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 -- 2240 2023-08-24 08:08:00 |
2 layout -1 word(s) 2239 2023-08-24 10:33:40 |

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.
Ali, I.I.; D’souza, C.; Singh, J.; Adeghate, E. The Role of Adropin in Health and Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/48408 (accessed on 06 August 2024).
Ali II, D’souza C, Singh J, Adeghate E. The Role of Adropin in Health and Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/48408. Accessed August 06, 2024.
Ali, Ifrah Ismail, Crystal D’souza, Jaipaul Singh, Ernest Adeghate. "The Role of Adropin in Health and Disease" Encyclopedia, https://encyclopedia.pub/entry/48408 (accessed August 06, 2024).
Ali, I.I., D’souza, C., Singh, J., & Adeghate, E. (2023, August 24). The Role of Adropin in Health and Disease. In Encyclopedia. https://encyclopedia.pub/entry/48408
Ali, Ifrah Ismail, et al. "The Role of Adropin in Health and Disease." Encyclopedia. Web. 24 August, 2023.
The Role of Adropin in Health and Disease
Edit

Adropin is a novel 76-amino acid-peptide that is expressed in different tissues and cells including the liver, pancreas, heart and vascular tissues, kidney, milk, serum, plasma and many parts of the brain. Adropin, encoded by the Enho gene, plays a crucial role in energy homeostasis. Therapeutic peptides show great potential in the treatment of many diseases.

adropin diabetes mellitus obesity

1. Introduction

The levels of adropin in blood circulation have been proposed to direct the metabolic state in skeletal muscle by influencing fuel selection preference towards glucose oxidation in the fed state [1]. Studies have shown that adropin regulates the expression of hepatic lipogenic genes and the PPARγ receptor (peroxisome proliferator-activated receptor gamma), the major regulator of lipogenesis [2]. Moreover, adropin regulates angiogenesis, increases blood flow, boosts capillary density and has a protective role for endothelial cells [3]. Apparently, the tissue level of adropin varies in several physiological and biological conditions such as multiple sclerosis [4], COVID-19 [5], gestational diabetes [6] obstructive sleep apnea [7], rheumatoid arthritis [8], coronary artery ectasia [9], acute mesenteric ischemia [10] and diabetic nephropathy [11].

2. Diabetes Mellitus

When adropin was first discovered, most of the attention was given to its role in lipid and carbohydrate metabolism and insulin resistance. Interestingly, some studies show that adropin deficiency plays a role in the development and progression of chronic diseases, such as diabetes mellitus. Zang et al. reported that serum concentrations of adropin were significantly decreased in Chinese type 2 diabetic patients, compared to control subjects [12]. Other studies confirmed this finding by reporting the downregulation in circulating adropin in adults with type 2 diabetes mellitus [13], liver disease [14] and children with type 1 diabetes mellitus [15]. In contrast, it was elucidated that higher insulin resistance and higher fasting plasma glucose positively correlated with serum adropin levels in patients with type 2 diabetes mellitus [16].
Additionally, low adropin levels have been shown to correlate with a risk of developing diabetic complications such as diabetic retinopathy [17], diabetic nephropathy [18] and gestational diabetes mellitus [19].
As it was reported that serum adropin levels vary between diabetic and normal subjects, several investigations tried to understand the mechanisms underlying these variations. For instance, it was demonstrated that hyperglycemia was associated with increased adropin expression, as well as the signal transducer and activator of transcription 3 (STAT3) activation in the liver of streptozotocin-induced diabetic rats. The mechanism underlying the elevation of adropin levels and Enho gene expression in the diabetic rats was suggested to be through STAT3 activation [20].
During the discovery of adropin, its physiological role was linked to glucose homeostasis. It is thus important to understand the potential role of adropin in controlling hyperglycemia and its effect on insulin-sensitive tissues.
In skeletal muscle, a study that was performed by Gao et al. showed that adropin played a crucial role in modulating glucose utilization in DIO mice with insulin resistance [1]. Adropin was able to promote glucose oxidation and diminish fatty acid oxidation in skeletal muscle, and that led to an increase in glucose uptake and enhanced mitochondrial function. The metabolic actions for enhancing mitochondrial function were mediated by suppressing the activity of peroxisome proliferator-activated receptor gamma coactivator-1a (PGC-1a), a transcription co-activator that regulates the expression of the genes that are involved in fatty acid oxidation. Moreover, adropin promoted skeletal muscle sensitization to insulin signaling actions by increasing insulin-induced Akt phosphorylation and the cell surface expression of glucose transporter 4 (GLUT4) [1].
In contrast, a study utilizing insulin-resistant hepatocytes showed that adropin could reduce glucose production in the liver. Adropin treatment downregulated the transcription of hepatic gluconeogenesis genes by inhibiting the binding site of transcription factors forkhead box protein O1(FoxO1) and cAMP-response element binding protein (CREB), along with their co-activators, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) and CREB regulated transcription coactivator 2 (CRTC2), respectively, to the promoter of gluconeogenesis genes [21][22]. FoxO1-PGC1α and CREB-CRTC2 complex promoter binding is required to activate the transcription of genes that are involved in hepatic glucose production [23][24].
Interestingly, this effect of adropin was not observed in insulin-sensitive hepatocytes [22]. This research team earlier reported that a downregulation in adropin expression led to systemic insulin resistance in mice that were introduced to a high-fat diet for a long period [25].
Furthermore, it was reported that adropin was associated with incretins in obese males with type 2 diabetes receiving a 3-month treatment of liraglutide [26]. There was an increase in plasma adropin levels in those subjects. Liraglutide is an anti-diabetic agent, a specific glucagon-like peptide-1 receptor (GLP-1) agonist, also known as incretin mimetic molecule. Incretins are endogenous peptide hormones that are secreted by the GI tract to stimulate insulin secretion from pancreatic β-cells after meals [27][28][29][30].
It is worth mentioning that irisin, which is a peptide hormone involved in glucose homeostasis [31][32], has a similar effect on incretins, specifically GLP-1. Both adropin and irisin can enhance glucose-stimulated insulin secretion [33]. In addition, the peptide apelin increased plasma GLP-1 levels in rats that were intraperitoneally injected with apelin-13 [34]. However, the role of adropin in inducing incretin secretion and augmenting incretin effect remains unclear and needs more study to elucidate the mechanism by which it regulates these GI tract hormones Table 1.

