Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1639 2022-06-22 07:58:07 |
2 layout Meta information modification 1639 2022-06-22 08:10:38 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kouvari, M.;  Sergi, D.;  D’cunha, N.M.;  Bulman, A.;  Panagiotakos, D.B.;  Naumovski, N. Non-Alcoholic Fatty Liver Disease and Cognition. Encyclopedia. Available online: https://encyclopedia.pub/entry/24317 (accessed on 24 April 2024).
Kouvari M,  Sergi D,  D’cunha NM,  Bulman A,  Panagiotakos DB,  Naumovski N. Non-Alcoholic Fatty Liver Disease and Cognition. Encyclopedia. Available at: https://encyclopedia.pub/entry/24317. Accessed April 24, 2024.
Kouvari, Matina, Domenico Sergi, Nathan M. D’cunha, Amanda Bulman, Demosthenes B. Panagiotakos, Nenad Naumovski. "Non-Alcoholic Fatty Liver Disease and Cognition" Encyclopedia, https://encyclopedia.pub/entry/24317 (accessed April 24, 2024).
Kouvari, M.,  Sergi, D.,  D’cunha, N.M.,  Bulman, A.,  Panagiotakos, D.B., & Naumovski, N. (2022, June 22). Non-Alcoholic Fatty Liver Disease and Cognition. In Encyclopedia. https://encyclopedia.pub/entry/24317
Kouvari, Matina, et al. "Non-Alcoholic Fatty Liver Disease and Cognition." Encyclopedia. Web. 22 June, 2022.
Non-Alcoholic Fatty Liver Disease and Cognition
Edit

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease globally, affecting approximately 25% of the general population. NAFLD occurs in the absence of excessive alcohol consumption and is closely associated with metabolic syndrome (MetS) and its components.

cognition liver steatosis non-alcoholic fatty liver disease

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease globally, affecting approximately 25% of the general population [1]. NAFLD rates are rising in parallel with the pandemics of obesity and type 2 diabetes mellitus (T2DM) [2]. NAFLD occurs in the absence of excessive alcohol consumption and is closely associated with metabolic syndrome (MetS) and its components [3]. Indeed, it represents the hepatic manifestation of the MetS. Additionally, NAFLD has metabolic and cardiovascular consequences that have been linked with metabolic complications, chronic kidney disease, cardiovascular disease (CVD), and malignancies, contributing to higher mortality. The latter is common in people with non-alcoholic steatohepatitis (NASH), which is characterised by both hepatic steatosis and inflammation [4]. All of these conditions related to NAFLD share low-grade inflammation and oxidative stress as common features, which, in turn, also play a role in extrahepatic diseases. In this context, the literature suggests an interaction between the liver and brain, adding one more indication regarding the intercorrelation between neurological and metabolic systems [5].

2. Observational Studies on the Association between NAFLD and Cognition

Within the last decade, only 11 observational studies have evaluated the association between NAFLD and cognition, with the vast majority published in the last 4 years [6][7][8][9][10][11][12][13][14][15][16][17][18][19]. The findings of these studies are summarised in Table 1. Five studies prospectively examined this association, presenting contradicting outcomes. The CARDIA study included middle-aged participants from the USA and revealed that the presence of NAFLD did not significantly affect the cognitive decline in a 5-year follow-up [7]. On the other side, a prospective study from a Chinese cohort revealed that NAFLD participants had greater cognitive decline compared with their free-of-NAFLD counterparts during a 4-year follow-up, especially within the subgroup of middle-aged individuals. Three studies in Europe—one in Italy, one in Germany, and one in Sweden—investigated the effect of NAFLD on long-term dementia risk, revealing either positive [17] or neutral [16][19] associations. However, in the case of the study from Sweden, once the histological features of liver steatosis were included in the model that predicted dementia risk, this significantly increased its predictive ability [19]. The remaining nine studies had a cross-sectional [8][9][10][11][13][15] or case-control [12][14][18] design. Only one of them, a sub-analysis in the Framingham study, revealed non-significant associations between NAFLD and cognition assessed via neuropsychological tests [9]. Four studies [6][11][12][14] evaluated the association between NAFLD and general cognitive performance using multiple neuropsychological tests. All of these studies reported that individuals with NAFLD had significantly lower cognitive performance overall, measured via different validated questionnaires. Three studies revealed significantly lower processing speed and attention in people with NAFLD compared with non-NAFLD controls [8][14][15]. Memory and learning domains were examined in three studies revealing mixed outcomes [9][14][15]. In particular, one study reported lower performance in memory and learning test scores within the NAFLD group [15], while no significant associations were identified in the other two studies. In one study [13], the association between NAFLD and the language domain of cognitive performance was evaluated, revealing lower scores in the presence of this pathophysiological condition. Three studies investigated the role of NAFLD on visuospatial perception [9][11][14]. Two out of the three studies found poorer visuospatial perception in people with NAFLD [11][14]. In addition to this, NAFLD was associated with lower scores in abstraction, figural creation, and mental flexibility, as revealed by three studies [8][9][14]. Brain aging was assessed in one study with individuals that were free of NAFLD having lower aging of the brain compared with their NAFLD counterparts [10].
Table 1. Characteristics of selected observational studies on the association between non-alcoholic fatty liver disease and cognitive function (n = 11).

