.; Kris-Etherton, P.M.; Sikand, G.; Aspry, K.E.; Soffer, D.E.; Willard, K.-E.; Maki, K.C. Review of Current Evidence and Clinical Recommendations on the Effects of Low-Carbohydrate and Very-Low-Carbohydrate (Including Ketogenic) Diets for the Management of Body Weight and Other Cardiometabolic Risk Factors: A Scientific Statement from the National Lipid Association Nutrition and Lifestyle Task Force. J. Clin. Lipidol. 2019, 13, 689–711.e1.
  76. Puchalska, P.; Crawford, P.A. Multi-Dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab. 2017, 25, 262–284.
  77. Bhanpuri, N.H.; Hallberg, S.J.; Williams, P.T.; McKenzie, A.L.; Ballard, K.D.; Campbell, W.W.; McCarter, J.P.; Phinney, S.D.; Volek, J.S. Cardiovascular Disease Risk Factor Responses to a Type 2 Diabetes Care Model Including Nutritional Ketosis Induced by Sustained Carbohydrate Restriction at 1 Year: An Open Label, Non-Randomized, Controlled Study. Cardiovasc. Diabetol. 2018, 17, 56.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 388
Revisions: 2 times (View History)
Update Date: 24 Nov 2021
1000/1000