3. Obesity

Obesity is a major health problem worldwide [39][40]. Studies performed on humans and animal models suggest that adropin may play a role in lipid metabolism and obesity.
C57BL/6J mice that were fed a high-fat diet exhibited a rapid increase in Enho gene expression, while fasting reduced the expression of this gene, when compared to the control mice. However, liver Enho gene expression declined when a high-fat diet was introduced to the mice for a longer period of time, suggesting a regulatory role of the Enho gene in nutrition, but the expression of adropin is diet-dependent [2]. Moreover, Kumar et al. generated an adropin knockout mice, which exhibited increased adiposity [41].
In humans, adropin level was negatively correlated with body mass index (BMI) [42] in diabetic patients, as overweight and obese patients had considerably reduced levels of adropin, compared to lean patients [12]. Moreover, when adropin levels were measured in plasma samples that were obtained from healthy subjects, it was found that the peptide levels correlated negatively with BMI and aging [43]. This observation was supported by studying patients who underwent bariatric surgery and monitoring serum adropin levels before and after surgery [44]. Serum adropin levels were higher 6 months after bariatric surgery than at baseline, leading to the conclusion that, in some patients, body mass reduction may restore the impaired production of adropin. Another study was conducted to investigate the role of adropin in children with obesity or metabolic syndrome [45]. The results of this research showed that—there was no significant difference between the plasma level of adropin in obese children, and those individuals with normal weight Table 1.

