You are on page 1of 11

1. Stumvoll, M., Goldstein, B. J., & Van Haeften, T. W. (2005).

Type 2 diabetes:
principles of pathogenesis and therapy. The Lancet, 365(9467), 1333-1346.

2. Bowie, D. (2018). Polyamine-mediated channel block of ionotropic glutamate


receptors and its regulation by auxiliary proteins. Journal of Biological
Chemistry, 293(48), 18789-18802.

3. Martens-Lobenhoffer, J., Postel, S., Tröger, U., & Bode-Böger, S. M. (2007).


Determination of ornithine in human plasma by hydrophilic interaction
chromatography–tandem mass spectrometry. Journal of Chromatography B, 855(2),
271-275.

4. Sun, H., Saeedi, P., Karuranga, S., Pinkepank, M., Ogurtsova, K., Duncan, B. B., &
Magliano, D. J. (2022). IDF Diabetes Atlas: Global, regional and country-level diabetes
prevalence estimates for 2021 and projections for 2045. Diabetes research and clinical
practice, 183, 109119.
5. Valdés, S., Rojo-Martínez, G., & Soriguer, F. (2007). Evolución de la prevalencia de
la diabetes tipo 2 en población adulta española. Medicina clínica, 129(9), 352-355.

6. Rodríguez Panós, B., Sanchos, C., García Gosálvez, F., Divisón, J. A., Artigao, L.
M., López Abril, J., & Puras, A. (2000). The prevalence of diabetes mellitus and its
asciation with other cardiovascular risk factors in the province of Albacete. The
vasculatr disease Group of Albacete (GEVA). Atención Primaria, 125, 166-171.
7. Abela, L., Simmons, L., Steindl, K., Schmitt, B., Mastrangelo, M., Joset, P., ... &
Plecko, B. (2016). N8-acetylspermidine as a potential plasma biomarker for Snyder-
Robinson syndrome identified by clinical metabolomics. Journal of inherited metabolic
disease, 39(1), 131-137.
8. Bachmann, A. S., & Geerts, D. (2018). Polyamine synthesis as a target of MYC
oncogenes. Journal of Biological Chemistry, 293(48), 18757-18769.
9. Weinstein, A. R., Sesso, H. D., Lee, I. M., Cook, N. R., Manson, J. E., Buring, J. E.,
& Gaziano, J. M. (2004). Relationship of physical activity vs body mass index with type
2 diabetes in women. Jama, 292(10), 1188-1194.
10. Lynch, J., Helmrich, S. P., Lakka, T. A., Kaplan, G. A., Cohen, R. D., Salonen, R.,
& Salonen, J. T. (1996). Moderately intense physical activities and high levels of
cardiorespiratory fitness reduce the risk of non-insulin-dependent diabetes mellitus in
middle-aged men. Archives of internal medicine, 156(12), 1307-1314.

11. Parsamanesh, N., Moossavi, M., Bahrami, A., Butler, A. E., & Sahebkar, A. (2018).
Therapeutic potential of curcumin in diabetic complications. Pharmacological
Research, 136, 181-193.

12. Bunney, P. E., Zink, A. N., Holm, A. A., Billington, C. J., & Kotz, C. M. (2017).
Orexin activation counteracts decreases in nonexercise activity thermogenesis (NEAT)
caused by high-fat diet. Physiology & behavior, 176, 139-14810.
13. Galicia-Garcia, U., Benito-Vicente, A., Jebari, S., Larrea-Sebal, A., Siddiqi, H.,
Uribe, K. B., ... & Martín, C. (2020). Pathophysiology of type 2 diabetes mellitus.
International journal of molecular sciences, 21(17), 6275.

14. Boland, B. B., Rhodes, C. J., & Grimsby, J. S. (2017). The dynamic plasticity of
insulin production in β-cells. Molecular metabolism, 6(9), 958-973.

15. Fu, Z., R Gilbert, E., & Liu, D. (2013). Regulation of insulin synthesis and secretion
and pancreatic Beta-cell dysfunction in diabetes. Current diabetes reviews, 9(1), 25-53.

