Does Hyperglycemia Downregulate Glucose Transporters in The Brain

Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Medical Hypotheses 139 (2020) 109614

Contents lists available at ScienceDirect

Medical Hypotheses
journal homepage: www.elsevier.com/locate/mehy

Does hyperglycemia downregulate glucose transporters in the brain? T


a b b b
Luana Lemos Leão , Gro Tangen , Maria Lage Barca , Knut Engedal ,
Sérgio Henrique S. Santosa,c, Frederico Sander M. Machadoa, Alfredo Maurício B. de Paulaa,

Renato Sobral Monteiro-Juniora,d,e,
a
Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil
b
Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
c
Institute of Agricultural Sciences, Universidade Federal de Minas Gerais, Brazil
d
Post-Graduate Program of Medicine (Neurology/Neuroscience), Federal Fluminense University, Niterói, Rio de Janeiro, Brazil
e
Neuroscience of Exercise Institute, Aroldo Tourinho Hospital, Montes Claros, MG, Brazil

A B S T R A C T

Diabetes is a metabolic condition associated with hyperglycemia manifested by the elevation of blood glucose levels occurring when the pancreas decreases or stops
the production of insulin, in case of insulin resistance or both. The current literature supports that insulin resistance may be responsible for the memory decline
associated with diabetes. Glucose transporters (GLUTs) are a family of proteins involved in glucose transport across biological membranes. GLUT-1 and GLUT-3 are
involved in glucose delivery to the brain. Evidence suggests that both transporters are downregulated in chronic peripheral hyperglycemia. Here we show the
mechanisms of glucose transport and its influence on cognitive function, including a hypothesis of how peripheral hyperglycemia related genes network interactions
may lead to glucose transporters downregulation and its possible consequences.

Introduction energy and are based on the glucose concentration gradient across cell
membrane [10]. In humans, 14 different GLUTs have been identified.
According to WHO Global Report on Diabetes, the number of adults Under basal metabolic conditions, most of these transporters, especially
living with this disease is 422 million, and in 2016, an estimated 1.6 the GLUT-4 strongly relies on insulin dependent mechanisms and pre-
million deaths were directly caused by diabetes [1,2]. In 2019, the sents a major role on glucose uptake of adipocytes, cardiac and skeletal
global diabetes prevalence was 463 million people, rising to 578 million muscle cells and plays an important function in whole-body glucose
by 2030 and to 700 million by 2045 [3]. Diabetes is a chronic metabolic homeostasis [11,12].
condition associated with hyperglycemia manifested by the elevation of In the last decade, studies have shown that diabetes is associated
blood glucose levels when pancreas decreases the production or stops with an increased risk of cognitive decline [13,14], affecting learning,
producing insulin (type 1 diabetes) or in cases of the cells’ insulin re- working and episodic memory, cognitive flexibility and speed proces-
sistance (type 2 diabetes) or both [4,5]. Insulin resistance or deficiency sing [15–18]. Cognitive impairment related to diabetes is traditionally
is associated with impairments in glucose metabolism disrupt brain associated with atherosclerosis, since diabetes has an influence in per-
energy balance increasing oxidative stress, reactive oxygen species ipheral vascular disease and dyslipidemia that may lead to small vessel
production that leads to DNA damage, and mitochondrial dysfunction, disease in the brain, which in turn may cause vascular dementia. Al-
all of which drive pro-apoptosis, pro-inflammatory, and the amyloid though, dyslipidemia has been also related to amyloid beta protein
beta protein cascades [6]. production [19–20]. However, more recent studies suggest that the
Two different family types of glucose transporter are found in the binding mechanisms between diabetes and dementia are related to
neurovascular unit at the blood–brain barrier. The sodium-dependent hyperinsulinemia and consequent insulin resistance, and this will cause
unidirectional transporters (SLC5), which 12 isoforms (SGLTs 1–12) dementia due to Alzheimer’s disease (AD), not vascular dementia
have been identified. However, the most prevalent transporters are the [21–24]. In fact, in type 2 diabetes, insulin is not capable to reduce the
sodium-independent bidirectional GLUT [7–9]. Glucose transporters levels of blood glucose after a meal. In most cases the reason for this is
(GLUT) are a family of integral membrane proteins that provide bi- that the insulin messenger signal no longer triggers the cellular cascade
directional transport of D-glucose and its analogues without consuming of events that leads to an increased uptake of glucose by cells [25].


