Carnosine (β-alanyl-L-histidine) is an endogenous dipeptide widely distributed in mammalian tissues, especially skeletal and cardiac muscle cells, and, to a lesser extent, in the brain. While early interest in carnosine was given because of its role in muscle cell metabolism and athletic performance, it has more recently gained attention for its potential application in several chronic diseases. Specifically, brain aging and neurodegenerative disorders have received particular attention, as a marked reduction in carnosine levels has been described in these conditions. Carnosine exerts a wide range of biological activities, including antioxidant, anti-inflammatory, anti-glycation, metal-chelating, and neuroprotective properties. Mechanistically, it acts by inhibiting the production of advanced glycation end products (AGEs), buffering cellular pH, and regulating intracellular nitric oxide signaling and mitochondrial function. Its safety profile, the lack of toxicity, and significant side effects support its application for long-term therapeutic use. In this review, we aim to recapitulate and discuss the effects, dosages, and administration routes of carnosine in preclinical in vivo models, with a particular focus on neurodegenerative disorders where it has been shown to reduce oxidative stress, suppress neuroinflammation, modulate protein aggregation, and preserve cognitive function, all key features of neurodegeneration. Despite promising findings, there are gaps in the knowledge on how carnosine affects synaptic plasticity, neuronal remodeling, and other processes that play a central role in the pathophysiology of neurodegenerative disorders. Additionally, clinical translation remains challenging due to inconsistencies across in vivo studies in terms of dosage, treatment duration, routes of administration, and disease models, which affect reproducibility and cross-study comparability. Therefore, while carnosine emerges as a multifunctional and well-tolerated molecule, further research is needed to clarify its therapeutic relevance in human diseases. In this review, we also address future perspectives and key methodological challenges that must be overcome to effectively translate carnosine's biological potential into clinical practice.
Citation: Saviana Antonella Barbati, Giuseppe Carota, Konstantinos Partsinevelos, Lucia Di Pietro, Anna Privitera, Vincenzo Cardaci, Andrea Graziani, Renata Mangione, Giuseppe Lazzarino, Barbara Tavazzi, Valentina Di Pietro, Emiliano Maiani, Francesco Bellia, Angela Maria Amorini, Giacomo Lazzarino, Shahid Pervez Baba, Giuseppe Caruso. Preclinical evidence and therapeutic perspectives on carnosine for the treatment of neurodegenerative disorders[J]. AIMS Neuroscience, 2025, 12(4): 444-513. doi: 10.3934/Neuroscience.2025025
Carnosine (β-alanyl-L-histidine) is an endogenous dipeptide widely distributed in mammalian tissues, especially skeletal and cardiac muscle cells, and, to a lesser extent, in the brain. While early interest in carnosine was given because of its role in muscle cell metabolism and athletic performance, it has more recently gained attention for its potential application in several chronic diseases. Specifically, brain aging and neurodegenerative disorders have received particular attention, as a marked reduction in carnosine levels has been described in these conditions. Carnosine exerts a wide range of biological activities, including antioxidant, anti-inflammatory, anti-glycation, metal-chelating, and neuroprotective properties. Mechanistically, it acts by inhibiting the production of advanced glycation end products (AGEs), buffering cellular pH, and regulating intracellular nitric oxide signaling and mitochondrial function. Its safety profile, the lack of toxicity, and significant side effects support its application for long-term therapeutic use. In this review, we aim to recapitulate and discuss the effects, dosages, and administration routes of carnosine in preclinical in vivo models, with a particular focus on neurodegenerative disorders where it has been shown to reduce oxidative stress, suppress neuroinflammation, modulate protein aggregation, and preserve cognitive function, all key features of neurodegeneration. Despite promising findings, there are gaps in the knowledge on how carnosine affects synaptic plasticity, neuronal remodeling, and other processes that play a central role in the pathophysiology of neurodegenerative disorders. Additionally, clinical translation remains challenging due to inconsistencies across in vivo studies in terms of dosage, treatment duration, routes of administration, and disease models, which affect reproducibility and cross-study comparability. Therefore, while carnosine emerges as a multifunctional and well-tolerated molecule, further research is needed to clarify its therapeutic relevance in human diseases. In this review, we also address future perspectives and key methodological challenges that must be overcome to effectively translate carnosine's biological potential into clinical practice.
| [1] |
Gulewitsch W, Amiradžibi S (1900) Ueber das Carnosin, eine neue organische Base des Fleischextractes. Ber Dtsch Chem Ges 33: 1902-1903. https://doi.org/10.1002/cber.19000330275
|
| [2] |
Jędrejko M, Kała K, Muszyńska B (2025) Anserine, Balenine, and Ergothioneine: Impact of Histidine-Containing Compounds on Exercise Performance-A Narrative Review. Nutrients 17: 828. https://doi.org/10.3390/nu17050828
|
| [3] |
Kawahara M, Tanaka KI, Kato-Negishi M (2018) Zinc, Carnosine, and Neurodegenerative Diseases. Nutrients 10: 147. https://doi.org/10.3390/nu10020147
|
| [4] |
Caruso G, Caraci F, Jolivet RB (2019) Pivotal role of carnosine in the modulation of brain cells activity: Multimodal mechanism of action and therapeutic potential in neurodegenerative disorders. Prog Neurobiol 175: 35-53. https://doi.org/10.1016/j.pneurobio.2018.12.004
|
| [5] |
Boldyrev AA, Aldini G, Derave W (2013) Physiology and pathophysiology of carnosine. Physiol Rev 93: 1803-1845. https://doi.org/10.1152/physrev.00039.2012
|
| [6] |
Teufel M, Saudek V, Ledig JP, et al. (2003) Sequence identification and characterization of human carnosinase and a closely related non-specific dipeptidase. J Biol Chem 278: 6521-6531. https://doi.org/10.1074/jbc.M209764200
|
| [7] |
Lenney JF, George RP, Weiss AM, et al. (1982) Human serum carnosinase: characterization, distinction from cellular carnosinase, and activation by cadmium. Clin Chim Acta 123: 221-231. https://doi.org/10.1016/0009-8981(82)90166-8
|
| [8] |
Lenney JF (1990) Human cytosolic carnosinase: evidence of identity with prolinase, a non-specific dipeptidase. Biol Chem Hoppe Seyler 371: 167-171. https://doi.org/10.1515/bchm3.1990.371.1.167
|
| [9] | Mokrushin AJIAnRanSb (2024) Dipeptide L-сarnosine (β-alanyl-L-histidine)—nervous tissue cryoprotector non-hybernate animals. Izvestiâ Akademii nauk. Rossijskaâ akademiâ nauk. Seriâ biologičeskaâ 2024: 346-357. https://doi.org/10.31857/S1026347024030064 |
| [10] |
Caruso G, Fresta CG, Martinez-Becerra F, et al. (2017) Carnosine modulates nitric oxide in stimulated murine RAW 264.7 macrophages. Mol Cell Biochem 431: 197-210. https://doi.org/10.1007/s11010-017-2991-3
|
| [11] |
Fresta CG, Fidilio A, Lazzarino G, et al. (2020) Modulation of Pro-Oxidant and Pro-Inflammatory Activities of M1 Macrophages by the Natural Dipeptide Carnosine. Int J Mol Sci 21: 776. https://doi.org/10.3390/ijms21030776
|
| [12] |
Spaas J, Franssen WMA, Keytsman C, et al. (2021) Carnosine quenches the reactive carbonyl acrolein in the central nervous system and attenuates autoimmune neuroinflammation. J Neuroinflammation 18: 255. https://doi.org/10.1186/s12974-021-02306-9
|
| [13] |