16. Rorsman, P., & Ashcroft, F. M. (2018). Pancreatic β-cell electrical activity and
insulin secretion: of mice and men. Physiological reviews, 98(1), 117-214

17. Islam, M. S. (2002). The ryanodine receptor calcium channel of β-cells: molecular
regulation and physiological significance. Diabetes, 51(5), 1299-1309.

18. Simon, J., Webb, T. E., King, B. F., Burnstock, G., & Barnard, E. A. (1995).
Characterisation of a recombinant P2Y purinoceptor. European Journal of
Pharmacology: Molecular Pharmacology, 291(3), 281-289.

19. Valera, S., Hussy, N., Evans, R. J., Adami, N., North, R. A., Surprenant, A., &
Buell, G. (1994). A new class of ligand-gated ion channel defined by P2x receptor for
extracellular ATP. Nature, 371(6497), 516-519

20. Christensen, A. A., & Gannon, M. (2019). The beta cell in type 2 diabetes. Current
diabetes reports, 19(9), 1-8.

21. Halban, P. A., Polonsky, K. S., Bowden, D. W., Hawkins, M. A., Ling, C., Mather,
K. J., & Weir, G. C. (2014). β-cell failure in type 2 diabetes: postulated mechanisms and
prospects for prevention and treatment. The Journal of Clinical Endocrinology &
Metabolism, 99(6), 1983-1992.

22. Yamamoto, W. R., Bone, R. N., Sohn, P., Syed, F., Reissaus, C. A., Mosley, A. L.,
& Evans-Molina, C. (2019). Endoplasmic reticulum stress alters ryanodine receptor
function in the murine pancreatic β cell. Journal of Biological Chemistry, 294(1), 168-
181.
23. Hicks, J. J., Torres-Ramos, Y. D., & Sierra-Vargas, M. P. (2006). Estrés oxidante.
Concepto y clasificación. Revista de endocrinología y nutrición, 14(4), 223-226

24. Halliwell, B., & Gutteridge, J. M. (2015). Free radicals in biology and medicine.
Oxford: Oxford University Press.

25. Guazzelli, P. A., Cittolin-Santos, G. F., Meira-Martins, L. A., Grings, M., Nonose,
Y., Lazzarotto, G. S., ... & de Assis, A. M. (2020). Acute liver failure induces glial
reactivity, oxidative stress and impairs brain energy metabolism in rats. Frontiers in
Molecular Neuroscience, 327.
26. Tangvarasittichai, S. (2015). Oxidative stress, insulin resistance, dyslipidemia and
type 2 diabetes mellitus. World journal of diabetes, 6(3), 456.

27. Maritim, A. C., Sanders, A., & Watkins Iii, J. B. (2003). Diabetes, oxidative stress,
and antioxidants: a review. Journal of biochemical and molecular toxicology, 17(1), 24-
38.

28. Cabello-Verrugio, C., Simon, F., Trollet, C., & Santibañez, J. F. (2016). Oxidative
stress in disease and aging: mechanisms and therapies 2016. Oxidative medicine and
cellular longevity, 4310469.

29. Færch, K., Vistisen, D., Pacini, G., Torekov, S. S., Johansen, N. B., Witte, D. R., &
Holst, J. J. (2016). Insulin resistance is accompanied by increased fasting glucagon and
delayed glucagon suppression in individuals with normal and impaired glucose
regulation. Diabetes, 65(11), 3473-3481.

30. Hall, J. E. (2015). Guyton and Hall textbook of medical physiology. e-Book:
Elsevier Health Sciences.

31. Robertson, R. P. (2006). Oxidative stress and impaired insulin secretion in type 2
diabetes. Current opinion in pharmacology, 6(6), 615-619.

32. Samuel, V. T., & Shulman, G. I. (2016). The pathogenesis of insulin resistance:
integrating signaling pathways and substrate flux. The Journal of clinical
investigation, 126(1), 12-22.

