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 -- 1843 2022-05-04 00:04:05 |
2 Format Correct Meta information modification 1843 2022-05-05 10:29:56 | |
3 Format Correct Meta information modification 1843 2022-05-05 10:49:45 |

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.
Križančić Bombek, L.; Čater, M. Skeletal Muscle Uncoupling Proteins in Obesity Mice Models. Encyclopedia. Available online: https://encyclopedia.pub/entry/22586 (accessed on 20 April 2024).
Križančić Bombek L, Čater M. Skeletal Muscle Uncoupling Proteins in Obesity Mice Models. Encyclopedia. Available at: https://encyclopedia.pub/entry/22586. Accessed April 20, 2024.
Križančić Bombek, Lidija, Maša Čater. "Skeletal Muscle Uncoupling Proteins in Obesity Mice Models" Encyclopedia, https://encyclopedia.pub/entry/22586 (accessed April 20, 2024).
Križančić Bombek, L., & Čater, M. (2022, May 04). Skeletal Muscle Uncoupling Proteins in Obesity Mice Models. In Encyclopedia. https://encyclopedia.pub/entry/22586
Križančić Bombek, Lidija and Maša Čater. "Skeletal Muscle Uncoupling Proteins in Obesity Mice Models." Encyclopedia. Web. 04 May, 2022.
Skeletal Muscle Uncoupling Proteins in Obesity Mice Models
Edit

Obesity and accompanying type 2 diabetes are among major and increasing worldwide problems that occur fundamentally due to excessive energy intake during its expenditure. Endotherms continuously consume a certain amount of energy to maintain core body temperature via thermogenic processes, mainly in brown adipose tissue and skeletal muscle. Skeletal muscle glucose utilization and heat production are significant and directly linked to body glucose homeostasis at rest, and especially during physical activity. However, this glucose balance is impaired in diabetic and obese states in humans and mice, and manifests as glucose resistance and altered muscle cell metabolism. Uncoupling proteins have a significant role in converting electrochemical energy into thermal energy without ATP generation. Different homologs of uncoupling proteins were identified, and their roles were linked to antioxidative activity and boosting glucose and lipid metabolism. From this perspective, uncoupling proteins were studied in correlation to the pathogenesis of diabetes and obesity and their possible treatments. Mice were extensively used as model organisms to study the physiology and pathophysiology of energy homeostasis. However, researchers should be aware of interstrain differences in mice models of obesity regarding thermogenesis and insulin resistance in skeletal muscles. 

uncoupling protein skeletal muscle obesity mouse models UCP1 UCP2 UCP3 Sex Differences in UCP Expression

1. UCP Homologs and Their Roles

The role of UCPs in the pathogenesis of diabetes mellitus has recently become a popular topic since five homologs have been found in mammals[1]. Their structure is similar, but their distribution in different tissues varies considerably[2]. The physiological functions of UCPs have been studied intensively in the last three decades, yet they are still not completely elucidated. They are known for their antioxidative activity[3][4] and as glucose and lipid metabolism enhancers or regulators. Several gene polymorphisms of UCP1, UCP2, and UCP3 have been found in human diabetic and obese individuals, linking them to the development of glucose metabolism and insulin signaling pathologies[2] (Figure 1). In contrast to UCP1, which can represent as much as 10% of proteins in the inner mitochondrial membrane[5], UCP2 and UCP3 usually comprise less than 0.1% of the membrane protein content. They need specific activation for their proton transporting function[6].
Metabolites 12 00259 g002 550
Figure 1. UCP homologs are present in different tissues and have distinct roles. Arrows up represent an increased activity, arrows down represent a decreased activity (created with BioRender.com, (accessed on 19 January 2022)).