Aloisi A, Barca A, Romano A, et al. (2013) Anti-aggregating effect of the naturally occurring dipeptide carnosine on aβ1-42 fibril formation. PLoS One 8: e68159. https://doi.org/10.1371/journal.pone.0068159
|
| [14] |
Hasanein P, Felegari Z (2017) Chelating effects of carnosine in ameliorating nickel-induced nephrotoxicity in rats. Can J Physiol Pharmacol 95: 1426-1432. https://doi.org/10.1139/cjpp-2016-0647
|
| [15] |
Baba SP, Hoetker JD, Merchant M, et al. (2013) Role of aldose reductase in the metabolism and detoxification of carnosine-acrolein conjugates. J Biol Chem 288: 28163-28179. https://doi.org/10.1074/jbc.M113.504753
|
| [16] |
Berezhnoy DS, Stvolinsky SL, Lopachev AV, et al. (2019) Carnosine as an effective neuroprotector in brain pathology and potential neuromodulator in normal conditions. Amino Acids 51: 139-150. https://doi.org/10.1007/s00726-018-2667-7
|
| [17] |
Caruso G (2022) Unveiling the Hidden Therapeutic Potential of Carnosine, a Molecule with a Multimodal Mechanism of Action: A Position Paper. Molecules 27: 3303. https://doi.org/10.3390/molecules27103303
|
| [18] |
Bonaccorso A, Privitera A, Grasso M, et al. (2023) The Therapeutic Potential of Novel Carnosine Formulations: Perspectives for Drug Development. Pharmaceuticals (Basel) 16: 778. https://doi.org/10.3390/ph16060778
|
| [19] |
Décombaz J, Beaumont M, Vuichoud J, et al. (2012) Effect of slow-release β-alanine tablets on absorption kinetics and paresthesia. Amino acids 43: 67-76. https://doi.org/10.1007/s00726-011-1169-7
|
| [20] |
Salatto RW, McGinnis GR, Davis DW, et al. (2021) Effects of Acute Beta-Alanine Ingestion and Immersion-Plus-Exercise on Connectedness to Nature and Perceived Pain. Int J Environ Res Public Health 18: 8134. https://doi.org/10.3390/ijerph18158134
|
| [21] |
Aldini G, de Courten B, Regazzoni L, et al. (2021) Understanding the antioxidant and carbonyl sequestering activity of carnosine: direct and indirect mechanisms. Free Radic Res 55: 321-330. https://doi.org/10.1080/10715762.2020.1856830
|
| [22] |
Chmielewska K, Dzierzbicka K, Inkielewicz-Stępniak I, et al. (2020) Therapeutic Potential of Carnosine and Its Derivatives in the Treatment of Human Diseases. Chem Res Toxicol 33: 1561-1578. https://doi.org/10.1021/acs.chemrestox.0c00010
|
| [23] |
Lee YT, Hsu CC, Lin MH, et al. (2005) Histidine and carnosine delay diabetic deterioration in mice and protect human low density lipoprotein against oxidation and glycation. Eur J Pharmacol 513: 145-150. https://doi.org/10.1016/j.ejphar.2005.02.010
|
| [24] |
Tanida M, Niijima A, Fukuda Y, et al. (2005) Dose-dependent effects of L-carnosine on the renal sympathetic nerve and blood pressure in urethane-anesthetized rats. Am J Physiol Regul Integr Comp Physiol 288: R447-455. https://doi.org/10.1152/ajpregu.00275.2004
|
| [25] |
Kurata H, Fujii T, Tsutsui H, et al. (2006) Renoprotective effects of l-carnosine on ischemia/reperfusion-induced renal injury in rats. J Pharmacol Exp Ther 319: 640-647. https://doi.org/10.1124/jpet.106.110122
|
| [26] |
Cuzzocrea S, Genovese T, Failla M, et al. (2007) Protective effect of orally administered carnosine on bleomycin-induced lung injury. Am J Physiol Lung Cell Mol Physiol 292: L1095-1104. https://doi.org/10.1152/ajplung.00283.2006
|
| [27] |
Mahmoud AH (2006) Comparative study between carnosine and fluvastatin in hypercholesterolemic rabbits. J Appl Sci 6: 1725-1730. https://doi.org/10.3923/jas.2006.1725.1730
|
| [28] |
Sauerhöfer S, Yuan G, Braun GS, et al. (2007) L-carnosine, a substrate of carnosinase-1, influences glucose metabolism. Diabetes 56: 2425-2432. https://doi.org/10.2337/db07-0177
|
| [29] |
Fouad AA, El-Rehany MA, Maghraby HK (2007) The hepatoprotective effect of carnosine against ischemia/reperfusion liver injury in rats. Eur J Pharmacol 572: 61-68. https://doi.org/10.1016/j.ejphar.2007.06.010
|
| [30] |
Kamei J, Ohsawa M, Miyata S, et al. (2008) Preventive effect of L-carnosine on changes in the thermal nociceptive threshold in streptozotocin-induced diabetic mice. Eur J Pharmacol 600: 83-86. https://doi.org/10.1016/j.ejphar.2008.10.002
|
| [31] |
Liu WH, Liu TC, Yin MC (2008) Beneficial effects of histidine and carnosine on ethanol-induced chronic liver injury. Food Chem Toxicol 46: 1503-1509. https://doi.org/10.1016/j.fct.2007.12.013
|
| [32] |
Mehmetçik G, Ozdemirler G, Koçak-Toker N, et al. (2008) Role of carnosine in preventing thioacetamide-induced liver injury in the rat. Peptides 29: 425-429. https://doi.org/10.1016/j.peptides.2007.11.008
|
| [33] |
Baykara B, Tekmen I, Pekcetin C, et al. (2009) The protective effects of carnosine and melatonin in ischemia-reperfusion injury in the rat liver. Acta Histochem 111: 42-51. https://doi.org/10.1016/j.acthis.2008.03.002
|
| [34] | Yan SL, Wu ST, Yin MC, et al. (2009) Protective effects from carnosine and histidine on acetaminophen-induced liver injury. J Food Sci 74: H259-265. https://doi.org/10.1111/j.1750-3841.2009.01330.x |
| [35] |
Fouad AA, Qureshi HA, Yacoubi MT, et al. (2009) Protective role of carnosine in mice with cadmium-induced acute hepatotoxicity. Food Chem Toxicol 47: 2863-2870. https://doi.org/10.1016/j.fct.2009.09.009
|
| [36] |
Kamal MA, Jiang H, Hu Y, et al. (2009) Influence of genetic knockout of Pept2 on the in vivo disposition of endogenous and exogenous carnosine in wild-type and Pept2 null mice. Am J Physiol Regul Integr Comp Physiol 296: R986-991. https://doi.org/10.1152/ajpregu.90744.2008
|
| [37] |
Renner C, Zemitzsch N, Fuchs B, et al. (2010) Carnosine retards tumor growth in vivo in an NIH3T3-HER2/neu mouse model. Mol Cancer 9: 1-7. https://doi.org/10.1186/1476-4598-9-2
|
| [38] |
Ozel Turkcu U, Bilgihan A, Biberoglu G, et al. (2010) Carnosine supplementation protects rat brain tissue against ethanol-induced oxidative stress. Mol Cell Biochem 339: 55-61. https://doi.org/10.1007/s11010-009-0369-x
|
| [39] |
Aydin AF, Küçükgergin C, Ozdemirler-Erata G, et al. (2010) The effect of carnosine treatment on prooxidant-antioxidant balance in liver, heart and brain tissues of male aged rats. Biogerontology 11: 103-109. https://doi.org/10.1007/s10522-009-9232-4
|
| [40] |
Aydin AF, Küskü-Kiraz Z, Doğru-Abbasoğlu S, et al. (2010) Effect of carnosine against thioacetamide-induced liver cirrhosis in rat. Peptides 31: 67-71. https://doi.org/10.1016/j.peptides.2009.11.028
|
| [41] |
Artun BC, Küskü-Kiraz Z, Güllüoğlu M, et al. (2010) The effect of carnosine pretreatment on oxidative stress and hepatotoxicity in binge ethanol administered rats. Hum Exp Toxicol 29: 659-665. https://doi.org/10.1177/0960327109359460
|
| [42] |
Riedl E, Pfister F, Braunagel M, et al. (2011) Carnosine prevents apoptosis of glomerular cells and podocyte loss in STZ diabetic rats. Cell Physiol Biochem 28: 279-288. https://doi.org/10.1159/000331740
|
| [43] |
Aldini G, Orioli M, Rossoni G, et al. (2011) The carbonyl scavenger carnosine ameliorates dyslipidaemia and renal function in Zucker obese rats. J Cell Mol Med 15: 1339-1354. https://doi.org/10.1111/j.1582-4934.2010.01101.x
|
| [44] |
Tsoi B, He RR, Yang DH, et al. (2011) Carnosine ameliorates stress-induced glucose metabolism disorder in restrained mice. J Pharmacol Sci 117: 223-229. https://doi.org/10.1254/jphs.11131fp
|
| [45] |
Mong MC, Chao CY, Yin MC (2011) Histidine and carnosine alleviated hepatic steatosis in mice consumed high saturated fat diet. Eur J Pharmacol 653: 82-88. https://doi.org/10.1016/j.ejphar.2010.12.001
|
| [46] |
Li YF, He RR, Tsoi B, et al. (2012) Anti-stress effects of carnosine on restraint-evoked immunocompromise in mice through spleen lymphocyte number maintenance. PLoS One 7: e33190. https://doi.org/10.1371/journal.pone.0033190
|
| [47] |