3. Pathogenetic Mechanisms Underpinning the Development of NAFLD

NAFLD represents the hepatic manifestation of the MetS. From a pathogenetic perspective, it arises as a consequence of an imbalance between triglyceride synthesis, fatty acid supply to the liver, triglyceride export via very-low-density lipoprotein (VLDL), and fatty acid oxidative capacity by the liver. This imbalance ultimately leads to the intrahepatic accumulation of triglycerides. Remarkably, all of these processes occur in a state of systemic insulin resistance and compensatory hyperinsulinemia [20]. Indeed, in the context of obesity and MetS, the underlying state of low-grade chronic inflammation promotes insulin resistance in metabolically active tissues, including adipose tissue [21]. Adipose tissue insulin resistance leads to an increase in fatty acid spill over from adipocytes due to the disinhibition of lipolysis [22]. This contributes to fatty acid oversupply in the liver, thereby fuelling the accumulation of triglycerides as lipid droplets within the hepatocytes. In support of the importance of this process in promoting NAFLD, the knockdown of fatty acid transport protein (FATP) 5, a fatty acid transporter expressed by hepatocytes, reversed steatosis in mice [23]. Another key pathogenetic process underpinning the accumulation of intrahepatic triglycerides is enhanced de novo lipogenesis, which is also a direct consequence of insulin resistance and, particularly, the compensatory hyperinsulinemia. De novo lipogenesis is under the transcriptional control of sterol regulatory element-binding protein 1c (SREBP1c), which, in turn, is regulated by insulin. However, de novo lipogenesis is not inhibited by insulin resistance. Instead, it is enhanced by hyperinsulinemia, which explains the increased hepatic de novo lipogenesis under insulin-resistant conditions [24]. A further pathogenetic mechanism underpinning the onset and progression of NAFLD is represented by impaired fatty acid oxidation, a metabolic pathway under the control of PPARα. In support of the role of this nuclear receptor and impaired fatty acid catabolism in the pathogenesis of NAFLD, PPARα-deficient ob/ob mice manifest more severe hepatic steatosis compared to their littermates due to decreased fatty acid oxidation [25].
However, data in humans relative to the relationship between fatty acid oxidation and NAFLD is controversial, with studies reporting either an increase, a decrease, or no changes in lipid catabolism in individuals with NAFLD [26]. Nevertheless, it must not be overlooked that even when there is an increase in fatty acid oxidation, the magnitude of such an increase may not be sufficient to cope with enhanced fatty acid supply. This compensatory response marked by an increase in fatty acid oxidation promotes an increase in reactive oxygen species (ROS), which further contributes to the mitochondrial dysfunction that characterises NAFLD [27]. In turn, mitochondrial dysfunction, due to the role of these organelles in fatty acid β-oxidation, contributes to both the inability of hepatocytes to cope with increased fatty acid supply as well as ROS production [28]. Finally, defects in triglycerides exported from the liver also contribute to NAFLD. In parallel with increased fatty acid oxidation, enhanced triglyceride export as part of VLDL also represents a mechanism to decrease hepatic lipid accumulation [29]. Once produced in the endoplasmic reticulum of hepatocytes, VLDL is channelled towards the Golgi apparatus, where mature VLDL are formed.