4. Cardiovascular Diseases

There is overwhelming evidence that cardiovascular diseases are common in patients suffering from diabetes mellitus [35][46][47][48][49][50][51]. Several studies and reports indicate the involvement of adropin in the functioning of the cardiovascular system. As mentioned previously, the immunoreactivity of adropin has been detected in many tissues, including the three layers of the heart [52].
High cardiac fatty acid oxidation rates and impaired cardiac insulin signaling are associated with decreased cardiac efficiency and various cardiac diseases [53]. Altamimi et al. investigated the effect of adropin on cardiac energy metabolism, insulin signaling and cardiac efficiency [54]. C57Bl/6 mice were injected with a secretable form of adropin (450 nmol/kg, i.p.) three times over 24 h, then they were fasted, and the hearts were isolated and perfused. Altamimi et al. demonstrated that adropin administration improved cardiac function, cardiac efficiency and coronary flow, compared to the untreated mice. Moreover, by measuring glucose and palmitate contribution in catabolic pathways for ATP production, the important role of adropin on the preference of cardiac glucose oxidation and the inhibition of cardiac fatty acid oxidation were reported. In cardiomyocytes, adropin regulates cell bioenergetics through GPR19 activation. The receptor activation leads to stimulation of p44/42 phosphorylation and, consequently, the downregulation of pyruvate dehydrogenase kinase 4 (PDK4) and pyruvate dehydrogenase (PDH) phosphorylation [55][56][57][58].
Hyperlipidemia is a risk factor that is associated with cardiovascular diseases [59]. Akcilar et al. demonstrated the role of a low dose of adropin in reducing hyperlipidemia in rats that were fed a high-fat diet. A reduction in the levels of serum triglycerides, total cholesterol and low-density lipoprotein cholesterol (LDL-C), as well as an increment in the level of high-density lipoprotein cholesterol (HDL-C) were reported [60]. Additionally, adropin administration reduced the mRNA expression of pro-inflammatory cytokines, tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). This suggests that adropin may have an anti-inflammatory role in the liver and possibly in other organs, including the heart.
Interestingly, a recent meta-analysis found an association between serum adropin and coronary artery disease (CAD) [61]. The study stated that the serum adropin level in patients with CAD was lower than in healthy individuals, indicating that the decrease in adropin concentration might play an important role in the development of CAD. Another cardiovascular disease, which has been correlated with adropin, is atrial fibrillation. Atrial fibrillation is a condition of abnormal heart rhythm [62][63]. Decreased serum adropin concentrations were found in atrial fibrillation patients compared with healthy controls. Patients with chronic atrial fibrillation had a significantly reduced serum adropin concentration compared with control patients. Hence, adropin deficiency may contribute to the development and progression of atrial fibrillation [64] Table 1.

5. Inflammation

Researchers and scientists have also investigated the role of adropin in inflammation and related diseases such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease in response to injury of the arterial wall and the formation of plaque [38]. Vascular inflammation stimulates the expression of certain adhesion molecules, such as intercellular adhesion molecule 1, vascular cell adhesion molecule 1 in endothelial cells. These adhesion molecules stimulate monocyte adhesion to endothelial cells and monocyte infiltration into the subendothelial space, causing an accumulation of macrophage foam cells. Chronic inflammation of the cardiovascular system is a common feature of chronic diseases including diabetes mellitus, hyperlipidemia and nosocomial conditions [65][66][67]. Hormones such as adropin and plant-based antioxidants have been used to mitigate the adverse effects of these vascular lesions. Adropin is expressed in human endothelial cells [3], and it has been shown earlier that adropin inhibits tumor necrosis factor α (TNFα). Sato et al. investigated the relationship between TNFα, monocyte adhesion, human endothelial cells, atherosclerosis and adropin. The experiment involved incubating human endothelial cells with adropin and TNFα and assessing the expression of the adhesion molecules that are involved in atherosclerosis. The results showed that an incubation of adropin alone had no significant effect on the mRNA expression of these adhesion molecules, which are usually stimulated by TNFα. However, when adropin and TNFα were both incubated with human endothelial cells, adropin suppressed the TNFα-induced mRNA expression of adhesion molecules, suggesting a role for adropin in the anti-atherosclerosis process by inhibiting endothelial cells’ adhesion molecules via the suppression of TNFα [68].
In the case of obesity, the infiltration of macrophages into adipose tissues causes chronic inflammation. Adipocytes secrete cytokines such as TNFα and MCP-1 that attract macrophages and regulatory T cells, leading to fat inflammation. Adropin regulates the expression of PPAR-γ by activating the AKT pathway, thus inhibiting the differentiation of 3T3-L1 preadipocytes into mature adipocytes and consequently reducing fat accumulation and fat inflammation [69].
In another study, lower adropin plasma levels and increased inflammation markers such as TNFα and interleukin-6 (IL-6) were reported in male patients with moderate and severe obstructive sleep apnea, compared to healthy individuals [70].
Furthermore, in order to investigate how adropin could affect hepatocyte inflammation and injury in nonalcoholic steatohepatitis (NASH), immunohistochemistry using the inflammation markers F4/80, CD45 and MCP-1, and a gene expression analysis for TNFα and IL-6 genes, were performed using liver tissues from adropin knockout C57BL/6J mice and the control wild-type, which were fed a methionine-choline deficient diet [71]. Methionine-choline deficient diet is the classic dietary model for studying NASH, and usually, rodents consuming this diet develop steatohepatitis, necroinflammation, and fibrosis, similar to human NASH [36]. The pathohistological analysis showed a higher signal of F4/80, CD45 and MCP1 and a substantial induction of genes TNFα and IL-6 in adropin knockout mice. These results indicate the presence of elevated inflammatory responses in adropin knockout mice, when compared to that of the wild-type mice [71] (Table 1).