Menini S, Iacobini C, Ricci C, et al. (2012) D-Carnosine octylester attenuates atherosclerosis and renal disease in ApoE null mice fed a Western diet through reduction of carbonyl stress and inflammation. Br J Pharmacol 166: 1344-1356. https://doi.org/10.1111/j.1476-5381.2012.01834.x
|
| [48] |
Peters V, Schmitt CP, Zschocke J, et al. (2012) Carnosine treatment largely prevents alterations of renal carnosine metabolism in diabetic mice. Amino Acids 42: 2411-2416. https://doi.org/10.1007/s00726-011-1046-4
|
| [49] |
Ansurudeen I, Sunkari VG, Grünler J, et al. (2012) Carnosine enhances diabetic wound healing in the db/db mouse model of type 2 diabetes. Amino Acids 43: 127-134. https://doi.org/10.1007/s00726-012-1269-z
|
| [50] |
Barski OA, Xie Z, Baba SP, et al. (2013) Dietary carnosine prevents early atherosclerotic lesion formation in apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol 33: 1162-1170. https://doi.org/10.1161/atvbaha.112.300572
|
| [51] |
Everaert I, Stegen S, Vanheel B, et al. (2013) Effect of beta-alanine and carnosine supplementation on muscle contractility in mice. Med Sci Sports Exerc 45: 43-51. https://doi.org/10.1249/MSS.0b013e31826cdb68
|
| [52] |
Sahin S, Oter S, Burukoğlu D, et al. (2013) The effects of carnosine in an experimental rat model of septic shock. Med Sci Monit Basic Res 19: 54-61. https://doi.org/10.12659/msmbr.883758
|
| [53] |
Kalaz EB, Çoban J, Aydın AF, et al. (2014) Carnosine and taurine treatments decreased oxidative stress and tissue damage induced by D-galactose in rat liver. J Physiol Biochem 70: 15-25. https://doi.org/10.1007/s13105-013-0275-2
|
| [54] |
Giriş M, Doğru-Abbasoğlu S, Kumral A, et al. (2014) Effect of carnosine alone or combined with α-tocopherol on hepatic steatosis and oxidative stress in fructose-induced insulin-resistant rats. J Physiol Biochem 70: 385-395. https://doi.org/10.1007/s13105-014-0314-7
|
| [55] |
Macarini JR, Maravai SG, Cararo JH, et al. (2014) Impairment of electron transfer chain induced by acute carnosine administration in skeletal muscle of young rats. Biomed Res Int 2014: 632986. https://doi.org/10.1155/2014/632986
|
| [56] |
Brown BE, Kim CH, Torpy FR, et al. (2014) Supplementation with carnosine decreases plasma triglycerides and modulates atherosclerotic plaque composition in diabetic apo E(-/-) mice. Atherosclerosis 232: 403-409. https://doi.org/10.1016/j.atherosclerosis.2013.11.068
|
| [57] |
Evran B, Karpuzoğlu H, Develi S, et al. (2014) Effects of carnosine on prooxidant-antioxidant status in heart tissue, plasma and erythrocytes of rats with isoproterenol-induced myocardial infarction. Pharmacol Rep 66: 81-86. https://doi.org/10.1016/j.pharep.2013.08.008
|
| [58] |
Peters V, Riedl E, Braunagel M, et al. (2014) Carnosine treatment in combination with ACE inhibition in diabetic rats. Regul Pept 194–195: 36-40. https://doi.org/10.1016/j.regpep.2014.09.005
|
| [59] |
Bao Y, Gao C, Hao W, et al. (2015) Effects of Dietary L-carnosine and Alpha-lipoic Acid on Growth Performance, Blood Thyroid Hormones and Lipid Profiles in Finishing Pigs. Asian-Australas J Anim Sci 28: 1465-1470. https://doi.org/10.5713/ajas.14.0604
|
| [60] |
Stegen S, Stegen B, Aldini G, et al. (2015) Plasma carnosine, but not muscle carnosine, attenuates high-fat diet-induced metabolic stress. Appl Physiol Nutr Metab 40: 868-876. https://doi.org/10.1139/apnm-2015-0042
|
| [61] |
Forsberg EA, Botusan IR, Wang J, et al. (2015) Carnosine decreases IGFBP1 production in db/db mice through suppression of HIF-1. J Endocrinol 225: 159-167. https://doi.org/10.1530/joe-14-0571
|
| [62] |
Wu T, Tao Y, Tsang F, et al. (2015) The effect of L-carnosine on the circadian resetting of clock genes in the heart of rats. Mol Biol Rep 42: 87-94. https://doi.org/10.1007/s11033-014-3745-x
|
| [63] |
Menini S, Iacobini C, Ricci C, et al. (2015) Protection from diabetes-induced atherosclerosis and renal disease by D-carnosine-octylester: effects of early vs late inhibition of advanced glycation end-products in Apoe-null mice. Diabetologia 58: 845-853. https://doi.org/10.1007/s00125-014-3467-6
|
| [64] |
Alsheblak MM, Elsherbiny NM, El-Karef A, et al. (2016) Protective effects of L-carnosine on CCl4 -induced hepatic injury in rats. Eur Cytokine Netw 27: 6-15. https://doi.org/10.1684/ecn.2016.0372
|
| [65] |
Ahshin-Majd S, Zamani S, Kiamari T, et al. (2016) Carnosine ameliorates cognitive deficits in streptozotocin-induced diabetic rats: Possible involved mechanisms. Peptides 86: 102-111. https://doi.org/10.1016/j.peptides.2016.10.008
|
| [66] |
Milewski K, Hilgier W, Fręśko I, et al. (2016) Carnosine Reduces Oxidative Stress and Reverses Attenuation of Righting and Postural Reflexes in Rats with Thioacetamide-Induced Liver Failure. Neurochem Res 41: 376-384. https://doi.org/10.1007/s11064-015-1821-9
|
| [67] |
Kumral A, Giriş M, Soluk-Tekkeşin M, et al. (2016) Beneficial effects of carnosine and carnosine plus vitamin E treatments on doxorubicin-induced oxidative stress and cardiac, hepatic, and renal toxicity in rats. Hum Exp Toxicol 35: 635-643. https://doi.org/10.1177/0960327115597468
|
| [68] |
Hasanein P, Kazemian-Mahtaj A, Khodadadi I (2016) Bioactive peptide carnosin protects against lead acetate-induced hepatotoxicity by abrogation of oxidative stress in rats. Pharm Biol 54: 1458-1464. https://doi.org/10.3109/13880209.2015.1104700
|
| [69] | Hasanein P, Teimuri-Far M (2015) Protective effect of bioactive peptide carnosine against lead-induced oxidative stress in kidney of rats. Cell Mol Biol (Noisy-le-grand) 61: 8-14. |
| [70] |
Fouad AA, Qutub HO, Al Rashed AS, et al. (2017) Therapeutic effect of carnosine in rat model of experimental liver carcinogenesis. Environ Toxicol Pharmacol 56: 10-14. https://doi.org/10.1016/j.etap.2017.08.021
|
| [71] |
Sun C, Wu Q, Zhang X, et al. (2017) Mechanistic Evaluation of the Protective Effect of Carnosine on Acute Lung Injury in Sepsis Rats. Pharmacology 100: 292-300. https://doi.org/10.1159/000479879
|
| [72] |
Albrecht T, Schilperoort M, Zhang S, et al. (2017) Carnosine Attenuates the Development of both Type 2 Diabetes and Diabetic Nephropathy in BTBR ob/ob Mice. Sci Rep 7: 44492. https://doi.org/10.1038/srep44492
|
| [73] |
Aydın AF, Bingül İ, Küçükgergin C, et al. (2017) Carnosine decreased oxidation and glycation products in serum and liver of high-fat diet and low-dose streptozotocin-induced diabetic rats. Int J Exp Pathol 98: 278-288. https://doi.org/10.1111/iep.12252
|
| [74] |
Fatih Aydın A, Küçükgergin C, Bingül İ, et al. (2017) Effect of Carnosine on Renal Function, Oxidation and Glycation Products in the Kidneys of High-Fat Diet/Streptozotocin-Induced Diabetic Rats. Exp Clin Endocrinol Diabetes 125: 282-289. https://doi.org/10.1055/s-0043-100117
|
| [75] |
Sahin S, Burukoglu Donmez D (2018) Effects of Carnosine (Beta-Alanyl-L-Histidine) in an Experimental Rat Model of Acute Kidney Injury Due to Septic Shock. Med Sci Monit 24: 305-316. https://doi.org/10.12659/msm.905181
|
| [76] |
Iacobini C, Menini S, Blasetti Fantauzzi C, et al. (2018) FL–926–16, a novel bioavailable carnosinase-resistant carnosine derivative, prevents onset and stops progression of diabetic nephropathy in db/db mice. Br J Pharmacol 175: 53-66. https://doi.org/10.1111/bph.14070
|
| [77] |
Deng J, Zhong YF, Wu YP, et al. (2018) Carnosine attenuates cyclophosphamide-induced bone marrow suppression by reducing oxidative DNA damage. Redox Biol 14: 1-6. https://doi.org/10.1016/j.redox.2017.08.003
|
| [78] |
Abplanalp W, Haberzettl P, Bhatnagar A, et al. (2019) Carnosine Supplementation Mitigates the Deleterious Effects of Particulate Matter Exposure in Mice. J Am Heart Assoc 8: e013041. https://doi.org/10.1161/jaha.119.013041
|
| [79] |