1.1. UCP1

Studies in rodents have shown that BAT starts to develop in the interscapular region during embryonic days E15–16 and that UCP1 mRNA expression increases around days E18–19 just before birth. The BAT continues to develop postnatally until between postnatal days P15–21 and remains present throughout adult life[7][8]. Recent research revealed the existence of two subpopulations of brown adipocytes in mice. One subpopulation has high thermogenic activity and high UCP1 expression, and the other has low thermogenic activity and low UCP1 expression[9]. At birth, all adipocytes express high levels of UCP1 and have a high thermogenic activity to meet newborns’ thermal requirements. Postnatally, some adipocytes begin to convert to the subpopulation with low UCP1 expression so that both subpopulations coexist in adult mice and might switch between each other during normal thermogenesis at room temperature. When exposed to cold, the transcription of genes in the subpopulation with the low UCP1 expression increases, thereby increasing the total thermogenic capacity of BAT[10]. During long-term cold exposure, de novo adipogenesis was observed in BAT[11][12]. In senescence, the capacity of adipocytes to increase UCP1 expression after cold exposure becomes impaired[10].
UCP1 mainly localizes to the inner mitochondrial membrane of BAT. Its proton conductance increases in elevated concentrations of long-chain free fatty acids (FFAs)[13] and is controlled by insulin[14][15]. Apart from BAT, recent studies also reported UCP1 expression in white adipose tissue, skeletal muscle, longitudinal smooth muscle layers, retinal cells, and Langerhans islet cells[2][16].
In skeletal muscle mitochondria, the expression of UCP1 reaches only 13% of the expression in BAT and increases the GDP-sensitive proton leak[17]. The roles of UCP1 are decreasing membrane potential, reducing reactive oxygen species (ROS) generation, increasing energy expenditure, and increasing nonshivering thermogenesis[18][19][20]. Compared to BAT, the ability of UCP1 in skeletal muscle to increase glutathione levels and reduce ROS production is far greater, suggesting different specific roles and possibly distinct mechanisms of UCP1 in both tissues[17]. Some research shows that diabetes and obesity development involve specific polymorphisms of the Ucp1 gene[21]. Mutations in Ucp1 affect the activity or expression of the UCP1 protein and reduce regulated or basal energy expenditure, resulting in altered pancreatic function and insulin secretion[22][23].

1.2. UCP2

UCP2 mRNA is expressed in many tissues, such as muscle, spleen, pancreas, kidney, central nervous system, and immune system. The UCP2 gene is already expressed during fetal life in murine skeletal muscle. Its expression increases immediately after birth, reaching a maximum on day 2, and steadily declines after that regardless of the lactating mother’s diet[24].
UCP2 is most widely present and highly expressed among UCPs in diabetic pancreatic beta-cells[25]; therefore, its involvement in diabetes development has been proposed. Its role in the pancreas as a negative regulator of insulin secretion has been studied intensively in ob/ob mice. The activation of UCP2 by ROS causes mitochondrial membrane proton leak, which reduces ATP synthesis in pancreatic β-cells and downregulates glucose-stimulated insulin secretion[26][27][28]. The ob/ob mice lacking UCP2 have increased ATP synthesis and glucose-stimulated insulin secretion from beta-cells in Langerhans islets[29][30]. DeSouza et al. (2007) used an antisense oligonucleotide to Ucp2 in ob/ob mice and Swiss mice with hyperlipidemic diet-induced obesity and diabetes to inhibit UCP2 expression, resulting in metabolic improvement[28]. Finally, results from a human study on ethnicity differences in UCP2 polymorphisms demonstrated that in Asians, the UCP2-866G/A polymorphism is protective against, while the UCP2 Ala55Val polymorphism is susceptible to, type 2 diabetes[31]. Similar traits might also exist in mice, but these have not been thoroughly researched yet.
One of the reported other roles of UCP2 is controlling immune cell activation by modulating MAPK pathways and mitochondrial ROS production[32][33]. Additionally, a neuroprotective role has been proposed. By regulating mitochondrial membrane potential, production of ROS, and calcium homeostasis, UCP2 modulates neuronal activity and inhibits cellular damage[34].

1.3. UCP3

UCP3 is expressed in skeletal muscle and BAT[35][36][37][38]. In BAT, UCP3 is almost one order of magnitude more abundant than in skeletal muscle or heart and is directly correlated with the abundance of UCP1[39]. The predominant isoform in skeletal muscle is UCP3, and its expression is highly skeletal-muscle-specific[40]. In mice, UCP3 mRNA levels were highest in skeletal muscle, followed by heart, white adipose tissue, and spleen, which was somewhat different than in rats, where the expression in tissues other than skeletal muscle was negligible[41].
UCP3 expression was almost undetectable in murine muscle tissue during fetal life. In contrast, its expression became noticeable soon after birth in response to suckling and lipid intake and steadily increased for 15 days. Interestingly, after 15 days of life, the UCP3 mRNA levels became dependent on dietary interventions. If lactating mice were fed regular high-carbohydrate chow, UCP3 expression levels in pups started to decrease, whereas if mothers were fed a high-fat diet, the levels of UCP3 expression in pups remained high[24]. Research shows that nutritional factors regulate UCP3 expression. Specifically, its expression is induced by elevated circulating FFAs, which is typical for fasting or starvation[24][42]. Pedraza et al. reported that the UCP3 expression in skeletal muscle is dramatically downregulated in lactating mice, and this effect is reversed with weaning. These changes come hand-in-hand with changes in circulating FFAs, which are reduced during lactation and return to normal after weaning[43].
Pancreatic beta cells also express UCP3[44], linking its role to energy expenditure, glucose metabolism, diabetes, and obesity[45][46]. Pancreatic UCP3 also affects insulin secretion but acts differently than UCP2[44]. In humans, the expression of the Ucp3 gene in skeletal muscle and pancreas of diabetic patients is decreased[47], suggesting Ucp3 involvement in the development of type 2 diabetes. Muscle UCP3 is also important in FFA metabolism. It protects mitochondria from oxidative stress induced by lipids and modulates insulin sensitivity[48], making it a potential player in type 2 diabetes development. UCP3 protein levels are upregulated when FFAs’ supply to the mitochondria exceeds their oxidative capacity and downregulates when oxidative capacity is improved.
The degradation of both UCP2 and UCP3 is very rapid[49], making their half-lives only approximately 30 min[50]. In comparison, the half-life of UCP1 is around 30 h[51]. The short half-lives of UCP2 and UCP3 enable rapid adjustments of their protein levels, which are needed when facing the rapidly changing metabolic needs and different rates of ROS production during mitochondrial oxidative processes. Because of this rapid degradation, the UCP2 protein level can decrease before the level of its mRNA drops[52]. It is crucial to consider this when evaluating data and drawing conclusions solely on mRNA expression.