33. Gier, B., Krippeit-Drews, P., Sheiko, T., Aguilar-Bryan, L., Bryan, J., Düfer, M., &
Drews, G. (2009). Suppression of K ATP channel activity protects murine pancreatic β
cells against oxidative stress. The Journal of clinical investigation, 119(11), 3246-3256.

34. Wang, J., & Wang, H. (2017). Oxidative stress in pancreatic beta cell
regeneration. Oxidative medicine and cellular longevity,1, 1-9.

35. Mafi, S., Mansoori, B., Taeb, S., Sadeghi, H., Abbasi, R., Cho, W. C., &
Rostamzadeh, D. (2022). mTOR-mediated regulation of immune responses in cancer
and tumor microenvironment. Frontiers in Immunology, 5724.

36. Reno, C. M., Puente, E. C., Sheng, Z., Daphna-Iken, D., Bree, A. J., Routh, V. H., ...
& Fisher, S. J. (2017). Brain GLUT4 knockout mice have impaired glucose tolerance,
decreased insulin sensitivity, and impaired hypoglycemic
counterregulation. Diabetes, 66(3), 587-597.

37. Iaccarino, G.; Sorriento, D. Insulin Resistance. Encyclopedia. Disponible online:


https://encyclopedia.pub/entry/6821 (Consultado en: 18 mayo 2022).

38. Lamming, Dudley. (2014). Diminished mTOR signaling: a common mode of action
for endocrine longevity factors. SpringerPlus. 3. 735. 10.1186/2193-1801-3-735.

39. Arpa-Gutierrez, F. J., Cruz-Martinez, A., Campos-Gonzalez, Y., Gutierrez-Molina,


M., Santiago-Perez, S., Perez-Conde, M. C., ... & Arenas-Barbero, J. (2005).
Mitochondrial respiratory chain diseases. Evaluation and variability in 52
patients. Revista de neurologia, 41(8), 449-454.

40. Hamilton, S. J., & Watts, G. F. (2013). Endothelial dysfunction in diabetes:


pathogenesis, significance, and treatment. The review of diabetic studies: RDS, 10(2-3),
133.

41. Bredt, D. S. (1999). Endogenous nitric oxide synthesis: biological functions and
pathophysiology. Free radical research, 31(6), 577-596.

42. Zhao, Y., Vanhoutte, P. M., & Leung, S. W. (2015). Vascular nitric oxide: Beyond
eNOS. Journal of pharmacological sciences, 129(2), 83-94.

43. Toda, N., & Herman, A. G. (2005). Gastrointestinal function regulation by nitrergic
efferent nerves. Pharmacological reviews, 57(3), 315-338.

44. Woodman, R. J., Chew, G. T., & Watts, G. F. (2005). Mechanisms, significance and
treatment of vascular dysfunction in type 2 diabetes mellitus. Drugs, 65(1), 31-74.

45. Mungrue, I. N., Bredt, D. S., Stewart, D. J., & Husain, M. (2003). From molecules
to mammals: what's NOS got to do with it? Acta Physiologica Scandinavica, 179(2),
123-135.

46. Lind, M., Hayes, A., Caprnda, M., Petrovic, D., Rodrigo, L., Kruzliak, P., & Zulli,
A. (2017). Inducible nitric oxide synthase: good or bad? Biomedicine &
Pharmacotherapy, 93, 370-375.

47. Gambardella, J., Khondkar, W., Morelli, M. B., Wang, X., Santulli, G., & Trimarco,
V. (2020). Arginine and endothelial function. Biomedicines, 8(8), 277.

48. Lundberg, J. O., Weitzberg, E., & Gladwin, M. T. (2008). The nitrate–nitrite–nitric
oxide pathway in physiology and therapeutics. Nature reviews Drug discovery, 7(2),
156-167.

49. Hamilton, S. J., & Watts, G. F. (2013). Disfunción endotelial en la diabetes:


patogénesis, importancia y tratamiento. La revisión de los estudios sobre la diabetes:
RDS, 10(2-3), 133.