Liu XQ, Jiang L, Lei L, et al. (2020) Carnosine alleviates diabetic nephropathy by targeting GNMT, a key enzyme mediating renal inflammation and fibrosis. Clin Sci (Lond) 134: 3175-3193. https://doi.org/10.1042/cs20201207
|
| [80] |
Zhao J, Conklin DJ, Guo Y, et al. (2020) Cardiospecific Overexpression of ATPGD1 (Carnosine Synthase) Increases Histidine Dipeptide Levels and Prevents Myocardial Ischemia Reperfusion Injury. J Am Heart Assoc 9: e015222. https://doi.org/10.1161/jaha.119.015222
|
| [81] |
Everaert I, He J, Hanssens M, et al. (2020) Carnosinase-1 overexpression, but not aerobic exercise training, affects the development of diabetic nephropathy in BTBR ob/ob mice. Am J Physiol Renal Physiol 318: F1030-f1040. https://doi.org/10.1152/ajprenal.00329.2019
|
| [82] |
Qiu J, Albrecht T, Zhang S, et al. (2020) Human carnosinase 1 overexpression aggravates diabetes and renal impairment in BTBR(Ob/Ob) mice. J Mol Med (Berl) 98: 1333-1346. https://doi.org/10.1007/s00109-020-01957-0
|
| [83] |
Weigand T, Colbatzky F, Pfeffer T, et al. (2020) A Global Cndp1-Knock-Out Selectively Increases Renal Carnosine and Anserine Concentrations in an Age- and Gender-Specific Manner in Mice. Int J Mol Sci 21: 4887. https://doi.org/10.3390/ijms21144887
|
| [84] |
Ommati MM, Farshad O, Ghanbarinejad V, et al. (2020) The Nephroprotective Role of Carnosine Against Ifosfamide-Induced Renal Injury and Electrolytes Imbalance is Mediated Via the Regulation of Mitochondrial Function and Alleviation of Oxidative Stress. Drug Res (Stuttg) 70: 49-56. https://doi.org/10.1055/a-1017-5085
|
| [85] |
Yaqub LS, Ayo JO, Habibu B, et al. (2021) Thermoregulatory, oxidative stress and lipid responses in prepartum ewes administered with L-carnosine during the hot-dry season. Trop Anim Health Prod 53: 388. https://doi.org/10.1007/s11250-021-02832-x
|
| [86] |
Stefani GP, Capalonga L, da Silva LR, et al. (2021) Effects of aerobic and resistance exercise training associated with carnosine precursor supplementation on maximal strength and V̇O(2max) in rats with heart failure. Life Sci 282: 119816. https://doi.org/10.1016/j.lfs.2021.119816
|
| [87] | Riger NA, Trushina EN, Mustafina OK, et al. (2021) [Pathogenetic mechanisms for the development of hematological disorders in induced fatty liver disease in Wistar rats and assessment of the regulatory effects of carnosine and α-lipoic acid]. Vopr Pitan 90: 6-19. https://doi.org/10.33029/0042-8833-2021-90-3-6-19 |
| [88] |
Gonçalves LS, Sales LP, Saito TR, et al. (2021) Histidine dipeptides are key regulators of excitation-contraction coupling in cardiac muscle: Evidence from a novel CARNS1 knockout rat model. Redox Biol 44: 102016. https://doi.org/10.1016/j.redox.2021.102016
|
| [89] |
Schwank-Xu C, Forsberg E, Bentinger M, et al. (2021) L-Carnosine Stimulation of Coenzyme Q10 Biosynthesis Promotes Improved Mitochondrial Function and Decreases Hepatic Steatosis in Diabetic Conditions. Antioxidants (Basel) 10: 793. https://doi.org/10.3390/antiox10050793
|
| [90] | Nooh MM, Rizk SM, Saied NM, et al. (2021) Carnosine Remedial Effect on Fertility of Male Rats Receiving Cyclophosphamide, Hydroxydaunomycin, Oncovin and Prednisone (CHOP). Andrologia 53: e14233. https://doi.org/10.1111/and.14233 |
| [91] |
Zhu W, Li YY, Zeng HX, et al. (2021) Carnosine alleviates podocyte injury in diabetic nephropathy by targeting caspase-1-mediated pyroptosis. Int Immunopharmacol 101: 108236. https://doi.org/10.1016/j.intimp.2021.108236
|
| [92] |
Busa P, Lee SO, Huang N, et al. (2022) Carnosine Alleviates Knee Osteoarthritis and Promotes Synoviocyte Protection via Activating the Nrf2/HO-1 Signaling Pathway: An In-Vivo and In-Vitro Study. Antioxidants (Basel) 11: 1209. https://doi.org/10.3390/antiox11061209
|
| [93] |
Rodriguez-Niño A, Pastene DO, Hettler SA, et al. (2022) Influence of carnosine and carnosinase-1 on diabetes-induced afferent arteriole vasodilation: implications for glomerular hemodynamics. Am J Physiol Renal Physiol 323: F69-F80. https://doi.org/10.1152/ajprenal.00232.2021
|
| [94] |
Park J, Jang J, Cha SR, et al. (2022) L-carnosine Attenuates Bleomycin-Induced Oxidative Stress via NFκB Pathway in the Pathogenesis of Pulmonary Fibrosis. Antioxidants (Basel) 11: 2462. https://doi.org/10.3390/antiox11122462
|
| [95] |
Zharikov AY, Kalnitsky AS, Mazko ON, et al. (2023) Effect of Carnosine on the Activity of Matrix Metalloproteinase-2 and Oxidative Stress in the Kidneys in Experimental Urate Nephrolithiasis. Bull Exp Biol Med 174: 326-329. https://doi.org/10.1007/s10517-023-05701-9
|
| [96] |
Ommati MM, Sabouri S, Niknahad H, et al. (2023) Pulmonary inflammation, oxidative stress, and fibrosis in a mouse model of cholestasis: the potential protective properties of the dipeptide carnosine. Naunyn Schmiedebergs Arch Pharmacol 396: 1129-1142. https://doi.org/10.1007/s00210-023-02391-y
|
| [97] |
Zhang S, Li Y, Liu X, et al. (2023) Carnosine alleviates kidney tubular epithelial injury by targeting NRF2 mediated ferroptosis in diabetic nephropathy. Amino Acids 55: 1141-1155. https://doi.org/10.1007/s00726-023-03301-5
|
| [98] |
Rathor R, Srivastava S, Suryakumar G (2023) A Comparative Biochemical Study Between L-Carnosine and beta-Alanine in Amelioration of Hypobaric Hypoxia-Induced Skeletal Muscle Protein Loss. High Alt Med Biol 24: 302-311. https://doi.org/10.1089/ham.2023.0014
|
| [99] |
Luo X, Li Y, Wang B, et al. (2023) Carnosine alleviates cisplatin-induced acute kidney injury by targeting Caspase-1 regulated pyroptosis. Biomed Pharmacother 167: 115563. https://doi.org/10.1016/j.biopha.2023.115563
|
| [100] |
Berdaweel IA, Monroe TB, Alowaisi AA, et al. (2023) Iron scavenging and suppression of collagen cross-linking underlie antifibrotic effects of carnosine in the heart with obesity. Front Pharmacol 14: 1275388. https://doi.org/10.3389/fphar.2023.1275388
|
| [101] |
Moreto F, Garcia JL, Ferreira A, et al. (2024) Quantitative proteomics study of carnosine effect in an animal model of Western diet-induced nonalcoholic fatty liver disease. J Biochem Mol Toxicol 38: e23644. https://doi.org/10.1002/jbt.23644
|
| [102] |
Grandini NA, Costa MR, Gregolin CS, et al. (2024) Effects of carnosine supplementation on markers for the pathophysiological development of metabolic dysfunction-associated steatotic liver disease in a diet-induced model. Mol Cell Endocrinol 582: 112138. https://doi.org/10.1016/j.mce.2023.112138
|
| [103] |
D'Amato A, Altomare A, Gilardoni E, et al. (2024) A quantitative proteomic approach to evaluate the efficacy of carnosine in a murine model of chronic obstructive pulmonary disease (COPD). Redox Biol 77: 103374. https://doi.org/10.1016/j.redox.2024.103374
|
| [104] |
Drenjančević I, Stupin A, Jukić I, et al. (2024) Oral Carnosine Supplementation Preserves Vascular Function of Sprague Dawley Rats on a High-Salt Diet via Restored Antioxidative Defence. Nutrients 17: 36. https://doi.org/10.3390/nu17010036
|
| [105] |
Xu X, Pastene DO, Qiu J, et al. (2025) Influence of carnosine supplementation on disease progression in a rat model of focal segmental glomerulosclerosis. Am J Physiol Renal Physiol 328: F599-f607. https://doi.org/10.1152/ajprenal.00017.2024
|
| [106] |
Meng Y, Xian T, Kang G, et al. (2024) Effects of dietary l-carnosine supplementation on the growth, intestinal microbiota, and serum metabolome of fattening lambs. Front Vet Sci 11: 1525783. https://doi.org/10.3389/fvets.2024.1525783
|
| [107] |
Li XY, Gu XY, Li XM, et al. (2025) Supplementation with carnosine, a food-derived bioactive dipeptide, alleviates dexamethasone-induced oxidative stress and bone impairment via the NRF2 signaling pathway. J Sci Food Agric 105: 1091-1104. https://doi.org/10.1002/jsfa.13899
|
| [108] |
