Metformin intake associates with better cognitive function in patients with Huntington's disease

David Hervás, Victoria Fornés-Ferrer, Ana Pilar Gómez-Escribano, María Dolores Sequedo, Carmen Peiró, José María Millán, Rafael P Vázquez-Manrique, David Hervás, Victoria Fornés-Ferrer, Ana Pilar Gómez-Escribano, María Dolores Sequedo, Carmen Peiró, José María Millán, Rafael P Vázquez-Manrique

Abstract

Huntington's disease (HD) is an inherited, dominant neurodegenerative disorder caused by an abnormal expansion of CAG triplets in the huntingtin gene (htt). Despite extensive efforts to modify the progression of HD thus far only symptomatic treatment is available. Recent work suggests that treating invertebrate and mice HD models with metformin, a well-known AMPK activator which is used worldwide to treat type 2-diabetes, reduces mutant huntingtin from cells and alleviates many of the phenotypes associated to HD. Herein we report statistical analyses of a sample population of participants in the Enroll-HD database, a world-wide observational study on HD, to assess the effect of metformin intake in HD patients respect to cognitive status using linear models. This cross-sectional study shows for the first time that the use of metformin associates with better cognitive function in HD patients.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Partial dependence plots showing the…
Fig 1. Partial dependence plots showing the interaction between metformin intake and HD status regarding cognitive scores.
(A-F) Result of the analysis of the different cognitive tests. G Graph showing the result of the analysis of all cognitive tests (Cognitive Score). These plots are produced using the estimates from the fitted linear regression models, so cognitive values are adjusted for age, gender, BMI and ISCED. The p-values included are assessing the effect of the interactions, that is, the differential effect of metformin intake in HD-patients compared to controls.
Fig 2. Partial dependence plot showing the…
Fig 2. Partial dependence plot showing the interaction between metformin intake and motor impairment (UHDRS Motorscore).
Result of the analysis of the UHDRS Motorscore. This plot was produced using the estimates from the fitted linear regression models, so the UHDRS Motorscore values are adjusted for age, gender, BMI and ISCED. The p-value is assessing the effect of the interaction, that is, the differential effect of metformin intake in HD-patients compared to controls.