6. Cell Proliferation and Differentiation

Far beyond the classical action, adropin can stimulate cell proliferation and differentiation. Lovren et al. showed that adropin has the ability to induce the proliferation and capillary-like tube formation of endothelial cells that stimulate angiogenesis [3]. Adropin upregulated endothelial NO synthase expression through VEGFR2 2-PI3K-Akt and VEGFR2-extracellular signal-regulated kinase pathways to reduce inflammation.
Furthermore, in rat primary preadipocytes and 3T3-L1 cells, preadipocyte proliferation was increased by adropin treatment, while the differentiation of those preadipocytes into mature adipocytes was reduced [37]. The suppression of adipogenic markers and lipid accumulation demonstrate the important role of adropin in the fight against obesity [69]. The same research team reported a similar effect of adropin on primary brown preadipocytes that were isolated from the interscapular region in rat [72].
On the other hand, it was reported that adropin downregulated the proliferation and migration of human aortic smooth muscle cells (HASMCs) in vitro, providing evidence that the peptide is also protective against atherosclerosis [68] (Table 1). The role of adropin in different tissues and organ systems is depicted in Figure 1.
Figure 1. Graphical representation of the function of adropin in different body tissues.

References

  1. Gao, S.; McMillan, R.P.; Zhu, Q.; Lopaschuk, G.D.; Hulver, M.W.; Butler, A.A. Therapeutic effects of adropin on glucose tolerance and substrate utilization in diet-induced obese mice with insulin resistance. Mol. Metab. 2015, 4, 310–324.
  2. Kumar, K.G.; Trevaskis, J.L.; Lam, D.D.; Sutton, G.M.; Koza, R.A.; Chouljenko, V.N.; Kousoulas, K.G.; Rogers, P.M.; Kesterson, R.A.; Thearle, M.; et al. Identification of Adropin as a Secreted Factor Linking Dietary Macronutrient Intake with Energy Homeostasis and Lipid Metabolism. Cell Metab. 2008, 8, 468–481.
  3. Lovren, F.; Pan, Y.; Quan, A.; Singh, K.K.; Shukla, P.C.; Gupta, M.; Al-Omran, M.; Teoh, H.; Verma, S. Adropin is a novel regulator of endothelial function. Circulation 2010, 11 (Suppl. S1), 185–192.
  4. Demirdöğen, F.; Akdağ, T.; Gündüz, Z.B.; Odabaş, F.Ö. Investigation of serum adropin levels and its relationship with hypothalamic atrophy in patients with multiple sclerosis. Mult. Scler. Relat. Disord. 2022, 66, 103948.
  5. Aydın, P.; Uzunçakmak, S.K.; Tör, İ.H.; Bilen, A.; Özden, A. Comparison of Serum Adropin Levels in Patients with Diabetes Mellitus, COVID-19, and COVID-19 with Diabetes Mellitus. Eurasian J. Med. 2022, 54, 197–201.
  6. Ruszała, M.; Pilszyk, A.; Niebrzydowska, M.; Kimber-Trojnar, Ż.; Trojnar, M.; Leszczyńska-Gorzelak, B. Novel Biomolecules in the Pathogenesis of Gestational Diabetes Mellitus 2.0. Int. J. Mol. Sci. 2022, 23, 4364.
  7. Kong, Z.; Liu, Y. Soluble Vascular Adhesion Protein-1 Level Correlates With Adropin and Inflammatory Biomarkers in Patients With Obstructive Sleep Apnea. Ear Nose Throat J. 2022. Online ahead of print.
  8. Simac, P.; Perkovic, D.; Bozic, I.; Bilopavlovic, N.; Martinovic, D.; Bozic, J. Serum Adropin Levels in Patients with Rheumatoid Arthritis. Life 2022, 12, 169.
  9. Uysal, B.A.; Kuyumcu, M.S. Serum irisin and adropin levels may be predictors for coronary artery ectasia. Clin. Exp. Hypertens. 2022, 44, 223–227.
  10. Kurt, E.; Tekin, E.; Kurt, N.; Bayramoglu, A. The role of adropin, HIF-1α and apelin biomarkers in the diagnosis of acute mesentaric ischemia. Am. J. Emerg. Med. 2022, 51, 223–227.