Tomonaga S, Tachibana T, Takagi T, et al. (2004) Effect of central administration of carnosine and its constituents on behaviors in chicks. Brain Res Bull 63: 75-82. https://doi.org/10.1016/j.brainresbull.2004.01.002
|
| [109] |
Tomonaga S, Tachibana T, Takahashi H, et al. (2005) Nitric oxide involves in carnosine-induced hyperactivity in chicks. Eur J Pharmacol 524: 84-88. https://doi.org/10.1016/j.ejphar.2005.09.008
|
| [110] | Zemke D, Krishnamurthy R, Majid A (2005) Carnosine is neuroprotective in a mouse model of stroke. Journal of Cerebral Blood Flow & Metabolism 25: S313-S313. https://doi.org/10.1038/sj.jcbfm.9591524.0313 |
| [111] |
Fedorova TN, Macletsova MG, Kulikov AV, et al. (2006) Carnosine protects from the oxidative stress induced by prenatal hypoxia. Dokl Biol Sci 408: 207-210. https://doi.org/10.1134/s001249660603001x
|
| [112] |
Dobrota D, Fedorova T, Stvolinsky S, et al. (2005) Carnosine protects the brain of rats and Mongolian gerbils against ischemic injury: after-stroke-effect. Neurochem Res 30: 1283-1288. https://doi.org/10.1007/s11064-005-8799-7
|
| [113] |
Jin CL, Yang LX, Wu XH, et al. (2005) Effects of carnosine on amygdaloid-kindled seizures in Sprague-Dawley rats. Neuroscience 135: 939-947. https://doi.org/10.1016/j.neuroscience.2005.06.066
|
| [114] |
Wu XH, Ding MP, Zhu-Ge ZB, et al. (2006) Carnosine, a precursor of histidine, ameliorates pentylenetetrazole-induced kindled seizures in rat. Neurosci Lett 400: 146-149. https://doi.org/10.1016/j.neulet.2006.02.031
|
| [115] |
Tsuneyoshi Y, Tomonaga S, Asechi M, et al. (2007) Central administration of dipeptides, beta-alanyl-BCAAs, induces hyperactivity in chicks. BMC Neurosci 8: 37. https://doi.org/10.1186/1471-2202-8-37
|
| [116] |
Rajanikant GK, Zemke D, Senut MC, et al. (2007) Carnosine is neuroprotective against permanent focal cerebral ischemia in mice. Stroke 38: 3023-3031. https://doi.org/10.1161/strokeaha.107.488502
|
| [117] |
Zhu YY, Zhu-Ge ZB, Wu DC, et al. (2007) Carnosine inhibits pentylenetetrazol-induced seizures by histaminergic mechanisms in histidine decarboxylase knock-out mice. Neurosci Lett 416: 211-216. https://doi.org/10.1016/j.neulet.2007.01.075
|
| [118] |
Tanida M, Gotoh H, Taniguchi H, et al. (2007) Effects of central injection of L-carnosine on sympathetic nerve activity innervating brown adipose tissue and body temperature in rats. Regul Pept 144: 62-71. https://doi.org/10.1016/j.regpep.2007.06.001
|
| [119] |
Tomonaga S, Yamane H, Onitsuka E, et al. (2008) Carnosine-induced antidepressant-like activity in rats. Pharmacol Biochem Behav 89: 627-632. https://doi.org/10.1016/j.pbb.2008.02.021
|
| [120] |
Min J, Senut MC, Rajanikant K, et al. (2008) Differential neuroprotective effects of carnosine, anserine, and N-acetyl carnosine against permanent focal ischemia. J Neurosci Res 86: 2984-2991. https://doi.org/10.1002/jnr.21744
|
| [121] |
Derave W, Jones G, Hespel P, et al. (2008) Creatine supplementation augments skeletal muscle carnosine content in senescence-accelerated mice (SAMP8). Rejuvenation Res 11: 641-647. https://doi.org/10.1089/rej.2008.0699
|
| [122] |
Kozan R, Sefil F, Bağirici F (2008) Anticonvulsant effect of carnosine on penicillin-induced epileptiform activity in rats. Brain Res 1239: 249-255. https://doi.org/10.1016/j.brainres.2008.08.019
|
| [123] |
Pekcetin C, Kiray M, Ergur BU, et al. (2009) Carnosine attenuates oxidative stress and apoptosis in transient cerebral ischemia in rats. Acta Biol Hung 60: 137-148. https://doi.org/10.1556/ABiol.60.2009.2.1
|
| [124] |
Feng ZY, Zheng XJ, Wang J (2009) Effects of carnosine on the evoked potentials in hippocampal CA1 region. J Zhejiang Univ Sci B 10: 505-511. https://doi.org/10.1631/jzus.B0820370
|
| [125] |
Aydín AF, Küskü-Kiraz Z, Doğru-Abbasoğlu S, et al. (2010) Effect of carnosine treatment on oxidative stress in serum, apoB-containing lipoproteins fraction and erythrocytes of aged rats. Pharmacol Rep 62: 733-739. https://doi.org/10.1016/s1734-1140(10)70331-5
|
| [126] |
Shen Y, He P, Fan YY, et al. (2010) Carnosine protects against permanent cerebral ischemia in histidine decarboxylase knockout mice by reducing glutamate excitotoxicity. Free Radic Biol Med 48: 727-735. https://doi.org/10.1016/j.freeradbiomed.2009.12.021
|
| [127] |
Tsai SJ, Kuo WW, Liu WH, et al. (2010) Antioxidative and anti-inflammatory protection from carnosine in the striatum of MPTP-treated mice. J Agric Food Chem 58: 11510-11516. https://doi.org/10.1021/jf103258p
|
| [128] |
Zhang X, Song L, Cheng X, et al. (2011) Carnosine pretreatment protects against hypoxia-ischemia brain damage in the neonatal rat model. Eur J Pharmacol 667: 202-207. https://doi.org/10.1016/j.ejphar.2011.06.003
|
| [129] |
Di Paola R, Impellizzeri D, Salinaro AT, et al. (2011) Administration of carnosine in the treatment of acute spinal cord injury. Biochem Pharmacol 82: 1478-1489. https://doi.org/10.1016/j.bcp.2011.07.074
|
| [130] |
Corona C, Frazzini V, Silvestri E, et al. (2011) Effects of dietary supplementation of carnosine on mitochondrial dysfunction, amyloid pathology, and cognitive deficits in 3xTg-AD mice. PLoS One 6: e17971. https://doi.org/10.1371/journal.pone.0017971
|
| [131] |
Faddah L, Baky NAA, Al-Rasheed NM, et al. (2012) Carnosine and cyclosporine A alleviate brain damage after traumatic brain injury in rats. Afr J Pharm Pharmacol 6: 3305-3312. https://doi.org/10.5897/AJPP12.533
|
| [132] | Ma J, Chen J, Bo S, et al. (2015) Protective effect of carnosine after chronic cerebral hypoperfusion possibly through suppressing astrocyte activation. Am J Transl Res 7: 2706-2715. |
| [133] |
Ma J, Xiong JY, Hou WW, et al. (2012) Protective effect of carnosine on subcortical ischemic vascular dementia in mice. CNS Neurosci Ther 18: 745-753. https://doi.org/10.1111/j.1755-5949.2012.00362.x
|
| [134] |
Çoban J, Bingül I, Yesil-Mizrak K, et al. (2013) Effects of carnosine plus vitamin E and betaine treatments on oxidative stress in some tissues of aged rats. Curr Aging Sci 6: 199-205. https://doi.org/10.2174/18746098112059990011
|
| [135] |
Herculano B, Tamura M, Ohba A, et al. (2013) β-alanyl-L-histidine rescues cognitive deficits caused by feeding a high fat diet in a transgenic mouse model of Alzheimer's disease. J Alzheimers Dis 33: 983-997. https://doi.org/10.3233/jad-2012-121324
|
| [136] |
Wang JP, Yang ZT, Liu C, et al. (2013) L-carnosine inhibits neuronal cell apoptosis through signal transducer and activator of transcription 3 signaling pathway after acute focal cerebral ischemia. Brain Res 1507: 125-133. https://doi.org/10.1016/j.brainres.2013.02.032
|
| [137] |
Bae ON, Majid A (2013) Role of histidine/histamine in carnosine-induced neuroprotection during ischemic brain damage. Brain Res 1527: 246-254. https://doi.org/10.1016/j.brainres.2013.07.004
|
| [138] |
Bae ON, Serfozo K, Baek SH, et al. (2013) Safety and efficacy evaluation of carnosine, an endogenous neuroprotective agent for ischemic stroke. Stroke 44: 205-212. https://doi.org/10.1161/strokeaha.112.673954
|
| [139] |
Park HS, Han KH, Shin JA, et al. (2014) The neuroprotective effects of carnosine in early stage of focal ischemia rodent model. J Korean Neurosurg Soc 55: 125-130. https://doi.org/10.3340/jkns.2014.55.3.125
|
| [140] |
Zhang H, Guo S, Zhang L, et al. (2014) Treatment with carnosine reduces hypoxia-ischemia brain damage in a neonatal rat model. Eur J Pharmacol 727: 174-180. https://doi.org/10.1016/j.ejphar.2014.01.023
|
| [141] |
Ji YS, Park JW, Heo H, et al. (2014) The neuroprotective effect of carnosine (β-alanyl-L-histidine) on retinal ganglion cell following ischemia-reperfusion injury. Curr Eye Res 39: 634-641. https://doi.org/10.3109/02713683.2013.855235
|
| [142] |