References

    1. Schulte J, Littleton JT (2011) The biological function of the Huntingtin protein and its relevance to Huntington's Disease pathology. Curr Trends Neurol 5: 65–78.
    1. Hatters DM (2008) Protein misfolding inside cells: the case of huntingtin and Huntington's disease. IUBMB Life 60: 724–728. doi:
    1. Arrasate M, Finkbeiner S (2012) Protein aggregates in Huntington's disease. Exp Neurol 238: 1–11. doi:
    1. Martinez-Horta S, Perez-Perez J, van Duijn E, Fernandez-Bobadilla R, Carceller M, Pagonabarraga J, et al. (2016) Neuropsychiatric symptoms are very common in premanifest and early stage Huntington's Disease. Parkinsonism Relat Disord.
    1. Paulsen JS, Langbehn DR, Stout JC, Aylward E, Ross CA, Nance M, et al. (2008) Detection of Huntington's disease decades before diagnosis: the Predict-HD study. J Neurol Neurosurg Psychiatry 79: 874–880. doi:
    1. Paulsen JS, Zhao H, Stout JC, Brinkman RR, Guttman M, Ross CA, et al. (2001) Clinical markers of early disease in persons near onset of Huntington's disease. Neurology 57: 658–662.
    1. Tabrizi SJ, Scahill RI, Durr A, Roos RA, Leavitt BR, Jones R, et al. (2011) Biological and clinical changes in premanifest and early stage Huntington's disease in the TRACK-HD study: the 12-month longitudinal analysis. Lancet Neurol 10: 31–42. doi:
    1. Vazquez-Manrique RP, Farina F, Cambon K, Sequedo MD, Parker AJ, Millán JM, et al. (2015) AMPK activation protects from neuronal dysfunction and vulnerability across nematode, cellular and mouse models of Huntington's disease. Hum Mol Genet.
    1. Ma TC, Buescher JL, Oatis B, Funk JA, Nash AJ, Carrier RL, et al. (2007) Metformin therapy in a transgenic mouse model of Huntington's disease. Neurosci Lett 411: 98–103. doi:
    1. Parker JA, Arango M, Abderrahmane S, Lambert E, Tourette C, Catoire H, et al. (2005) Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat Genet 37: 349–350. doi:
    1. Trettel F, Rigamonti D, Hilditch-Maguire P, Wheeler VC, Sharp AH, Persichetti F, et al. (2000) Dominant phenotypes produced by the HD mutation in STHdh(Q111) striatal cells. Hum Mol Genet 9: 2799–2809.
    1. Chen L, Shu Y, Liang X, Chen EC, Yee SW, Zur AA, et al. (2014) OCT1 is a high-capacity thiamine transporter that regulates hepatic steatosis and is a target of metformin. Proc Natl Acad Sci U S A 111: 9983–9988. doi:
    1. Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, Albright RA, et al. (2014) Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 510: 542–546. doi:
    1. Ju TC, Chen HM, Chen YC, Chang CP, Chang C, Chern Y, et al. (2014) AMPK-alpha1 functions downstream of oxidative stress to mediate neuronal atrophy in Huntington's disease. Biochim Biophys Acta 1842: 1668–1680. doi:
    1. Ju TC, Chen HM, Lin JT, Chang CP, Chang WC, Kang JJ, et al. (2011) Nuclear translocation of AMPK-alpha1 potentiates striatal neurodegeneration in Huntington's disease. J Cell Biol 194: 209–227. doi:
    1. Velazquez EM, Mendoza S, Hamer T, Sosa F, Glueck CJ (1994) Metformin therapy in polycystic ovary syndrome reduces hyperinsulinemia, insulin resistance, hyperandrogenemia, and systolic blood pressure, while facilitating normal menses and pregnancy. Metabolism 43: 647–654.
    1. Tamayo T, Rosenbauer J, Wild SH, Spijkerman AM, Baan C, Forouhi NG, et al. (2014) Diabetes in Europe: an update. Diabetes Res Clin Pract 103: 206–217. doi:
    1. Zilliox LA, Chadrasekaran K, Kwan JY, Russell JW (2016) Diabetes and Cognitive Impairment. Curr Diab Rep 16: 87 doi:
    1. Rojas LB, Gomes MB (2013) Metformin: an old but still the best treatment for type 2 diabetes. Diabetol Metab Syndr 5: 6 doi:
    1. Lexis CP, van der Horst-Schrivers AN, Lipsic E, Valente MA, Muller Kobold AC, de Boer RA, et al. (2015) The effect of metformin on cardiovascular risk profile in patients without diabetes presenting with acute myocardial infarction: data from the Glycometabolic Intervention as adjunct to Primary Coronary Intervention in ST Elevation Myocardial Infarction (GIPS-III) trial. BMJ Open Diabetes Res Care 3: e000090 doi:
    1. Li D (2011) Metformin as an antitumor agent in cancer prevention and treatment. J Diabetes 3: 320–327. doi:
    1. Pryor R, Cabreiro F (2015) Repurposing metformin: an old drug with new tricks in its binding pockets. Biochem J 471: 307–322. doi:
    1. Lee SK, Lee JO, Kim JH, Kim SJ, You GY, Moon JW, et al. (2011) Metformin sensitizes insulin signaling through AMPK-mediated PTEN down-regulation in preadipocyte 3T3-L1 cells. J Cell Biochem 112: 1259–1267. doi:
    1. Sarabia V, Lam L, Burdett E, Leiter LA, Klip A (1992) Glucose transport in human skeletal muscle cells in culture. Stimulation by insulin and metformin. J Clin Invest 90: 1386–1395. doi:
    1. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F (2012) Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond) 122: 253–270.
    1. El-Mir MY, Nogueira V, Fontaine E, Averet N, Rigoulet M, Leverve X (2000) Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J Biol Chem 275: 223–228.
    1. Lu M, Su C, Qiao C, Bian Y, Ding J, Hu G. (2016) Metformin Prevents Dopaminergic Neuron Death in MPTP/P-Induced Mouse Model of Parkinson's Disease via Autophagy and Mitochondrial ROS Clearance. Int J Neuropsychopharmacol.
    1. Aziz NA, van der Burg JM, Landwehrmeyer GB, Brundin P, Stijnen T, EHDI Study Group, et al. (2008) Weight loss in Huntington disease increases with higher CAG repeat number. Neurology 71: 1506–1513. doi:
    1. Mochel F, Charles P, Seguin F, Barritault J, Coussieu C, Perin L, et al. (2007) Early energy deficit in Huntington disease: identification of a plasma biomarker traceable during disease progression. PLoS One 2: e647 doi:
    1. Popovic V, Svetel M, Djurovic M, Petrovic S, Doknic M, Pekic S, et al. (2004) Circulating and cerebrospinal fluid ghrelin and leptin: potential role in altered body weight in Huntington's disease. Eur J Endocrinol 151: 451–455.
    1. van der Burg JM, Winqvist A, Aziz NA, Maat-Schieman ML, Roos RA, Bates GP, et al. (2011) Gastrointestinal dysfunction contributes to weight loss in Huntington's disease mice. Neurobiol Dis 44: 1–8. doi:
    1. Huntington-Study-Group (1996) Unified Huntington's Disease Rating Scale: reliability and consistency. Huntington Study Group. Mov Disord 11: 136–142. doi:

Source: PubMed

Подписаться