1.4. Other UCPs

UCP4 and UCP5 are mainly expressed in the central nervous system, where they play roles in brain metabolism and thermoregulatory heat production and are therefore often named neuronal UCPs[53][54]. However, their expression has also been determined in skeletal muscle, controlling energy expenditure and lipid oxidation. UCP5 is expressed in human skeletal muscle in three different isoforms, with UCP5L being the most abundant isoform, followed by UCP5S and UCP5SI[55]. UCP4 and UCP5 have a similar role in the protection against oxidative stress and mitochondrial dysfunction as other homologs[56]. High levels of UCP5 mRNA have been detected in testes and lower levels in the kidneys and liver[55].

2. Sex Differences in UCP Expression

Studies with rodents of both genders have shown significant sex-associated differences in the regulation of UCPs, which occur due to sex hormones and other distinct gender-based biological functions[57]. Sex hormone receptors are localized in the mitochondria of specific cells and can affect mitochondrial physiology[58]. In rodents, sex hormones influence different features of skeletal muscle, such as fiber diameter and myosin heavy-chain expression[59]. They also regulate UCP1 expression in brown adipocytes[60][61].
Age plays a vital role in the sex dimorphism of UCP expression. In prepubertal age in mice, UCPs are expressed at similar levels in both sexes, with significant differences, especially in UCP1 and UCP3 expression, being observed only later in adulthood. Expression of these proteins decreased with time in adult males, while in females, UCP1 and UCP3 expression decreased during young adulthood and increased later[62]. This age-dependent UCPs expression pattern correlates with weight gain. In several studies, weight gain with aging was more significant in males than in female mice, which showed a slight increase in body weight with senescence. This finding suggests that upregulation of UCP1 and UCP3 in BAT helps female mice avoid triglyceride accumulation in skeletal muscle and prevents obesity development[45][62][63].
Caloric diet feeding causes different overweight-induced expression of UCP3 in muscle and UCP1 in BAT in males than in females. Females tend to have a higher capacity to store fat when food is in excess than males, resulting in weight gain[64]. On the other hand, experiments with fasting showed interesting sex-dependent differences in UCP expression. Bazhan et al. (2019) studied sex asymmetry in the fasting effects on the transcription of the Ucp3 gene in muscle. A significant upregulation of muscle Ucp3 occurred in females after fasting for 24 h, while these changes were much less evident in males[65].