50. Thomas, D. D., Liu, X., Kantrow, S. P., & Lancaster, J. R. (2001). The biological
lifetime of nitric oxide: implications for the perivascular dynamics of NO and
O2. Proceedings of the National Academy of Sciences, 98(1), 355-360.

51. Adams, L., Franco, M. C., & Estevez, A. G. (2015). Reactive nitrogen species in
cellular signaling. Experimental biology and medicine, 240(6), 711-717.

52. Schulze, E., & Steiger, E. (1886). Ueber einen neuen stickstoffhaltigen Bestandtheil
der Keimlinge von Lupinus luteus. Berichte der deutschen chemischen
Gesellschaft, 19(1), 1177-1180.
53. Pytliak, Marek & Mechírová, Viola., (2008). Koncentrácie asymetrického
dimetylarginínu ako marker kardiovaskulárneho rizika. Cardiol., 17(4):157–161.

54. Fitch, C. A., Platzer, G., Okon, M., Garcia‐Moreno E, B., & McIntosh, L. P. (2015).
Arginine: Its pKa value revisited. Protein science, 24(5), 752-761.

55. Wu, G., & Morris Jr, S. M. (1998). Arginine metabolism: nitric oxide and
beyond. Biochemical Journal, 336(1), 1-17.

56. Hsu, C. N., & Tain, Y. L. (2019). Impact of arginine nutrition and metabolism
during pregnancy on offspring outcomes. Nutrients, 11(7), 1452-1467.

57. Liu, Z., & Barrett, E. J. (2002). Human protein metabolism: its measurement and
regulation. American Journal of Physiology-Endocrinology and Metabolism, 283(6),
1105-1112.

58. Dhanakoti, S. N., Brosnan, J. T., Herzberg, G. R., & Brosnan, M. E. (1990). Renal
arginine synthesis: studies in vitro and in vivo. American Journal of Physiology-
Endocrinology and Metabolism, 259(3), 437.

59. Wu, G., Meininger, C. J., McNeal, C. J., Bazer, F. W., & Rhoads, J. M. (2021). Role
of L-arginine in nitric oxide synthesis and health in humans. In Amino Acids in
Nutrition and Health (pp. 167-187). Springer, Cham.

60. Wu, G., Davis, P. K., Flynn, N. E., Knabe, D. A., & Davidson, J. T. (1997).
Endogenous synthesis of arginine plays an important role in maintaining arginine
homeostasis in postweaning growing pigs. The Journal of nutrition, 127(12), 2342-
2349.

61. Strobel, J., Müller, F., Zolk, O., Endreß, B., König, J., Fromm, M. F., & Maas, R.
(2013). Transport of asymmetric dimethylarginine (ADMA) by cationic amino acid
transporter 2 (CAT2), organic cation transporter 2 (OCT2) and multidrug and toxin
extrusion protein 1 (MATE1). Amino acids, 45(4), 989-1002.

62. Closs, E. I., Basha, F. Z., Habermeier, A., & Förstermann, U. (1997). Interference
ofl-arginine analogues withl-arginine transport mediated by the y+ carrier hCAT-
2B. Nitric oxide, 1(1), 65-73.

63. Wu, G., Bazer, F. W., Davis, T. A., Kim, S. W., Li, P., Marc Rhoads, J., ... & Yin,
Y. (2009). Arginine metabolism and nutrition in growth, health and disease. Amino
acids, 37(1), 153-168.

64. Chien-an, A. H., Lin, W. W., Obie, C., & Valle, D. (1999). Molecular enzymology
of mammalian Δ1-pyrroline-5-carboxylate synthase: alternative splice donor utilization
generates isoforms with different sensitivity to ornithine inhibition. Journal of
Biological Chemistry, 274(10), 6754-6762.

65. Boelens, P. G., van Leeuwen, P. A., Dejong, C. H., & Deutz, N. E. (2005). Intestinal
renal metabolism of L-citrulline and L-arginine following enteral or parenteral infusion
of L-alanyl-L-[2, 15N] glutamine or L-[2, 15N] glutamine in mice. American Journal of
Physiology-Gastrointestinal and Liver Physiology, 289(4), 679-685.