  11. Es-Haghi, A.; Al-Abyadh, T.; Mehrad-Majd, H. The Clinical Value of Serum Adropin Level in Early Detection of Diabetic Nephropathy. Kidney Blood Press. Res. 2021, 46, 734–740.
  12. Zang, H.; Jiang, F.; Cheng, X.; Xu, H.; Hu, X. Serum adropin levels are decreased in chinese type 2 diabetic patients and negatively correlated with body mass index. Endocr. J. 2018, 65, 685–691.
  13. Chen, S.; Zeng, K.; Liu, Q.C.; Guo, Z.; Zhang, S.; Chen, X.R.; Lin, J.H.; Wen, J.P.; Zhao, C.F.; Lin, X.H.; et al. Adropin deficiency worsens HFD-induced metabolic defects. Cell Death Dis. 2017, 8, e3008.
  14. Li, N.; Xie, G.; Zhou, B.; Qu, A.; Meng, H.; Liu, J.; Wang, G. Serum Adropin as a Potential Biomarker for Predicting the Development of Type 2 Diabetes Mellitus in Individuals With Metabolic Dysfunction-Associated Fatty Liver Disease. Front. Physiol. 2021, 12, 696163.
  15. Polkowska, A.; Pasierowska, I.E.; Pasławska, M.; Pawluczuk, E.; Bossowski, A. Assessment of Serum Concentrations of Adropin, Afamin, and Neudesin in Children with Type 1 Diabetes. BioMed Res. Int. 2019, 2019, 6128410.
  16. Hosseini, A.; Shanaki, M.; Emamgholipour, S.; Nakhjavani, M.; Razi, F.; Golmohammadi, T. Elevated serum levels of adropin in patients with type 2 diabetes mellitus and its association with insulin resistance. J. Biol. Today’s World 2016, 5, 44–49.
  17. Li, S.; Sun, J.; Hu, W.; Liu, Y.; Lin, D.; Duan, H.; Liu, F. The association of serum and vitreous adropin concentrations with diabetic retinopathy. Ann. Clin. Biochem. Int. J. Lab. Med. 2019, 56, 253–258.
  18. Hu, W.; Chen, L. Association of Serum Adropin Concentrations with Diabetic Nephropathy. Mediat. Inflamm. 2016, 2016, 6038261.
  19. Beigi, A.; Shirzad, N.; Nikpour, F.; Nasli Esfahani, E.; Emamgholipour, S.; Bandarian, F. Association between serum adropin levels and gestational diabetes mellitus; a case–control study. Gynecol. Endocrinol. 2015, 31, 939–941.
  20. Kuo, F.Y.; Cheng, K.C.; Li, Y.; Cheng, J.T.; Tsai, C.C. Promotion of adropin expression by hyperglycemia is associated with STAT3 activation in diabetic rats. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 2269–2277.
  21. Chen, X.; Chen, S.; Shen, T.; Yang, W.; Chen, Q.; Zhang, P.; You, Y.; Sun, X.; Xu, H.; Tang, Y.; et al. Adropin regulates hepatic glucose production via PP2A/AMPK pathway in insulin-resistant hepatocytes. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020, 34, 10056–10072.
  22. Ozkan, A.; Aslan, M.A.; Sinen, O.; Munzuroglu, M.; Derin, N.; Parlak, H.; Bulbul, M.; Agar, A. Effects of adropin on learning and memory in rats tested in the Morris water maze. Hippocampus 2022, 32, 253–263.
  23. Puigserver, P.; Rhee, J.; Donovan, J.; Walkey, C.J.; Yoon, J.C.; Oriente, F.; Kitamura, Y.; Altomonte, J.; Dong, H.H.; Accili, D.; et al. Insulin-regulated hepatic gluconeogenesis through FOXO1–PGC-1a interaction. Nature 2003, 423, 545–550.
  24. Hill, M.J.; Suzuki, S.; Segars, J.H.; Kino, T. CRTC2 is a coactivator of GR and couples GR and CREB in the regulation of hepatic gluconeogenesis. Mol. Endocrinol. 2016, 30, 104–117.
  25. Thapa, D.; Xie, B.; Manning, J.R.; Zhang, M.; Stoner, M.W.; Huckestein, B.R.; Edmunds, L.R.; Zhang, X.; Dedousis, N.L.; O’Doherty, R.M.; et al. Adropin reduces blood glucose levels in mice by limiting hepatic glucose production. Physiol. Rep. 2019, 7, e14043.