References

  1. Stefan Krauss; Chen-Yu Zhang; Bradford B. Lowell; The mitochondrial uncoupling-protein homologues. Nature Reviews Molecular Cell Biology 2005, 6, 248-261, 10.1038/nrm1592.
  2. Jing Liu; Ji Li; Wen-Jian Li; Chun-Ming Wang; The Role of Uncoupling Proteins in Diabetes Mellitus. Journal of Diabetes Research 2013, 2013, 1-7, 10.1155/2013/585897.
  3. Ryan J. Mailloux; Mary-Ellen Harper; Uncoupling proteins and the control of mitochondrial reactive oxygen species production. Free Radical Biology and Medicine 2011, 51, 1106-1115, 10.1016/j.freeradbiomed.2011.06.022.
  4. Petr Ježek; Blanka Holendová; Keith D. Garlid; Martin Jabůrek; Mitochondrial Uncoupling Proteins: Subtle Regulators of Cellular Redox Signaling. Antioxidants & Redox Signaling 2018, 29, 667-714, 10.1089/ars.2017.7225.
  5. Barbara Cannon; Jan Nedergaard; Brown Adipose Tissue: Function and Physiological Significance. Physiological Reviews 2004, 84, 277-359, 10.1152/physrev.00015.2003.
  6. Telma Cristina Esteves; Martin D. Brand; The reactions catalysed by the mitochondrial uncoupling proteins UCP2 and UCP3. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms 2005, 1709, 35-44, 10.1016/j.bbabio.2005.06.002.
  7. Marta Giralt; Immaculada Martin; Roser Iglesias; Octavi Vinas; Francesc Villarroya; Teresa Mampel; Ontogeny and perinatal modulation of gene expression in rat brown adipose tissue. Unaltered iodothyronine 5'-deiodinase activity is necessary for the response to environmental temperature at birth. JBIC Journal of Biological Inorganic Chemistry 1990, 193, 297-302, 10.1111/j.1432-1033.1990.tb19336.x.
  8. Josef Houštěk; Jan Kopecky; Zdeněk Rychter; Tomáš Soukup; Uncoupling protein in embryonic brown adipose tissue — existence of nonthermogenic and thermogenic mitochondria. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms 1988, 935, 19-25, 10.1016/0005-2728(88)90103-x.
  9. Abhijit Babaji Shinde; Anying Song; Qiong A. Wang; Brown Adipose Tissue Heterogeneity, Energy Metabolism, and Beyond. Frontiers in Endocrinology 2021, 12, 651763, 10.3389/fendo.2021.651763.
  10. Anying Song; Wenting Dai; Min Jee Jang; Leonard Medrano; Zhuo Li; Hu Zhao; Mengle Shao; Jiayi Tan; Aimin Li; Tinglu Ning; et al.Marcia M. MillerBrian ArmstrongJanice M. HussYi ZhuYong LiuViviana GradinaruXiwei WuLei JiangPhilipp E. SchererQiong A. Wang Low- and high-thermogenic brown adipocyte subpopulations coexist in murine adipose tissue. Journal of Clinical Investigation 2019, 130, 247-257, 10.1172/jci129167.
  11. L. J. Bukowiecki; A. Géloën; A. J. Collet; Proliferation and differentiation of brown adipocytes from interstitial cells during cold acclimation. American Journal of Physiology-Cell Physiology 1986, 250, C880-C887, 10.1152/ajpcell.1986.250.6.c880.
  12. Yun‐Hee Lee; Anelia P. Petkova; Anish A. Konkar; James G. Granneman; Cellular origins of cold-induced brown adipocytes in adult mice. The FASEB Journal 2014, 29, 286-299, 10.1096/fj.14-263038.
  13. Andriy Fedorenko; Polina V. Lishko; Yuriy Kirichok; Mechanism of Fatty-Acid-Dependent UCP1 Uncoupling in Brown Fat Mitochondria. Cell 2012, 151, 400-413, 10.1016/j.cell.2012.09.010.
  14. R Burcelin; J Kande; D Ricquier; J Girard; Changes in uncoupling protein and GLUT4 glucose transporter expressions in interscapular brown adipose tissue of diabetic rats: relative roles of hyperglycaemia and hypoinsulinaemia. Biochemical Journal 1993, 291, 109-113, 10.1042/bj2910109.
  15. Matthias J. Betz; Sven Enerbäck; Targeting thermogenesis in brown fat and muscle to treat obesity and metabolic disease. Nature Reviews Endocrinology 2017, 14, 77-87, 10.1038/nrendo.2017.132.