66. Levillain, O. (2012). Expression and function of arginine-producing and consuming-


enzymes in the kidney. Amino acids, 42(4), 1237-1252.

67. Bratt, J. M., Zeki, A. A., Last, J. A., & Kenyon, N. J. (2011). Competitive
metabolism of L-arginine: arginase as a therapeutic target in asthma. Journal of
biomedical research, 25(5), 299-308.

68. Cabello-Verrugio, C., Simon, F., Trollet, C., & Santibañez, J. F. (2016). Oxidative
stress in disease and aging: mechanisms and therapies 2016. Oxidative medicine and
cellular longevity, 4310469.

69. Kövamees, O., Shemyakin, A., Checa, A., Wheelock, C. E., Lundberg, J. O.,
Östenson, C. G., & Pernow, J. (2016). Arginase inhibition improves microvascular
endothelial function in patients with type 2 diabetes mellitus. The Journal of Clinical
Endocrinology & Metabolism, 101(11), 3952-3958.

70. Ash, D. E., Cox, J. D., & Christianson, D. W. (2000). Arginase: a binuclear
manganese metalloenzyme. Met Ions Biol Syst, 37, 407-428.

71. Kim, J. H., Bugaj, L. J., Oh, Y. J., Bivalacqua, T. J., Ryoo, S., Soucy, K. G., ... &
Berkowitz, D. E. (2009). Arginase inhibition restores NOS coupling and reverses
endothelial dysfunction and vascular stiffness in old rats. Journal of applied
physiology, 107(4), 1249-1257.

72. Förstermann, U., & Sessa, W. C. (2012). Nitric oxide synthases: regulation and
function. European heart journal, 33(7), 829-837.

73. Hamilton, S. J., & Watts, G. F. (2013). Endothelial dysfunction in diabetes:


pathogenesis, significance, and treatment. The review of diabetic studies: RDS, 10(2-3),
133.

74. Pernow, J., & Jung, C. (2013). Arginase as a potential target in the treatment of
cardiovascular disease: reversal of arginine steal? Cardiovascular research, 98(3), 334-
343.

75. Bredt, D. S., & Snyder, S. H. (1990). Isolation of nitric oxide synthetase, a
calmodulin-requiring enzyme. Proceedings of the National Academy of Sciences, 87(2),
682-685.

76. Chin-Dusting, J. P. F., Willems, L., & Kaye, D. M. (2007). L-arginine transporters
in cardiovascular disease: a novel therapeutic target. Pharmacology &
therapeutics, 116(3), 428-436.

77. Closs, E. I., Simon, A., Vékony, N., & Rotmann, A. (2004). Plasma membrane
transporters for arginine. The Journal of nutrition, 134(10), 2752-2759.
78. Blanc, R. S., & Richard, S. (2017). Arginine methylation: the coming of
age. Molecular cell, 65(1), 8-24.

79. Morales, Y., Cáceres, T., May, K., & Hevel, J. M. (2016). Biochemistry and
regulation of the protein arginine methyltransferases (PRMTs). Archives of biochemistry
and biophysics, 590, 138-152.

80. San Miguel, A., San Miguel, R., & Gil, F. M. (2008). Dimetilarginina asimétrica
(ADMA) en diferentes enfermedades. Revista del Laboratorio Clínico, 1(3), 113-121.

81. Tain, Y. L., & Hsu, C. N. (2017). Toxic dimethylarginines: asymmetric


dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA). Toxins, 9(3), 92.

82. Rodrigues-Krause, J., Krause, M., Rocha, I. M. G. D., Umpierre, D., & Fayh, A. P.
T. (2018). Association of l-arginine supplementation with markers of endothelial
function in patients with cardiovascular or metabolic disorders: a systematic review and
meta-analysis. Nutrients, 11(1), 15.

83. Bahrami, D., Mozaffari-Khosravi, H., & Zavar-Reza, J. (2019). The effect of oral L-
arginine supplementation on lipid profile, glycemic status, and insulin resistance in
patients with metabolic syndrome: a randomized, double-blind, placebo-controlled
trial. Mediterranean journal of nutrition and metabolism, 12(1), 79-90.