  26. Kurir, T.T.; Miličević, T.; Novak, A.; Vilović, M.; Božić, J. Adropin—potential link in cardiovascular protection for obese male type 2 diabetes mellitus patients treated with liraglutide. Acta Clin. Croat. 2020, 59, 344–350.
  27. Tasyurek, H.M.; Altunbas, H.A.; Balci, M.K.; Sanlioglu, S. Incretins: Their physiology and application in the treatment of diabetes mellitus. Diabetes/Metab. Res. Rev. 2014, 30, 354–371.
  28. Lotfy, M.; Singh, J.; Kalász, H.; Tekes, K.; Adeghate, E. Medicinal Chemistry and Applications of Incretins and DPP-4 Inhibitors in the Treatment of Type 2 Diabetes Mellitus. Open Med. Chem. J. 2011, 5, 82–92.
  29. Adeghate, J.O.; D’Souza, C.; Kántor, O.; Tariq, S.; Souid, A.K.; Adeghate, E. Early (5-Day) Onset of Diabetes Mellitus Causes Degeneration of Photoreceptor Cells, Overexpression of Incretins, and Increased Cellular Bioenergetics in Rat Retina. Cells 2021, 10, 1981.
  30. Lotfy, M.; Singh, J.; Rashed, H.; Tariq, S.; Zilahi, E.; Adeghate, E. The effect of glucagon-like peptide-1 in the management of diabetes mellitus: Cellular and molecular mechanisms. Cell Tissue Res. 2014, 358, 343–358.
  31. Mahgoub, M.O.; D’Souza, C.; Al Darmaki, R.; Baniyas, M.; Adeghate, E. An update on the role of irisin in the regulation of endocrine and metabolic functions. Peptides 2018, 104, 15–23.
  32. Behera, J.; Ison, J.; Voor, M.J.; Tyagi, N. Exercise-Linked Skeletal Irisin Ameliorates Diabetes-Associated Osteoporosis by Inhibiting the Oxidative Damage-Dependent miR-150-FNDC5/Pyroptosis Axis. Diabetes 2022, db210573.
  33. Marrano, N.; Biondi, G.; Borrelli, A.; Cignarelli, A.; Perrini, S.; Laviola, L.; Giorgino, F.; Natalicchio, A. Irisin and incretin hormones: Similarities, differences, and implications in type 2 diabetes and obesity. Biomolecules 2021, 11, 286.
  34. Wattez, J.S.; Ravallec, R.; Cudennec, B.; Knauf, C.; Dhulster, P.; Valet, P.; Breton, C.; Vieau, D.; Lesage, J. Apelin stimulates both cholecystokinin and glucagon-like peptide 1 secretions in vitro and in vivo in rodents. Peptides 2013, 48, 134–136.
  35. Howarth, F.C.; Jacobson, M.; Shafiullah, M.; Adeghate, E. Long-term effects of type 2 diabetes mellitus on heart rhythm in the Goto-Kakizaki rat. Exp. Physiol. 2008, 93, 362–369.
  36. Marcolin, É.; Forgiarini, L.F.; Tieppo, J.; Dias, A.S.; de Freitas, L.A.R.; Marroni, N.P. Methionine- and choline-deficient diet induces hepatic changes characteristic of non-alcoholic steatohepatitis. Arq. De Gastroenterol. 2011, 48, 72–79.
  37. Jasaszwili, M.; Wojciechowicz, T.; Billert, M.; Strowski, M.Z.; Nowak, K.W.; Skrzypski, M. Effects of adropin on proliferation and differentiation of 3T3-L1 cells and rat primary preadipocytes. Mol. Cell. Endocrinol. 2019, 496, 110532.
  38. Hansson, G.K.; Libby, P. The immune response in atherosclerosis: A double-edged sword. Nat. Rev. Immunol. 2006, 6, 508–519.
  39. Jaberi, S.A.; Cohen, A.; Saeed, Z.; Ojha, S.; Singh, J.; Adeghate, E. Obesity: Molecular Mechanisms, Epidemiology, Complications and Pharmacotherapy. In Cellular and Biochemical Mechanisms of Obesity; Springer: Cham, Switzerland, 2021; pp. 249–266.
  40. Hart, D.A. Obesity, the Obesity Epidemic, and Metabolic Dysfunction: The Conundrum Presented by the Disconnect between Evolution and Modern Societies. J. Biomed. Sci. Eng. 2021, 14, 203–211.
  41. Ganesh-Kumar, K.; Zhang, J.; Gao, S.; Rossi, J.; McGuinness, O.P.; Halem, H.H.; Culler, M.D.; Mynatt, R.L.; Butler, A.A. Adropin deficiency is associated with increased adiposity and insulin resistance. Obesity 2012, 20, 1394–1402.