References

  1. Kanwal, F.; Shubrook, J.H.; Younossi, Z.; Natarajan, Y.; Bugianesi, E.; Rinella, M.E.; Harrison, S.A.; Mantzoros, M.; Pfotenhauer, K.; Klein, S.; et al. Preparing for the NASH epidemic: A call to action. Metabolism 2021, 122, 154822.
  2. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Obesity and nonalcoholic fatty liver disease: From pathophysiology to therapeutics. Metabolism 2018, 92, 82–97.
  3. Muzurović, E.; Mikhailidis, D.P.; Mantzoros, C. Non-alcoholic fatty liver disease, insulin resistance, metabolic syndrome and their association with vascular risk. Metabolism 2021, 119, 154770.
  4. Mantovani, A.; Scorletti, E.; Mosca, A.; Alisi, A.; Byrne, C.D.; Targher, G. Complications, morbidity and mortality of nonalcoholic fatty liver disease. Metabolism 2020, 111, 154170.
  5. George, E.S.; Sood, S.; Daly, R.M.; Tan, S.-Y. Is there an association between non-alcoholic fatty liver disease and cognitive function? A systematic review. BMC Geriatr. 2022, 22, 47.
  6. Liu, Q.; Liu, C.; Hu, F.; Deng, X.; Zhang, Y. Non-alcoholic Fatty Liver Disease and Longitudinal Cognitive Changes in Middle-Aged and Elderly Adults. Front. Med. 2022, 8, 2642.
  7. Gerber, Y.; VanWagner, L.B.; Yaffe, K.; Terry, J.G.; Rana, J.S.; Reis, J.P.; Sidney, S. Non-alcoholic fatty liver disease and cognitive function in middle-aged adults: The CARDIA study. BMC Gastroenterol. 2021, 21, 96.
  8. Weinstein, A.; de Avila, L.; Paik, J.; Golabi, P.; Escheik, C.; Gerber, L.; Younossi, Z.M. Cognitive Performance in Individuals With Non-Alcoholic Fatty Liver Disease and/or Type 2 Diabetes Mellitus. J. Psychosom. Res. 2018, 59, 567–574.
  9. Weinstein, G.; Davis-Plourde, K.; Himali, J.J.; Zelber-Sagi, S.; Beiser, A.S.; Seshadri, S. P2-562: Non-Alcoholic fatty liver disease, liver fibrosis score and cognitive function in middle-aged adults: The Framingham study. Liver Int. 2019, 39, 1713–1721.
  10. Weinstein, G.; Zelber-Sagi, S.; Preis, S.R.; Beiser, A.; DeCarli, C.; Speliotes, E.K.; Satizabal, C.L.; Vasan, R.S.; Seshadri, S. Association of Nonalcoholic Fatty Liver Disease With Lower Brain Volume in Healthy Middle-aged Adults in the Framingham Study. JAMA Neurol. 2018, 75, 97–104.
  11. Filipović, B.; Marković, O.; Đurić, V.; Filipović, B. Cognitive Changes and Brain Volume Reduction in Patients with Nonalcoholic Fatty Liver Disease. Can. J. Gastroenterol. Hepatol. 2018, 2018, 1–6.
  12. Tuttolomondo, A.; Petta, S.; Casuccio, A.; Maida, C.; Della Corte, V.; Daidone, M.; Di Raimondo, D.; Pecoraro, R.; Fonte, R.; Cirrincione, A.; et al. Reactive hyperemia index (RHI) and cognitive performance indexes are associated with histologic markers of liver disease in subjects with non-alcoholic fatty liver disease (NAFLD): A case control study. Cardiovasc. Diabetol. 2018, 17, 28.
  13. Takahashi, A.; Kono, S.; Wada, A.; Oshima, S.; Abe, K.; Imaizumi, H.; Fujita, M.; Hayashi, M.; Okai, K.; Miura, I.; et al. Reduced brain activity in female patients with non-alcoholic fatty liver disease as measured by near-infrared spectroscopy. PLoS ONE 2017, 12, e0174169.
  14. Celikbilek, A.; Celikbilek, M.; Bozkurt, G. Cognitive assessment of patients with nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2018, 30, 944–950.
  15. Seo, S.W.; Gottesman, R.F.; Clark, J.M.; Hernaez, R.; Chang, Y.; Kim, C.; Ha, K.H.; Guallar, E.; Lazo, M. Nonalcoholic fatty liver disease is associated with cognitive function in adults. Neurology 2016, 86, 1136–1142.