  16. Michèle M Sale; Fang-Chi Hsu; Nicholette D Palmer; Candace J Gordon; Keith L Keene; Hermina M Borgerink; Arun J Sharma; Richard N Bergman; Kent D Taylor; Mohammed F Saad; et al.Jill M Norris The uncoupling protein 1 gene, UCP1, is expressed in mammalian islet cells and associated with acute insulin response to glucose in African American families from the IRAS Family Study. BMC Endocrine Disorders 2007, 7, 1-1, 10.1186/1472-6823-7-1.
  17. Adjeitey CN, Mailloux RJ, Dekemp RA, Harper ME.; Mitochondrial uncoupling in skeletal muscle by UCP1 augments energy expenditure and glutathione content while mitigating ROS production.. Am J Physiol Endocrinol Metab 2013, 305(3), E405-E415, doi:10.1152/ajpendo.00057.2013.
  18. ite Gaudry MJ, Campbell KL, Jastroch M; Evolution of UCP1. Handb Exp Pharmacol 2019, 251, 127-141, doi:10.1007/164_2018_116.
  19. Janne Orava; Pirjo Nuutila; Martin E. Lidell; Vesa Oikonen; Tommi Noponen; Tapio Viljanen; Mika Scheinin; Markku Taittonen; Tarja Niemi; Sven Enerbäck; et al.Kirsi A. Virtanen Different Metabolic Responses of Human Brown Adipose Tissue to Activation by Cold and Insulin. Cell Metabolism 2011, 14, 272-279, 10.1016/j.cmet.2011.06.012.
  20. Edward T. Chouchani; Lawrence Kazak; Bruce M. Spiegelman; New Advances in Adaptive Thermogenesis: UCP1 and Beyond. Cell Metabolism 2018, 29, 27-37, 10.1016/j.cmet.2018.11.002.
  21. Jun-Jing Jia; Yun-Bo Tian; Zhen-Hui Cao; Lin-Li Tao; Xi Zhang; Si-Zhen Gao; Chang-Rong Ge; Qiu-Ye Lin; M. Jois; The polymorphisms of UCP1 genes associated with fat metabolism, obesity and diabetes. Molecular Biology Reports 2009, 37, 1513-1522, 10.1007/s11033-009-9550-2.
  22. A Hamann; J Tafel; B Büsing; H Münzberg; A Hinney; H Mayer; W Siegfried; D Ricquier; H Greten; J Hebebrand; et al.S Matthaei Analysis of the uncoupling protein-1 (UCP1) gene in obese and lean subjects: Identification of four amino acid variants. International Journal of Obesity 1998, 22, 939-941, 10.1038/sj.ijo.0800725.
  23. Kil Soo Kim; Dae-Yeon Cho; Young Joo Kim; Sun Mi Choi; Jong Yeol Kim; Seung Uoo Shin; Yoo Sik Yoon; The finding of new genetic polymorphism of UCP-1 A-1766G and its effects on body fat accumulation. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 2005, 1741, 149-155, 10.1016/j.bbadis.2004.11.026.
  24. S Brun; M C Carmona; T Mampel; O Viñas; M Giralt; R Iglesias; F Villarroya; Uncoupling protein-3 gene expression in skeletal muscle during development is regulated by nutritional factors that alter circulating non-esterified fatty acids.. FEBS Letters 1999, 453, 205-209, 10.1016/s0014-5793(99)00722-x.
  25. Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, Hagen T, Vidal-Puig AJ, Boss O, Kim YB, Zheng XX, Wheeler MB, Shulman GI, Chan CB, Lowell BB; Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction, and type 2 diabetes. Cell 2001, 105(6), 745-755, doi:10.1016/s0092-8674(01)00378-6.
  26. Stefan Krauss; Chen-Yu Zhang; Luca Scorrano; Louise T. Dalgaard; Julie St-Pierre; Shane T. Grey; Bradford B. Lowell; Superoxide-mediated activation of uncoupling protein 2 causes pancreatic β cell dysfunction. Journal of Clinical Investigation 2003, 112, 1831-1842, 10.1172/jci19774.
  27. Zhongmin Alex Ma; Zhengshan Zhao; John Turk; Mitochondrial Dysfunction and β-Cell Failure in Type 2 Diabetes Mellitus. Experimental Diabetes Research 2011, 2012, 1-11, 10.1155/2012/703538.
  28. Cláudio T. De Souza; Eliana P. Araújo; Luiz F. Stoppiglia; José R. Pauli; Eduardo Ropelle; Silvana A. Rocco; Rodrigo M. Marin; Kleber G. Franchini; José B. Carvalheira; Mário J. Saad; et al.Antonio C. BoscheroEverardo M. CarneiroLício A. Velloso Inhibition of UCP2 expression reverses diet-induced diabetes mellitus by effects on both insulin secretion and action. The FASEB Journal 2007, 21, 1153-1163, 10.1096/fj.06-7148com.
  29. Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, Hagen T, Vidal-Puig AJ, Boss O, Kim YB, Zheng XX, Wheeler MB, Shulman GI, Chan CB, Lowell BB; Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction, and type 2 diabetes. Cell 2001, 105(6), 745-55, doi: 10.1016/s0092-8674(01)00378-6.