  42. Erman, H.; Ozdemir, A.; Sitar, M.E.; Cetin, S.I.; Boyuk, B. Role of serum adropin measurement in the assessment of insulin resistance in obesity. J. Investig. Med. Off. Publ. Am. Fed. Clin. Res. 2021, 69, 1318–1323.
  43. Butler, A.A.; Tam, C.S.; Stanhope, K.L.; Wolfe, B.M.; Ali, M.R.; O’Keeffe, M.; St-Onge, M.P.; Ravussin, E.; Havel, P.J. Low Circulating Adropin Concentrations with Obesity and Aging Correlate with Risk Factors for Metabolic Disease and Increase after Gastric Bypass Surgery in Humans. J. Clin. Endocrinol. Metab. 2012, 97, 3783–3791.
  44. Glück, M.; Glück, J.; Wiewióra, M.; Rogala, B.; Piecuch, J. Serum Irisin, Adropin, and Preptin in Obese Patients 6 Months After Bariatric Surgery. Obes. Surg. 2019, 29, 3334–3341.
  45. Kocaoglu, C.; Buyukinan, M.; Erdem, S.S.; Ozel, A. Are obesity and metabolic syndrome associated with plasma adropin levels in children? J. Pediatric Endocrinol. Metab. 2015, 28, 1293–1297.
  46. Adeghate, E.A.; Kalász, H.; Al Jaberi, S.; Adeghate, J.; Tekes, K. Tackling type 2 diabetes-associated cardiovascular and renal comorbidities: A key challenge for drug development. Expert Opin. Investig. Drugs 2021, 30, 85–93.
  47. Singh, R.M.; Waqar, T.; Howarth, F.C.; Adeghate, E.; Bidasee, K.; Singh, J. Hyperglycemia-induced cardiac contractile dysfunction in the diabetic heart. Heart Fail. Rev. 2018, 23, 37–54.
  48. Lotfy, M.; Adeghate, J.; Kalasz, H.; Singh, J.; Adeghate, E. Chronic Complications of Diabetes Mellitus: A Mini Review. Curr. Diabetes Rev. 2017, 13, 3–10.
  49. Adeghate, E.; Singh, J. Structural changes in the myocardium during diabetes-induced cardiomyopathy. Heart Fail. Rev. 2014, 19, 15–23.
  50. Adeghate, E.; Schattner, P.; Dunn, E. An update on the etiology and epidemiology of diabetes mellitus. Ann. N. Y. Acad. Sci. 2006, 1084, 1–29.
  51. Adeghate, E. Molecular and cellular basis of the aetiology and management of diabetic cardiomyopathy: A short review. Mol. Cell. Biochem. 2004, 261, 187–191.
  52. Aydin, S.; Kuloglu, T.; Aydin, S.; Eren, M.N.; Yilmaz, M.; Kalayci, M.; Sahin, I.; Kocaman, N.; Citil, C.; Kendir, Y. Expression of adropin in rat brain, cerebellum, kidneys, heart, liver, and pancreas in streptozotocin-induced diabetes. Mol. Cell. Biochem. 2013, 380, 73–81.
  53. Fillmore, N.; Mori, J.; Lopaschuk, G.D. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br. J. Pharmacol. 2014, 171, 2080–2090.
  54. Altamimi, T.R.; Gao, S.; Karwi, Q.G.; Fukushima, A.; Rawat, S.; Wagg, C.S.; Zhang, L.; Lopaschuk, G.D. Adropin regulates cardiac energy metabolism and improves cardiac function and efficiency. Metab. Clin. Exp. 2019, 98, 37–48.
  55. Thapa, D.; Xie, B.; Zhang, M.; Stoner, M.W.; Manning, J.R.; Huckestein, B.R.; Edmunds, L.R.; Mullett, S.J.; McTiernan, C.F.; Wendell, S.G.; et al. Adropin treatment restores cardiac glucose oxidation in pre-diabetic obese mice. J. Mol. Cell. Cardiol. 2019, 129, 174–178.
  56. Arkadievich, O.D. Metabolic markers of myocardium insulin resistance in dogs with heart failure. Open Vet. J. 2021, 10, 363–370.
  57. Thapa, D.; Xie, B.; Mushala, B.; Zhang, M.; Manning, J.R.; Bugga, P.; Stoner, M.W.; Jurczak, M.J.; Scott, I. Diet-induced obese mice are resistant to improvements in cardiac function resulting from short-term adropin treatment. Curr. Res. Physiol. 2022, 5, 55–62.
  58. Mushala, B.A.S.; Scott, I. Adropin: A hepatokine modulator of vascular function and cardiac fuel metabolism. Am. J. Physiol. Circ. Physiol. 2021, 320, H238–H244.