Inozemtsev AN, Berezhnoy DS, Fedorova TN, et al. (2014) The effect of the natural dipeptide carnosine on learning of rats under the conditions of negative reinforcement. Dokl Biol Sci 454: 16-18. https://doi.org/10.1134/s0012496614010177
|
| [143] |
Albayrak S, Atci İ B, Kalayci M, et al. (2015) Effect of carnosine, methylprednisolone and their combined application on irisin levels in the plasma and brain of rats with acute spinal cord injury. Neuropeptides 52: 47-54. https://doi.org/10.1016/j.npep.2015.06.004
|
| [144] |
Dai YJ, Wu DC, Feng B, et al. (2015) Protective effect of carnosine on febrile seizures in immature mice. Neurosci Lett 588: 95-100. https://doi.org/10.1016/j.neulet.2014.12.061
|
| [145] |
Macedo LW, Cararo JH, Maravai SG, et al. (2016) Acute Carnosine Administration Increases Respiratory Chain Complexes and Citric Acid Cycle Enzyme Activities in Cerebral Cortex of Young Rats. Mol Neurobiol 53: 5582-5590. https://doi.org/10.1007/s12035-015-9475-9
|
| [146] | Russo R, Adornetto A, Cavaliere F, et al. (2015) Intravitreal injection of forskolin, homotaurine, and L-carnosine affords neuroprotection to retinal ganglion cells following retinal ischemic injury. Mol Vis 21: 718-729. |
| [147] |
Zhang ZY, Sun BL, Yang MF, et al. (2015) Carnosine attenuates early brain injury through its antioxidative and anti-apoptotic effects in a rat experimental subarachnoid hemorrhage model. Cell Mol Neurobiol 35: 147-157. https://doi.org/10.1007/s10571-014-0106-1
|
| [148] |
Afshin-Majd S, Khalili M, Roghani M, et al. (2015) Carnosine exerts neuroprotective effect against 6-hydroxydopamine toxicity in hemiparkinsonian rat. Mol Neurobiol 51: 1064-1070. https://doi.org/10.1007/s12035-014-8771-0
|
| [149] |
Aydın AF, Çoban J, Doğan-Ekici I, et al. (2016) Carnosine and taurine treatments diminished brain oxidative stress and apoptosis in D-galactose aging model. Metab Brain Dis 31: 337-345. https://doi.org/10.1007/s11011-015-9755-0
|
| [150] |
Ma J, Bo SH, Lu XT, et al. (2016) Protective effects of carnosine on white matter damage induced by chronic cerebral hypoperfusion. Neural Regen Res 11: 1438-1444. https://doi.org/10.4103/1673-5374.191217
|
| [151] |
Baky NA, Fadda L, Al-Rasheed NM, et al. (2016) Neuroprotective effect of carnosine and cyclosporine-A against inflammation, apoptosis, and oxidative brain damage after closed head injury in immature rats. Toxicol Mech Methods 26: 1-10. https://doi.org/10.3109/15376516.2015.1070224
|
| [152] |
Banerjee S, Poddar MK (2016) Aging-induced changes in brain regional serotonin receptor binding: Effect of Carnosine. Neuroscience 319: 79-91. https://doi.org/10.1016/j.neuroscience.2016.01.032
|
| [153] | Al-Rasheed NM, Fadda L, Mohamed AM, et al. (2016) Regulating effect of carnosine and/or l-arginine on the expression of inflammatory molecules induced nephropathy in the hypoxic rat model. Braz Arch Biol Technol 59: e16150622. https://doi.org/10.1590/1678-4324-2016150622 |
| [154] |
Kaneko J, Enya A, Enomoto K, et al. (2017) Anserine (beta-alanyl-3-methyl-L-histidine) improves neurovascular-unit dysfunction and spatial memory in aged AβPPswe/PSEN1dE9 Alzheimer's-model mice. Sci Rep 7: 12571. https://doi.org/10.1038/s41598-017-12785-7
|
| [155] |
Zhao J, Shi L, Zhang LR (2017) Neuroprotective effect of carnosine against salsolinol-induced Parkinson's disease. Exp Ther Med 14: 664-670. https://doi.org/10.3892/etm.2017.4571
|
| [156] |
Keskin A, Kanar BG, Kanar RG, et al. (2017) An investigation on the impact of carnosine on the myocardium in lower extremity ischemia-reperfusion injury in rats. Int J Cardiovasc Acad 3: 109-113. https://doi.org/10.1016/j.ijcac.2016.11.054
|
| [157] |
Stvolinsky SL, Fedorova TN, Devyatov AA, et al. (2017) [A neuroprotective action of carnosine in conditions of experimental focal cerebral ischemia-reperfusion]. Zh Nevrol Psikhiatr Im S S Korsakova 117: 60-64. https://doi.org/10.17116/jnevro201711712260-64
|
| [158] |
Xie RX, Li DW, Liu XC, et al. (2017) Carnosine Attenuates Brain Oxidative Stress and Apoptosis After Intracerebral Hemorrhage in Rats. Neurochem Res 42: 541-551. https://doi.org/10.1007/s11064-016-2104-9
|
| [159] |
Aydin F, Kalaz EB, Kucukgergin C, et al. (2018) Carnosine Treatment Diminished Oxidative Stress and Glycation Products in Serum and Tissues of D-Galactose-Treated Rats. Curr Aging Sci 11: 10-15. https://doi.org/10.2174/1871530317666170703123519
|
| [160] |
Devyatov AA, Fedorova TN, Stvolinsky SL, et al. (2018) [Study of the neuroprotective effects of carnosine in an experimental model of focal cerebral ischemia/reperfusion]. Biomed Khim 64: 344-348. https://doi.org/10.18097/pbmc20186404344
|
| [161] |
Fedorova TN, Devyatov AA, Berezhnoi DS, et al. (2018) Oxidative Status in Different Areas of the Cerebral Cortex of Wistar Rats during Focal Ischemia and Its Modulation with Carnosine. Bull Exp Biol Med 165: 746-750. https://doi.org/10.1007/s10517-018-4256-x
|
| [162] | Wang AH, Ma Q, Wang X, et al. (2018) Protective effects of beef decoction rich in carnosine on cerebral ischemia injury by permanent middle cerebral artery occlusion in rats. Exp Ther Med 15: 1321-1329. https://doi.org/10.3892/etm.2017.5524 |
| [163] |
Barca A, Gatti F, Spagnolo D, et al. (2018) Responsiveness of Carnosine Homeostasis Genes in the Pancreas and Brain of Streptozotocin-Treated Mice Exposed to Dietary Carnosine. Int J Mol Sci 19: 1713. https://doi.org/10.3390/ijms19061713
|
| [164] |
Qi Z, Yu X, Xu P, et al. (2018) l-Homocarnosine, l-carnosine, and anserine attenuate brain oxidative damage in a pentylenetetrazole-induced epilepsy model of ovariectomized rats. 3 Biotech 8: 363. https://doi.org/10.1007/s13205-018-1357-1
|
| [165] |
Tiwari N, Bhatia P, Kumar A, et al. (2018) Potential of carnosine, a histamine precursor in rat model of bilateral common carotid artery occlusion-induced vascular dementia. Fundam Clin Pharmacol 32: 516-531. https://doi.org/10.1111/fcp.12376
|
| [166] |
Colín-Barenque L, Bizarro-Nevares P, González Villalva A, et al. (2018) Neuroprotective effect of carnosine in the olfactory bulb after vanadium inhalation in a mouse model. Int J Exp Pathol 99: 180-188. https://doi.org/10.1111/iep.12285
|
| [167] |
Bermúdez ML, Skelton MR, Genter MB (2018) Intranasal carnosine attenuates transcriptomic alterations and improves mitochondrial function in the Thy1-aSyn mouse model of Parkinson's disease. Mol Genet Metab 125: 305-313. https://doi.org/10.1016/j.ymgme.2018.08.002
|
| [168] |
Bermúdez ML, Seroogy KB, Genter MB (2019) Evaluation of Carnosine Intervention in the Thy1-aSyn Mouse Model of Parkinson's Disease. Neuroscience 411: 270-278. https://doi.org/10.1016/j.neuroscience.2019.05.026
|
| [169] |
Jain S, Kim ES, Kim D, et al. (2020) Comparative Cerebroprotective Potential of d- and l-Carnosine Following Ischemic Stroke in Mice. Int J Mol Sci 21: 3053. https://doi.org/10.3390/ijms21093053
|
| [170] |
Dai Z, Lu XY, Zhu WL, et al. (2020) Carnosine ameliorates age-related dementia via improving mitochondrial dysfunction in SAMP8 mice. Food Funct 11: 2489-2497. https://doi.org/10.1039/c9fo02453k
|
| [171] |
Ommati MM, Heidari R, Ghanbarinejad V, et al. (2020) The neuroprotective properties of carnosine in a mouse model of manganism is mediated via mitochondria regulating and antioxidative mechanisms. Nutr Neurosci 23: 731-743. https://doi.org/10.1080/1028415x.2018.1552399
|
| [172] |
Virdi JK, Bhanot A, Jaggi AS, et al. (2020) Investigation on beneficial role of l-carnosine in neuroprotective mechanism of ischemic postconditioning in mice: possible role of histidine histamine pathway. Int J Neurosci 130: 983-998. https://doi.org/10.1080/00207454.2020.1715393
|
| [173] |
Devyatov AA, Fedorova TN, Berezhnoy DS, et al. (2020) Mechanisms of Neuroprotective Action of Hesperetin and Carnosine in Focal Ischemia of the Brain in Rats. Bull Exp Biol Med 169: 242-245. https://doi.org/10.1007/s10517-020-04859-w