  30. Christine A. Robson-Doucette; Sobia Sultan; Emma M. Allister; Jakob D. Wikstrom; Vasilij Koshkin; Alpana Bhattacharjee; Kacey J. Prentice; Samuel B. Sereda; Orian S. Shirihai; Michael B. Wheeler; et al. Beta-Cell Uncoupling Protein 2 Regulates Reactive Oxygen Species Production, Which Influences Both Insulin and Glucagon Secretion. Diabetes 2011, 60, 2710-2719, 10.2337/db11-0132.
  31. Rong Huang; Tingting Cai; Yunting Zhou; Yuming Wang; Huiying Wang; Ziyang Shen; Wenqing Xia; Xiaomei Liu; Bo Ding; Yong Luo; et al.Rengna YanHuiqin LiJindan WuJianhua Ma Ethnicity Differences in the Association of UCP1-3826A/G, UCP2-866G/A and Ala55Val, and UCP3-55C/T Polymorphisms with Type 2 Diabetes Mellitus Susceptibility: An Updated Meta-Analysis. BioMed Research International 2021, 2021, 1-14, 10.1155/2021/3482879.
  32. Yalin Emre; Tobias Nübel; Uncoupling protein UCP2: When mitochondrial activity meets immunity. FEBS Letters 2010, 584, 1437-1442, 10.1016/j.febslet.2010.03.014.
  33. Sabrina Diano; Tamas L. Horvath; Mitochondrial uncoupling protein 2 (UCP2) in glucose and lipid metabolism. Trends in Molecular Medicine 2012, 18, 52-58, 10.1016/j.molmed.2011.08.003.
  34. P Andy Li Suresh L Mehta; P. Andy Li; Neuroprotective Role of Mitochondrial Uncoupling Protein 2 in Cerebral Stroke. British Journal of Pharmacology 2009, 29, 1069-1078, 10.1038/jcbfm.2009.4.
  35. Olivier Boss; Sonia Samec; Ariane Paoloni-Giacobino; Colette Rossier; Abdul Dulloo; Josiane Seydoux; Patrick Muzzin; Jean-Paul Giacobino; Uncoupling protein-3: a new member of the mitochondrial carrier family with tissue-specific expression. FEBS Letters 1997, 408, 39-42, 10.1016/s0014-5793(97)00384-0.
  36. Antonio Vidal-Puig; Gemma Solanes; Danica Grujic; Jeffrey S. Flier; Bradford B. Lowell; UCP3: An Uncoupling Protein Homologue Expressed Preferentially and Abundantly in Skeletal Muscle and Brown Adipose Tissue. Biochemical and Biophysical Research Communications 1997, 235, 79-82, 10.1006/bbrc.1997.6740.
  37. Da-Wei Gong; Yufang He; Michael Karas; Marc Reitman; Uncoupling Protein-3 Is a Mediator of Thermogenesis Regulated by Thyroid Hormone, beta3-Adrenergic Agonists, and Leptin. Journal of Biological Chemistry 1997, 272, 24129-24132, 10.1074/jbc.272.39.24129.
  38. Antonio Vidal-Puig; Danica Grujic; Chen-Yu Zhang; Thilo Hagen; Olivier Boss; Yasuo Ido; Alicja Szczepanik; Jennifer Wade; Vamsi Mootha; Ronald Cortright; et al.Deborah MuoioBradford B. Lowell Energy Metabolism in Uncoupling Protein 3 Gene Knockout Mice. Journal of Biological Chemistry 2000, 275, 16258-16266, 10.1074/jbc.m910179199.
  39. Karolina E. Hilse; Anastasia V. Kalinovich; Anne Rupprecht; Alina Smorodchenko; Ute Zeitz; Katrin Staniek; Reinhold G. Erben; Elena E. Pohl; The expression of UCP3 directly correlates to UCP1 abundance in brown adipose tissue. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2015, 1857, 72-78, 10.1016/j.bbabio.2015.10.011.
  40. Vian Azzu; Martin Jastroch; Ajit S. Divakaruni; Martin D. Brand; The regulation and turnover of mitochondrial uncoupling proteins. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms 2010, 1797, 785-791, 10.1016/j.bbabio.2010.02.035.
  41. Lukáš Alán; Katarína Smolková; Eva Kronusová; Jitka Šantorová; Petr Ježek; Absolute levels of transcripts for mitochondrial uncoupling proteins UCP2, UCP3, UCP4, and UCP5 show different patterns in rat and mice tissues. Journal of Bioenergetics and Biomembranes 2009, 41, 71-78, 10.1007/s10863-009-9201-2.
  42. D. S. Weigle; L. E. Selfridge; M. W. Schwartz; R. J. Seeley; D. E. Cummings; P. J. Havel; J. L. Kuijper; H. BeltrandelRio; Elevated free fatty acids induce uncoupling protein 3 expression in muscle: a potential explanation for the effect of fasting. Diabetes 1998, 47, 298-302, 10.2337/diabetes.47.2.298.