  59. Poss, J.; Custodis, F.; Werner, C.; Weingartner, O.; Bohm, M.; Laufs, U. Cardiovascular Disease and Dyslipidemia: Beyond LDL. Curr. Pharm. Des. 2011, 17, 861–870.
  60. Akcılar, R.; Emel Koçak, F.; Şimşek, H.; Akcılar, A.; Bayat, Z.; Ece, E.; Kökdaşgil, H. The effect of adropin on lipid and glucose metabolism in rats with hyperlipidemia. Iran. J. Basic Med. Sci. 2016, 19, 245–251.
  61. Zheng, J.; Liu, M.; Chen, L.; Yin, F.; Zhu, X.; Gou, J.; Zeng, W.; Lv, Z. Association between serum adropin level and coronary artery disease: A systematic review and meta-analysis. Cardiovasc. Diagn. Ther. 2019, 9, 1–7.
  62. Nattel, S. New ideas about atrial fibrillation 50 years on. Nature 2002, 415, 219–226.
  63. Sultan, A.; Jacobson, M.; Adeghate, E.; Oulhaj, A.; Shafiullah, M.; Qureshi, A.; Howarth, F.C. Effects of obesity and diabesity on heart rhythm in the Zucker rat. Clin. Exp. Pharmacol. Physiol. 2021, 48, 735–747.
  64. Wang, B.; Xue, Y.; Shang, F.; Ni, S.; Liu, X.; Fan, B.; Wang, H. Association of serum adropin with the presence of atrial fibrillation and atrial remodeling. J. Clin. Lab. Anal. 2019, 33, e22672.
  65. Meeran, M.; Al Taee, H.; Azimullah, S.; Tariq, S.; Adeghate, E.; Ojha, S. β-Caryophyllene, a natural bicyclic sesquiterpene attenuates doxorubicin-induced chronic cardiotoxicity via activation of myocardial cannabinoid type-2 (CB2) receptors in rats. Chem. Biol. Interact. 2019, 304, 158–167.
  66. Al-Taee, H.; Azimullah, S.; Meeran, M.; Alaraj Almheiri, M.K.; Al Jasmi, R.A.; Tariq, S.; Ab Khan, M.; Adeghate, E.; Ojha, S. β-caryophyllene, a dietary phytocannabinoid attenuates oxidative stress, inflammation, apoptosis and prevents structural alterations of the myocardium against doxorubicin-induced acute cardiotoxicity in rats: An in vitro and in vivo study. Eur. J. Pharmacol. 2019, 858, 172467.
  67. Mahgoub, E.; Kumaraswamy, S.M.; Kader, K.H.; Venkataraman, B.; Ojha, S.; Adeghate, E.; Rajesh, M. Genipin attenuates cisplatin-induced nephrotoxicity by counteracting oxidative stress, inflammation, and apoptosis. Biomed. Pharmacother. 2017, 93, 1083–1097.
  68. Sato, K.; Yamashita, T.; Shirai, R.; Shibata, K.; Okano, T.; Yamaguchi, M.; Mori, Y.; Hirano, T.; Watanabe, T. Adropin contributes to anti-atherosclerosis by suppressing monocyte-endothelial cell adhesion and smooth muscle cell proliferation. Int. J. Mol. Sci. 2018, 19, 1293.
  69. Jasaszwili, M.; Billert, M.; Strowski, M.Z.; Nowak, K.W.; Skrzypski, M. Adropin as A Fat-Burning Hormone with Multiple Functions-Review of a Decade of Research. Molecules 2020, 25, 549.
  70. Bozic, J.; Borovac, J.A.; Galic, T.; Kurir, T.T.; Supe-Domic, D.; Dogas, Z. Adropin and inflammation biomarker levels in male patients with obstructive sleep apnea: A link with glucose metabolism and sleep parameters. J. Clin. Sleep Med. 2018, 14, 1109–1118.
  71. Chen, X.; Xue, H.; Fang, W.; Chen, K.; Chen, S.; Yang, W.; Shen, T.; Chen, X.; Zhang, P.; Ling, W. Adropin protects against liver injury in nonalcoholic steatohepatitis via the Nrf2 mediated antioxidant capacity. Redox Biol. 2019, 21, 101068.
  72. Jasaszwili, M.; Wojciechowicz, T.; Strowski, M.Z.; Nowak, K.W.; Skrzypski, M. Adropin stimulates proliferation but suppresses differentiation in rat primary brown preadipocytes. Arch. Biochem. Biophys. 2020, 692, 108536.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 371
Revisions: 2 times (View History)
Update Date: 24 Aug 2023
1000/1000
Video Production Service