|
| [174] |
Attia H, Fadda L, Al-Rasheed N, et al. (2020) Carnosine and L-arginine attenuate the downregulation of brain monoamines and gamma aminobutyric acid; reverse apoptosis and upregulate the expression of angiogenic factors in a model of hemic hypoxia in rats. Naunyn Schmiedebergs Arch Pharmacol 393: 381-394. https://doi.org/10.1007/s00210-019-01738-8
|
| [175] |
Kim EH, Kim ES, Shin D, et al. (2021) Carnosine Protects against Cerebral Ischemic Injury by Inhibiting Matrix-Metalloproteinases. Int J Mol Sci 22: 7495. https://doi.org/10.3390/ijms22147495
|
| [176] |
Brown JM, Baker LS, Seroogy KB, et al. (2021) Intranasal Carnosine Mitigates α-Synuclein Pathology and Motor Dysfunction in the Thy1-aSyn Mouse Model of Parkinson's Disease. ACS Chem Neurosci 12: 2347-2359. https://doi.org/10.1021/acschemneuro.1c00096
|
| [177] |
Banerjee S, Mukherjee B, Poddar MK, et al. (2021) Carnosine improves aging-induced cognitive impairment and brain regional neurodegeneration in relation to the neuropathological alterations in the secondary structure of amyloid beta (Aβ). J Neurochem 158: 710-723. https://doi.org/10.1111/jnc.15357
|
| [178] |
Łochyński D, Pawlak M, Everaert I, et al. (2022) Motor Unit Fatigability following Chronic Carnosine Supplementation in Aged Rats. Nutrients 14: 514. https://doi.org/10.3390/nu14030514
|
| [179] | Arslan A, Acer N, Kesici H, et al. (2022) Stereological Study on the Effect of Carnosine on of Purkinje Cells in the Cerebellum of Rats Exposed to 900 MHz Electromagnetic Field. Turk Neurosurg 32: 618-624. https://doi.org/10.5137/1019-5149.JTN.35313-21.2 |
| [180] |
Peng D, Xia Q, Guan L, et al. (2022) Carnosine Improves Cognitive Impairment Through Promoting SIRT6 Expression and Inhibiting Endoplasmic Reticulum Stress in a Diabetic Encephalopathy Model. Rejuvenation Res 25: 79-88. https://doi.org/10.1089/rej.2022.0002
|
| [181] |
Tsuji T, Furuhara K, Gerasimenko M, et al. (2022) Oral Supplementation with L-Carnosine Attenuates Social Recognition Deficits in CD157KO Mice via Oxytocin Release. Nutrients 14: 803. https://doi.org/10.3390/nu14040803
|
| [182] |
Hegazy MA, Abdelmonsif DA, Zeitoun TM, et al. (2022) Swimming exercise versus L-carnosine supplementation for Alzheimer's dementia in rats: implication of circulating and hippocampal FNDC5/irisin. J Physiol Biochem 78: 109-124. https://doi.org/10.1007/s13105-021-00845-6
|
| [183] |
Ndolo RO, Yu L, Zhao Y, et al. (2023) Carnosine-Based Reversal of Diabetes-Associated Cognitive Decline via Activation of the Akt/mTOR Pathway and Modulation of Autophagy in a Rat Model of Type 2 Diabetes Mellitus. Dement Geriatr Cogn Disord 52: 156-168. https://doi.org/10.1159/000530605
|
| [184] |
Hu X, Fukui Y, Feng T, et al. (2023) Neuroprotective effects of carnosine in a mice stroke model concerning oxidative stress and inflammatory response. J Neurol Sci 447: 120608. https://doi.org/10.1016/j.jns.2023.120608
|
| [185] |
Rivi V, Caruso G, Caraci F, et al. (2024) Behavioral and transcriptional effects of carnosine in the central ring ganglia of the pond snail Lymnaea stagnalis. J Neurosci Res 102: e25371. https://doi.org/10.1002/jnr.25371
|
| [186] |
Shen J, Xu J, Wen Y, et al. (2024) Carnosine ameliorates postoperative cognitive dysfunction of aged rats by limiting astrocytes pyroptosis. Neurotherapeutics 21: e00359. https://doi.org/10.1016/j.neurot.2024.e00359
|
| [187] |
Chern H, Caruso G, Desaire H, et al. (2025) Carnosine Mitigates Cognitive Impairment and Dopamine Release in an Okadaic Acid-Induced Zebrafish Model with Alzheimer's Disease-like Symptoms. ACS Chem Neurosci 16: 790-801. https://doi.org/10.1021/acschemneuro.4c00596
|
| [188] |
Khama-Murad AX, Pavlinova LI, Mokrushin AA (2008) Neurotropic effect of exogenous L-carnosine in cultured slices of the olfactory cortex from rat brain. Bull Exp Biol Med 146: 1-3. https://doi.org/10.1007/s10517-008-0227-y
|
| [189] |
Solana-Manrique C, Sanz FJ, Martínez-Carrión G, et al. (2022) Antioxidant and Neuroprotective Effects of Carnosine: Therapeutic Implications in Neurodegenerative Diseases. Antioxidants (Basel) 11: 848. https://doi.org/10.3390/antiox11050848
|
| [190] |
Caruso G, Fresta CG, Musso N, et al. (2019) Carnosine Prevents Abeta-Induced Oxidative Stress and Inflammation in Microglial Cells: A Key Role of TGF-beta1. Cells 8: 64. https://doi.org/10.3390/cells8010064
|
| [191] |
Kubota M, Kobayashi N, Sugizaki T, et al. (2020) Carnosine suppresses neuronal cell death and inflammation induced by 6-hydroxydopamine in an in vitro model of Parkinson's disease. PLoS One 15: e0240448. https://doi.org/10.1371/journal.pone.0240448
|
| [192] |
Aloisi A, Barca A, Romano A, et al. (2013) Anti-aggregating effect of the naturally occurring dipeptide carnosine on abeta1-42 fibril formation. PLoS One 8: e68159. https://doi.org/10.1371/journal.pone.0068159
|
| [193] |
Attanasio F, Convertino M, Magno A, et al. (2013) Carnosine inhibits Abeta(42) aggregation by perturbing the H-bond network in and around the central hydrophobic cluster. Chembiochem 14: 583-592. https://doi.org/10.1002/cbic.201200704
|
| [194] |
Rivi V, Carota G, Tascedda F, et al. (2025) Carnosine modulates Aβ-induced transcriptional aberrations in murine microglial cells. Curr Res Pharmacol Drug Discov 8: 100221. https://doi.org/10.1016/j.crphar.2025.100221
|
| [195] |
Greco V, Naletova I, Ahmed IMM, et al. (2020) Hyaluronan-carnosine conjugates inhibit Aβ aggregation and toxicity. Sci Rep 10: 15998. https://doi.org/10.1038/s41598-020-72989-2
|
| [196] |
Distefano A, Caruso G, Oliveri V, et al. (2022) Neuroprotective Effect of Carnosine Is Mediated by Insulin-Degrading Enzyme. ACS Chem Neurosci 13: 1588-1593. https://doi.org/10.1021/acschemneuro.2c00201
|
| [197] |
Cardaci V, Di Pietro L, Zupan MC, et al. (2025) Characterizing oxidative stress induced by Abeta oligomers and the protective role of carnosine in primary mixed glia cultures. Free Radic Biol Med 229: 213-224. https://doi.org/10.1016/j.freeradbiomed.2025.01.030
|
| [198] |
Mizuno D, Kawahara M, Konoha-Mizuno K, et al. (2024) The Role of Zinc in the Development of Vascular Dementia and Parkinson's Disease and the Potential of Carnosine as Their Therapeutic Agent. Biomedicines 12: 1296. https://doi.org/10.3390/biomedicines12061296
|
| [199] |
Mizuno D, Konoha-Mizuno K, Mori M, et al. (2015) Protective activity of carnosine and anserine against zinc-induced neurotoxicity: a possible treatment for vascular dementia. Metallomics 7: 1233-1239. https://doi.org/10.1039/c5mt00049a
|
| [200] |
Kawahara M, Sadakane Y, Mizuno K, et al. (2020) Carnosine as a Possible Drug for Zinc-Induced Neurotoxicity and Vascular Dementia. Int J Mol Sci 21: 2570. https://doi.org/10.3390/ijms21072570
|
| [201] |
Khama-Murad A, Mokrushin AA, Pavlinova LI (2011) Neuroprotective properties of l-carnosine in the brain slices exposed to autoblood in the hemorrhagic stroke model in vitro. Regul Pept 167: 65-69. https://doi.org/10.1016/j.regpep.2010.11.007
|
| [202] |
Zhang L, Yao K, Fan Y, et al. (2012) Carnosine protects brain microvascular endothelial cells against rotenone-induced oxidative stress injury through histamine H1 and H2 receptors in vitro. Clin Exp Pharmacol Physiol 39: 1019-1025. https://doi.org/10.1111/1440-1681.12019
|
| [203] |
Takahashi S, Nakashima Y, Toda K (2009) Carnosine facilitates nitric oxide production in endothelial f-2 cells. Biol Pharm Bull 32: 1836-1839. https://doi.org/10.1248/bpb.32.1836
|
| [204] |
Spina-Purrello V, Giliberto S, Barresi V, et al. (2010) Modulation of PARP-1 and PARP-2 expression by L-carnosine and trehalose after LPS and INFgamma-induced oxidative stress. Neurochem Res 35: 2144-2153. https://doi.org/10.1007/s11064-010-0297-x