84. Lucotti, P., Setola, E., Monti, L. D., Galluccio, E., Costa, S., Sandoli, E. P., ... &
Piatti, P. (2006). Beneficial effects of a long-term oral L-arginine treatment added to a
hypocaloric diet and exercise training program in obese, insulin-resistant type 2 diabetic
patients. American Journal of Physiology-Endocrinology and Metabolism, 291(5),
E906-E912.

85. Mohamadin, A. M., Hammad, L. N., El‐Bab, M. F., & Gawad, H. S. A. (2007). Can
nitric oxide‐generating compounds improve the oxidative stress response in
experimentally diabetic rats? Clinical and experimental pharmacology and
physiology, 34(7), 586-593.

86. Karimi, E., Hatami, E., Ghavami, A., Hadi, A., Darand, M., & Askari, G. (2021).
Effects of L-arginine supplementation on biomarkers of glycemic control: a systematic
review and meta‐analysis of randomised clinical trials. Archives of Physiology and
Biochemistry, 1-11.

87. Rad, E. Y., Nazarian, B., Saboori, S., Falahi, E., & Hekmatdoost, A. (2020). Effects
of l-arginine supplementation on glycemic profile: Evidence from a systematic review
and meta-analysis of clinical trials. Journal of Integrative Medicine, 18(4), 284-291.

88. Niu, M., Xiang, L., Liu, Y., Zhao, Y., Yuan, J., Dai, X., & Chen, H. (2017).
Adiponectin induced AMP-activated protein kinase impairment mediates insulin
resistance in Bama mini-pig fed high-fat and high-sucrose diet. Asian-Australasian
journal of animal sciences, 30(8), 1190-1197.

89. Sansbury, B. E., & Hill, B. G. (2014). Regulation of obesity and insulin resistance
by nitric oxide. Free radical biology and medicine, 73, 383-399.
90. Achari, A. E., & Jain, S. K. (2017). Adiponectin, a therapeutic target for obesity,
diabetes, and endothelial dysfunction. International journal of molecular
sciences, 18(6), 1321, 1-17.

91. Monti, L. D., Setola, E., Lucotti, P. C. G., Marrocco‐Trischitta, M. M., Comola, M.,
Galluccio, E., ... & Piatti, P. M. (2012). Effect of a long‐term oral l‐arginine
supplementation on glucose metabolism: a randomized, double‐blind, placebo‐
controlled trial. Diabetes, obesity and metabolism, 14(10), 893-900.

92. Bai, Y., Sun, L., Yang, T., Sun, K., Chen, J., & Hui, R. (2009). Increase in fasting
vascular endothelial function after short-term oral L-arginine is effective when baseline
flow-mediated dilation is low: a meta-analysis of randomized controlled trials. The
American journal of clinical nutrition, 89(1), 77-84.

93. Drexler, H., Fischell, T. A., Pinto, F. J., Chenzbraun, A., Botas, J., Cooke, J. P., &
Alderman, E. L. (1994). Effect of L-arginine on coronary endothelial function in cardiac
transplant recipients. Relation to vessel wall morphology. Circulation, 89(4), 1615-
1623.

94. Creager, M. A., Gallagher, S. J., Girerd, X. J., Coleman, S. M., Dzau, V. J., &
Cooke, J. P. (1992). L-arginine improves endothelium-dependent vasodilation in
hypercholesterolemic humans. The Journal of clinical investigation, 90(4), 1248-1253.

95. Besset, A., Bonardet, A., Rondouin, G., Descomps, B., & Passouant, P. (1982).
Increase in sleep related GH and Prl secretion after chronic arginine aspartate
administration in man. European Journal of Endocrinology, 99(1), 18-23.

96. Adams, M. R., McCredie, R., Jessup, W., Robinson, J., Sullivan, D., & Celermajer,
D. S. (1997). Oral L-arginine improves endothelium-dependent dilatation and reduces
monocyte adhesion to endothelial cells in young men with coronary artery
disease. Atherosclerosis, 129(2), 261-269.