  16. Labenz, C.; Kostev, K.; Kaps, L.; Galle, P.R.; Schattenberg, J.M. Incident dementia in elderly patients with nonalcoholic fatty liver disease in Germany. Dig. Dis. Sci. 2021, 61, 3179–3185.
  17. Solfrizzi, V.; Scafato, E.; Custodero, C.; Loparco, F.; Ciavarella, A.; Panza, F.; Seripa, D.; Imbimbo, B.P.; Lozupone, M.; Napoli, N.; et al. Liver fibrosis score, physical frailty, and the risk of dementia in older adults: The Italian Longitudinal Study on Aging. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2020, 6, e12065.
  18. Elliott, C.; Frith, J.; Day, C.P.; Jones, D.E.J.; Newton, J.L. Functional Impairment in Alcoholic Liver Disease and Non-alcoholic Fatty Liver Disease Is Significant and Persists over 3 Years of Follow-Up. Am. J. Dig. Dis. 2013, 58, 2383–2391.
  19. Shang, Y.; Nasr, P.; Ekstedt, M.; Widman, L.; Stål, P.; Hultcrantz, R.; Kechagias, S.; Hagström, H. Non-alcoholic fatty liver disease does not increase dementia risk although histology data might improve risk prediction. JHEP Rep. 2020, 3, 100218.
  20. Ilan, Y. Leaky gut and the liver: A role for bacterial translocation in nonalcoholic steatohepatitis. World J. Gastroenterol. 2012, 18, 2609–2618.
  21. Zhu, B.; Guo, X.; Xu, H.; Jiang, B.; Li, H.; Wang, Y.; Yin, Q.; Zhou, T.; Cai, J.J.; Glaser, S.; et al. Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. Lab. Investig. 2021, 101, 328–340.
  22. Samala, N.; Tersey, S.A.; Chalasani, N.; Anderson, R.M.; Mirmira, R.G. Molecular mechanisms of nonalcoholic fatty liver disease: Potential role for 12-lipoxygenase. J. Diabetes Its Complicat. 2017, 31, 1630–1637.
  23. Doege, H.; Grimm, D.; Falcon, A.; Tsang, B.; Storm, T.A.; Xu, H.; Ortegon, A.M.; Kazantzis, M.; Kay, M.A.; Stahl, A. Silencing of Hepatic Fatty Acid Transporter Protein 5 in Vivo Reverses Diet-induced Non-alcoholic Fatty Liver Disease and Improves Hyperglycemia. J. Biol. Chem. 2008, 283, 22186–22192.
  24. Ipsen, D.H.; Lykkesfeldt, J.; Tveden-Nyborg, P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell. Mol. Life Sci. 2018, 75, 3313–3327.
  25. Gao, Q.; Jia, Y.; Yang, G.; Zhang, X.; Boddu, P.C.; Petersen, B.; Narsingam, S.; Zhu, Y.-J.; Thimmapaya, B.; Kanwar, Y.S.; et al. PPARα-Deficient ob/ob Obese Mice Become More Obese and Manifest Severe Hepatic Steatosis Due to Decreased Fatty Acid Oxidation. Am. J. Pathol. 2015, 185, 1396–1408.
  26. Croci, I.; Byrne, N.M.; Choquette, S.; Hills, A.P.; Chachay, V.S.; Clouston, A.D.; O’Moore-Sullivan, T.M.; Macdonald, G.A.; Prins, J.B.; Hickman, I.J. Whole-body substrate metabolism is associated with disease severity in patients with non-alcoholic fatty liver disease. Gut 2012, 62, 1625–1633.
  27. Simões, I.C.M.; Fontes, A.; Pinton, P.; Zischka, H.; Wieckowski, M.R. Mitochondria in non-alcoholic fatty liver disease. Int. J. Biochem. Cell Biol. 2018, 95, 93–99.
  28. Meex, R.C.R.; Blaak, E.E. Mitochondrial Dysfunction is a Key Pathway that Links Saturated Fat Intake to the Development and Progression of NAFLD. Mol. Nutr. Food Res. 2020, 65, e1900942.
  29. Perry, R.J.; Samuel, V.T.; Petersen, K.F.; Shulman, G.I. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature 2014, 510, 84–91.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 389
Revisions: 2 times (View History)
Update Date: 24 Jun 2022
1000/1000