|
| [205] |
Ou-Yang L, Liu Y, Wang BY, et al. (2018) Carnosine suppresses oxygen-glucose deprivation/recovery-induced proliferation and migration of reactive astrocytes of rats in vitro. Acta Pharmacol Sin 39: 24-34. https://doi.org/10.1038/aps.2017.126
|
| [206] |
Fresta CG, Hogard ML, Caruso G, et al. (2017) Monitoring carnosine uptake by RAW 264.7 macrophage cells using microchip electrophoresis with fluorescence detection. Anal Methods 9: 402-408. https://doi.org/10.1039/C6AY03009B
|
| [207] |
Fresta CG, Chakraborty A, Wijesinghe MB, et al. (2018) Non-toxic engineered carbon nanodiamond concentrations induce oxidative/nitrosative stress, imbalance of energy metabolism, and mitochondrial dysfunction in microglial and alveolar basal epithelial cells. Cell Death Dis 9: 245. https://doi.org/10.1038/s41419-018-0280-z
|
| [208] |
Yamashita S, Sato M, Matsumoto T, et al. (2018) Mechanisms of carnosine-induced activation of neuronal cells. Biosci Biotechnol Biochem 82: 683-688. https://doi.org/10.1080/09168451.2017.1413325
|
| [209] |
Kramarenko GG, Markova ED, Ivanova-Smolenskaya IA, et al. (2001) Peculiarities of carnosine metabolism in a patient with pronounced homocarnosinemia. Bull Exp Biol Med 132: 996-999. https://doi.org/10.1023/a:1013687832424
|
| [210] |
Bessman SP, Baldwin R (1962) Imidazole aminoaciduria in cerebromacular degeneration. Science 135: 789-791. https://doi.org/10.1126/science.135.3506.789
|
| [211] |
Levenson J, Lindahl-Kiessling K, Rayner S (1964) Carnosine excretion in juvenile amaurotic idiocy. Lancet 2: 756-757. https://doi.org/10.1016/s0140-6736(64)92581-4
|
| [212] |
Fonteh AN, Harrington RJ, Tsai A, et al. (2007) Free amino acid and dipeptide changes in the body fluids from Alzheimer's disease subjects. Amino Acids 32: 213-224. https://doi.org/10.1007/s00726-006-0409-8
|
| [213] |
Chao de la Barca JM, Rondet-Courbis B, Ferré M, et al. (2020) A Plasma Metabolomic Profiling of Exudative Age-Related Macular Degeneration Showing Carnosine and Mitochondrial Deficiencies. J Clin Med 9: 631. https://doi.org/10.3390/jcm9030631
|
| [214] |
Balion CM, Benson C, Raina PS, et al. (2007) Brain type carnosinase in dementia: a pilot study. BMC Neurol 7: 38. https://doi.org/10.1186/1471-2377-7-38
|
| [215] |
Wassif WS, Sherwood RA, Amir A, et al. (1994) Serum carnosinase activities in central nervous system disorders. Clin Chim Acta 225: 57-64. https://doi.org/10.1016/0009-8981(94)90027-2
|
| [216] |
Virani SS, Alonso A, Aparicio HJ, et al. (2021) Heart Disease and Stroke Statistics-2021 Update: A Report From the American Heart Association. Circulation 143: e254-e743. https://doi.org/10.1161/cir.0000000000000950
|
| [217] |
Davis CK, Laud PJ, Bahor Z, et al. (2016) Systematic review and stratified meta-analysis of the efficacy of carnosine in animal models of ischemic stroke. J Cereb Blood Flow Metab 36: 1686-1694. https://doi.org/10.1177/0271678x16658302
|
| [218] |
Anderson NE, Broad JB, Bonita R (1995) Delays in hospital admission and investigation in acute stroke. BMJ 311: 162. https://doi.org/10.1136/bmj.311.6998.162
|
| [219] |
Stvolinsky S, Kukley M, Dobrota D, et al. (2000) Carnosine protects rats under global ischemia. Brain Res Bull 53: 445-448. https://doi.org/10.1016/s0361-9230(00)00366-x
|
| [220] |
Kopach O, Rusakov DA, Sylantyev S (2022) Multi-target action of β-alanine protects cerebellar tissue from ischemic damage. Cell Death Dis 13: 747. https://doi.org/10.1038/s41419-022-05159-z
|
| [221] |
Joshi P, Perni M, Limbocker R, et al. (2021) Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response. Commun Biol 4: 843. https://doi.org/10.1038/s42003-021-02218-7
|
| [222] |
Pulido A, Hulbert B, Giese H, et al. (2023) Copper Chelation via beta-alanine extends lifespan in a C. elegans model of Alzheimer's Disease. Brain Disorders 10: 100076. https://doi.org/10.1016/j.dscb.2023.100076
|
| [223] |
Javonillo DI, Tran KM, Phan J, et al. (2021) Systematic Phenotyping and Characterization of the 3xTg-AD Mouse Model of Alzheimer's Disease. Front Neurosci 15: 785276. https://doi.org/10.3389/fnins.2021.785276
|
| [224] |
Kulikova O, Troshev D, Berezhnoy D, et al. (2023) Neuroprotective Efficacy of a Nanomicellar Complex of Carnosine and Lipoic Acid in a Rat Model of Rotenone-Induced Parkinson's Disease. Antioxidants (Basel) 12: 1215. https://doi.org/10.3390/antiox12061215
|
| [225] |
Ali AN, Su L, Newton J, et al. (2025) Dietary Carnosine Supplementation in Healthy Human Volunteers: A Safety, Tolerability, Plasma and Brain Concentration Study. Nutrients 17: 2130. https://doi.org/10.3390/nu17132130
|
| [226] |
Schilly KM, Gunawardhana SM, Wijesinghe MB, et al. (2020) Biological applications of microchip electrophoresis with amperometric detection: in vivo monitoring and cell analysis. Anal Bioanal Chem 412: 6101-6119. https://doi.org/10.1007/s00216-020-02647-z
|
| [227] |
Toviwek B, Koonawootrittriron S, Suwanasopee T, et al. (2024) Why Bestatin Prefers Human Carnosinase 2 (CN2) to Human Carnosinase 1 (CN1). J Phys Chem B 128: 11876-11884. https://doi.org/10.1021/acs.jpcb.4c05571
|
| [228] |
Peppers SC, Lenney JF (1988) Bestatin inhibition of human tissue carnosinase, a non-specific cytosolic dipeptidase. Biol Chem Hoppe Seyler 369: 1281-1286. https://doi.org/10.1515/bchm3.1988.369.2.1281
|
| [229] |
Qiu J, Hauske SJ, Zhang S, et al. (2019) Identification and characterisation of carnostatine (SAN9812), a potent and selective carnosinase (CN1) inhibitor with in vivo activity. Amino Acids 51: 7-16. https://doi.org/10.1007/s00726-018-2601-z
|
| [230] |
Toviwek B, Suwanasopee T, Koonawootrittriron S, et al. (2023) Binding Modes of Carnostatine, Homocarnosine, and Ophidine to Human Carnosinase 1. ACS Omega 8: 42966-42975. https://doi.org/10.1021/acsomega.3c06139
|
| [231] |
Vistoli G, Orioli M, Pedretti A, et al. (2009) Design, synthesis, and evaluation of carnosine derivatives as selective and efficient sequestering agents of cytotoxic reactive carbonyl species. ChemMedChem 4: 967-975. https://doi.org/10.1002/cmdc.200800433
|
| [232] |
Masuoka N, Yoshimine C, Hori M, et al. (2019) Effects of Anserine/Carnosine Supplementation on Mild Cognitive Impairment with APOE4. Nutrients 11: 1626. https://doi.org/10.3390/nu11071626
|
| [233] |
Szcześniak D, Budzeń S, Kopeć W, et al. (2014) Anserine and carnosine supplementation in the elderly: Effects on cognitive functioning and physical capacity. Arch Gerontol Geriatr 59: 485-490. https://doi.org/10.1016/j.archger.2014.04.008
|
| [234] |
Rokicki J, Li L, Imabayashi E, et al. (2015) Daily Carnosine and Anserine Supplementation Alters Verbal Episodic Memory and Resting State Network Connectivity in Healthy Elderly Adults. Front Aging Neurosci 7: 219. https://doi.org/10.3389/fnagi.2015.00219
|
| [235] |
Hisatsune T, Kaneko J, Kurashige H, et al. (2016) Effect of Anserine/Carnosine Supplementation on Verbal Episodic Memory in Elderly People. J Alzheimers Dis 50: 149-159. https://doi.org/10.3233/jad-150767
|
| [236] |
Katakura Y, Totsuka M, Imabayashi E, et al. (2017) Anserine/Carnosine Supplementation Suppresses the Expression of the Inflammatory Chemokine CCL24 in Peripheral Blood Mononuclear Cells from Elderly People. Nutrients 9: 1199. https://doi.org/10.3390/nu9111199
|
| [237] |
Ding Q, Tanigawa K, Kaneko J, et al. (2018) Anserine/Carnosine Supplementation Preserves Blood Flow in the Prefrontal Brain of Elderly People Carrying APOE e4. Aging Dis 9: 334-345. https://doi.org/10.14336/ad.2017.0809
|
| [238] |
Goto K, Maemura H, Takamatsu K, et al. (2011) Hormonal responses to resistance exercise after ingestion of carnosine and anserine. J Strength Cond Res 25: 398-405. https://doi.org/10.1519/JSC.0b013e3181bac43c
|