97. Stapleton, P. A., Goodwill, A. G., James, M. E., Brock, R. W., & Frisbee, J. C.
(2010). Hypercholesterolemia and microvascular dysfunction: interventional
strategies. Journal of inflammation, 7(1), 1-10.

98. Horowitz, J. D., & Heresztyn, T. (2007). An overview of plasma concentrations of


asymmetric dimethylarginine (ADMA) in health and disease and in clinical studies:
methodological considerations. Journal of Chromatography B, 851(1-2), 42-50.

99. Tran, C. T., Leiper, J. M., & Vallance, P. (2003). The ddah/adma/nos
pathway. Atherosclerosis Supplements, 4(4), 33-40.

100. Sydow, K., & Münzel, T. (2003). ADMA and oxidative stress. Atherosclerosis
Supplements, 4(4), 41-51.

101. Kohli, R., Meininger, C. J., Haynes, T. E., Yan, W., Self, J. T., & Wu, G. (2004).
Dietary L-arginine supplementation enhances endothelial nitric oxide synthesis in
streptozotocin-induced diabetic rats. The Journal of nutrition, 134(3), 600-608.
102. Lucotti, P., Monti, L., Setola, E., La Canna, G., Castiglioni, A., Rossodivita, A., ...
& Piatti, P. (2009). Oral L-arginine supplementation improves endothelial function and
ameliorates insulin sensitivity and inflammation in cardiopathic nondiabetic patients
after an aortocoronary bypass. Metabolism, 58(9), 1270-1276.

103. Zarezadeh, M., Emami, M. R., Kord-Varkane, H., Mousavi, S. M., Alizadeh, H.,
Asbaghi, O., ... & Khorshidi, M. (2020). The effect of oral L-arginine supplementation
on asymmetric dimethylarginine levels: A systematic review and meta-analysis of
randomized clinical trials. Advances in Integrative Medicine, 7(2), 61-66.

104. Shiraseb, F., Asbaghi, O., Bagheri, R., Wong, A., Figueroa, A., & Mirzaei, K.
(2021). The Effect of L-arginine Supplementation On Blood Pressure in Adults: A
Systematic Review and Dose-response Meta-analysis of Randomized Clinical Trials.
Advances in Nutrition. Epub.

105. Dong, J. Y., Qin, L. Q., Zhang, Z., Zhao, Y., Wang, J., Arigoni, F., & Zhang, W.
(2011). Effect of oral L-arginine supplementation on blood pressure: a meta-analysis of
randomized, double-blind, placebo-controlled trials. American heart journal, 162(6),
959-965.

106. Chobanian, A. V., Bakris, G. L., Black, H. R., Cushman, W. C., Green, L. A., Izzo
Jr, J. L., ... & National High Blood Pressure Education Program Coordinating
Committee. (2003). Seventh report of the joint national committee on prevention,
detection, evaluation, and treatment of high blood pressure. hypertension, 42(6), 1206-
1252.

107. Melik, Z., Zaletel, P., Virtic, T., & Cankar, K. (2017). L-arginine as dietary
supplement for improving microvascular function. Clinical hemorheology and
microcirculation, 65(3), 205-217.

108. Khalaf, D., Krüger, M., Wehland, M., Infanger, M., & Grimm, D. (2019). The
effects of oral l-arginine and l-citrulline supplementation on blood
pressure. Nutrients, 11(7), 1679, 1-14.

109. Rajapakse, N. W., De Miguel, C., Das, S., & Mattson, D. L. (2008). Exogenous L-
arginine ameliorates angiotensin II-induced hypertension and renal damage in
rats. Hypertension, 52(6), 1084-1090.

110. Kelly, L. K., Wedgwood, S., Steinhorn, R. H., & Black, S. M. (2004). Nitric oxide
decreases endothelin-1 secretion through the activation of soluble guanylate
cyclase. American Journal of Physiology-Lung Cellular and Molecular
Physiology, 286(5), 984-991.