  43. N Pedraza; G Solanes; M C Carmona; R Iglesias; O Viñas; T Mampel; M Vazquez; M Giralt; F Villarroya; Impaired expression of the uncoupling protein-3 gene in skeletal muscle during lactation: fibrates and troglitazone reverse lactation-induced downregulation of the uncoupling protein-3 gene.. Diabetes 2000, 49, 1224-1230, 10.2337/diabetes.49.7.1224.
  44. Yunfeng Li; Kathrin Maedler; Luan Shu; Leena Haataja; UCP-2 and UCP-3 Proteins Are Differentially Regulated in Pancreatic Beta-Cells. PLoS ONE 2008, 3, e1397, 10.1371/journal.pone.0001397.
  45. Sheila R. Costford; Shehla N. Chaudhry; Sean A. Crawford; Mahmoud Salkhordeh; Mary-Ellen Harper; Long-term high-fat feeding induces greater fat storage in mice lacking UCP3. American Journal of Physiology-Endocrinology and Metabolism 2008, 295, E1018-E1024, 10.1152/ajpendo.00779.2007.
  46. Graham P. Holloway; Swati S. Jain; Veronic Bezaire; Xiao Xia Han; Jan F. C. Glatz; Joost J. F. P. Luiken; Mary-Ellen Harper; Arend Bonen; FAT/CD36-null mice reveal that mitochondrial FAT/CD36 is required to upregulate mitochondrial fatty acid oxidation in contracting muscle. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 2009, 297, R960-R967, 10.1152/ajpregu.91021.2008.
  47. Patrick Schrauwen; Marco Mensink; Gert Schaart; Esther Moonen-Kornips; Jean-Pierre Sels; Ellen E. Blaak; Aaron P. Russell; Matthijs K. C. Hesselink; Reduced Skeletal Muscle Uncoupling Protein-3 Content in Prediabetic Subjects and Type 2 Diabetic Patients: Restoration by Rosiglitazone Treatment. The Journal of Clinical Endocrinology & Metabolism 2006, 91, 1520-1525, 10.1210/jc.2005-1572.
  48. Miranda Nabben; Joris Hoeks; Mitochondrial uncoupling protein 3 and its role in cardiac- and skeletal muscle metabolism. Physiology & Behavior 2008, 94, 259-269, 10.1016/j.physbeh.2007.11.039.
  49. Vian Azzu; Martin D. Brand; The on-off switches of the mitochondrial uncoupling proteins. Trends in Biochemical Sciences 2010, 35, 298-307, 10.1016/j.tibs.2009.11.001.
  50. Sophie Rousset; Julien Mozo; Geneviève Dujardin; Yalin Emre; Sandrine Masscheleyn; Daniel Ricquier; Anne-Marie Cassard-Doulcier; UCP2 is a mitochondrial transporter with an unusual very short half-life. FEBS Letters 2007, 581, 479-482, 10.1016/j.febslet.2007.01.010.
  51. P Puigserver; David Herron; M Gianotti; A Palou; Barbara Cannon; Jan Nedergaard; Induction and degradation of the uncoupling protein thermogenin in brown adipocytes in vitro and in vivo. Evidence for a rapidly degradable pool. Biochemical Journal 1992, 284, 393-398, 10.1042/bj2840393.
  52. Claire Pecqueur; Marie-Clotilde Alves-Guerra; Chantal Gelly; Corinne Lévi-Meyrueis; Elodie Couplan; Sheila Collins; Daniel Ricquier; Frederic Bouillaud; Bruno Miroux; Uncoupling Protein 2, in Vivo Distribution, Induction upon Oxidative Stress, and Evidence for Translational Regulation. Journal of Biological Chemistry 2001, 276, 8705-8712, 10.1074/jbc.m006938200.
  53. Daniel Sanchis; Christophe Fleury; Nathalie Chomiki; Marc Goubern; Quinling Huang; Maria Neverova; Francine Grégoire; Juliet Easlick; Serge Raimbault; Corinne Lévi-Meyrueis; et al.Bruno MirouxSheila CollinsMichael SeldinDenis RichardCraig WardenFrédéric BouillaudDaniel Ricquier BMCP1, a Novel Mitochondrial Carrier with High Expression in the Central Nervous System of Humans and Rodents, and Respiration Uncoupling Activity in Recombinant Yeast. Journal of Biological Chemistry 1998, 273, 34611-34615, 10.1074/jbc.273.51.34611.
  54. Weiguang Mao; Xing Xian Yu; Alan Zhong; Wenlu Li; Jennifer Brush; Steven W Sherwood; Sean H Adams; Guohua Pan; UCP4, a novel brain-specific mitochondrial protein that reduces membrane potential in mammalian cells. FEBS Letters 1999, 443, 326-330, 10.1016/s0014-5793(98)01713-x.