111. Laplante, M. A., Wu, R., Moreau, P., & de Champlain, J. (2005). Endothelin
mediates superoxide production in angiotensin II-induced hypertension in rats. Free
Radical Biology and Medicine, 38(5), 589-596.
112. Muniyappa, R., & Yavuz, S. (2013). Metabolic actions of angiotensin II and
insulin: a microvascular endothelial balancing act. Molecular and cellular
endocrinology, 378(1-2), 59-69.

113. McNeal, C. J., Meininger, C. J., Wilborn, C. D., Tekwe, C. D., & Wu, G. (2018).
Safety of dietary supplementation with arginine in adult humans. Amino Acids, 50(9),
1215-1229.

114. Wascher, T. C., Posch, K., Wallner, S., Hermetter, A., Kostner, G. M., & Graier,
W. F. (1997). Vascular effects of L-arginine: anything beyond a substrate for the NO-
synthase? Biochemical and biophysical research communications, 234(1), 35-38.

115. Leone, A., Moncada, S., Vallance, P., Calver, A., & Collier, J. (1992).
Accumulation of an endogenous inhibitor of nitric oxide synthesis in chronic renal
failure. The Lancet, 339(8793), 572-575.

116. Conlon, K., Collins, T., & Kidd, C. (1996). Modulation of vagal actions on heart
rate produced by inhibition of nitric oxide synthase in the anaesthetized
ferret. Experimental Physiology: Translation and Integration, 81(3), 547-550.

117. Shiraseb, F., Asbaghi, O., Bagheri, R., Wong, A., Figueroa, A., & Mirzaei, K.
(2021). The Effect of L-arginine Supplementation on Blood Pressure in Adults: A
Systematic Review and Dose-response Meta-analysis of Randomized Clinical
Trials. Advances in Nutrition, 1-17.

118. Huang, J., Ladeiras, D., Yu, Y., Ming, X. F., & Yang, Z. (2021). Detrimental
effects of chronic L-arginine rich food on aging kidney. Frontiers in pharmacology,
2126.

119. Mousavi, S. M., Milajerdi, A., Fatahi, S., Rahmani, J., Zarezadeh, M., Ghaedi, E.,
& Varkaneh, H. K. (2019). The effect of L-arginine supplementation on obesity-related
indices: A systematic review and meta-analysis of randomized clinical trials.
International Journal for Vitamin and Nutrition Research. Epub.

120. Hurt, R. T., Ebbert, J. O., Schroeder, D. R., Croghan, I. T., Bauer, B. A., McClave,
S. A., ... & McClain, C. J. (2014). L-arginine for the treatment of centrally obese
subjects: a pilot study. Journal of dietary supplements, 11(1), 40-52.

121. Tan, B., Yin, Y., Liu, Z., Li, X., Xu, H., Kong, X., ... & Wu, G. (2009). Dietary L-
arginine supplementation increases muscle gain and reduces body fat mass in growing-
finishing pigs. Amino acids, 37(1), 169-175.

122. Jensen, N. S. O., Camargo, T. F. B., & Bergamaschi, D. P. (2016). Comparison of


methods to measure body fat in 7-to-10-year-old children: a systematic review. Public
Health, 133, 3-13.

123. Jobgen, W., Meininger, C. J., Jobgen, S. C., Li, P., Lee, M. J., Smith, S. B., ... &
Wu, G. (2009). Dietary L-arginine supplementation reduces white fat gain and enhances
skeletal muscle and brown fat masses in diet-induced obese rats. The Journal of
nutrition, 139(2), 230-237.
124. Benton, C. R., Holloway, G. P., Han, X. X., Yoshida, Y., Snook, L. A., Lally, J., ...
& Bonen, A. (2010). Increased levels of peroxisome proliferator-activated receptor
gamma, coactivator 1 alpha (PGC-1α) improve lipid utilisation, insulin signalling and
glucose transport in skeletal muscle of lean and insulin-resistant obese Zucker
rats. Diabetologia, 53(9), 2008-2019.

You might also like