  55. Xiaolin Yang; Richard E. Pratley; Stephen Tokraks; P. Antonio Tataranni; Paska A. Permana; UCP5/BMCP1 transcript isoforms in human skeletal muscle: relationship of the short-insert isoform with lipid oxidation and resting metabolic rates. Molecular Genetics and Metabolism 2002, 75, 369-373, 10.1016/s1096-7192(02)00008-2.
  56. David B. Ramsden; Philip W.‐L. Ho; Jessica W.‐M. Ho; Hui‐Fang Liu; Danny H.‐F. So; Ho‐Man Tse; Koon‐Ho Chan; Shu‐Leong Ho; Human neuronal uncoupling proteins 4 and 5 (UCP4 and UCP5): structural properties, regulation, and physiological role in protection against oxidative stress and mitochondrial dysfunction. Brain and Behavior 2012, 2, 468-478, 10.1002/brb3.55.
  57. A M Rodríguez; A Palou; Uncoupling proteins: gender dependence and their relation to body weight control. International Journal of Obesity 2004, 28, 500-502, 10.1038/sj.ijo.0802588.
  58. Shao-Hua Yang; Ran Liu; Evelyn J. Perez; Yi Wen; Stanley M. Stevens; Thomas Valencia; Anne-Marie Brun-Zinkernagel; Laszlo Prokai; Yvonne Will; James Dykens; et al.Peter KoulenJames W. Simpkins Mitochondrial localization of estrogen receptor beta. Proceedings of the National Academy of Sciences 2004, 101, 4130-4135, 10.1073/pnas.0306948101.
  59. Jane M. Eason; Gail A. Schwartz; Grace K. Pavlath; Arthur W. English; Sexually dimorphic expression of myosin heavy chains in the adult mouse masseter. Journal of Applied Physiology 2000, 89, 251-258, 10.1152/jappl.2000.89.1.251.
  60. A. M. Rodríguez; M. Monjo; P. Roca; A. Palou; Opposite actions of testosterone and progesterone on UCP1 mRNA expression in cultured brown adipocytes. Cellular and Molecular Life Sciences 2002, 59, 1714-1723, 10.1007/pl00012499.
  61. Adamo Valle; Francisco García-Palmer; Jordi Oliver Oliver; Pilar Roca; Sex Differences in Brown Adipose Tissue Thermogenic Features During Caloric Restriction. Cellular Physiology and Biochemistry 2007, 19, 195-204, 10.1159/000099207.
  62. Michael Moschinger; Karolina E. Hilse; Anne Rupprecht; Ute Zeitz; Reinhold G. Erben; Thomas Rülicke; Elena E. Pohl; Age-related sex differences in the expression of important disease-linked mitochondrial proteins in mice. Biology of Sex Differences 2019, 10, 1-10, 10.1186/s13293-019-0267-1.
  63. Céline Aguer; Oliver Fiehn; Erin L. Seifert; Véronic Bézaire; John K. Meissen; Amanda Daniels; Kyle Scott; Jean‐Marc Renaud; Marta Padilla; David R. Bickel; et al.Michael DysartSean H. AdamsMary‐Ellen Harper Muscle uncoupling protein 3 overexpression mimics endurance training and reduces circulating biomarkers of incomplete β‐oxidation. The FASEB Journal 2013, 27, 4213-4225, 10.1096/fj.13-234302.
  64. A M Rodríguez; A Palou; Uncoupling proteins: gender-dependence and their relation to body weight control. International Journal of Obesity 2004, 28, 327-329, 10.1038/sj.ijo.0802579.
  65. Nadezhda Bazhan; Tatiana Jakovleva; Natalia Feofanova; Elena Denisova; Anastasia Dubinina; Natalia Sitnikova; Elena Makarova; Sex Differences in Liver, Adipose Tissue, and Muscle Transcriptional Response to Fasting and Refeeding in Mice. Cells 2019, 8, 1529, 10.3390/cells8121529.
  66. A M Rodríguez; A Palou; Uncoupling proteins: gender-dependence and their relation to body weight control. International Journal of Obesity 2004, 28, 327-329, 10.1038/sj.ijo.0802579.
  67. Nadezhda Bazhan; Tatiana Jakovleva; Natalia Feofanova; Elena Denisova; Anastasia Dubinina; Natalia Sitnikova; Elena Makarova; Sex Differences in Liver, Adipose Tissue, and Muscle Transcriptional Response to Fasting and Refeeding in Mice. Cells 2019, 8, 1529, 10.3390/cells8121529.
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: 797
Revisions: 3 times (View History)
Update Date: 05 May 2022
1000/1000