Corresponding author at: Grupo de Estudos e Pesquisas em Neurociência, Exercício, Saúde e Esporte - GENESEs - Universidade Estadual de Montes Claros, Av.
Doutor Rui Braga s/n, Vila Mauricéia, Montes Claros, Minas Gerais, Brazil.
E-mail address: [email protected] (R.S. Monteiro-Junior).

https://doi.org/10.1016/j.mehy.2020.109614
Received 23 December 2019; Received in revised form 30 January 2020; Accepted 5 February 2020
0306-9877/ © 2020 Elsevier Ltd. All rights reserved.
L.L. Leão, et al. Medical Hypotheses 139 (2020) 109614

The human brain is almost entirely dependent upon glucose as an homeostasis is strongly implicated by a variety of genetically en-
energy source, taking in around 100–150 g of glucose per day [26]. Due gineered mouse models. Dysfunctional GLUT-4 in skeletal muscle is the
to the restrictive permeability of the blood–brain barrier (BBB) and the main cause of peripheral insulin resistance [47,48].
relative lack of local brain carbohydrate storage, the CNS heavily relies
upon BBB expression of transporters for the delivery of key nutrients The basis of glucose transport in the brain
and solutes to the brain [27]. The delivery of blood glucose to the brain
requires crossing of glucose mediated by glucose transporter proteins. Glucose availability in the CNS is critical for neuronal function, and
Within the central nervous system, GLUT-1, mostly expressed in red glucose levels in the brain regulate local neuronal activity and whole-
blood cells and endothelial cells in heavily glycosylated form (55 kDa) body energy metabolism [49]. Among the membrane transport pro-
and in astrocytes in low glycosylated form (45 kDa), involves glucose teins, GLUT-1 is known to be a key transporter of glucose transport into
movement across the BBB’s endothelial cells [28]. Furthermore, the the brain across the BBB acting to maintain central nervous system
GLUT-3 aids glucose to pass through the neural cell membrane [29,30]. glucose homeostasis. Expression of different isoforms of the GLUT has
Studies have shown that chronic hypoglycemia upregulates the GLUT-1 also been identified at BBB endothelial cells although with relative
and GLUT-3 gene expression and increases their protein abundance. lower levels of GLUT-3 and GLUT-4 [50]. Considering the distribution
However, whether the expression of GLUT-1 and GLUT-3 is reduced in and properties of GLUT transporters, GLUT-1 and GLUT-3 will be
diabetes is unclear and is controversial. Any deficiency in GLUT treated together as they are both particularly involved in delivery of
transporter proteins leads to a major impact on brain energy metabo- glucose to the brain. As glucose is the obligatory substrate for cerebral
lism [31,32]. metabolism under normal conditions, glucose transport is fundamental
Studies have shown that vascular endothelial cells exposed to high in this organ [51,35].
glucose levels downregulate the rate of glucose transport by reducing Glucose transport into the brain is a complex process involving the
GLUT-1 mRNA and protein expression, as well as GLUT-1 plasma endothelial cells of small blood vessels, glial cells, particularly astro-
membrane localization [33,34]. In response to these recent findings, a cytes and neurons. GLUT-1 is highly expressed in the endothelial cells
couple of interesting questions arose: 1] could hyperglycemia be a of the microvasculature of the brain and is responsible for glucose
cause of downregulation of GLUT transporters in the brain? 2] if the transfer across the BBB. The transport of glucose to neurons is mediated
downregulation of GLUTs occurs in the brain, could it be one of the by astrocytes [52], which also express GLUT-1. GLUT-1 is localized to
mechanisms related to AD? Therefore, the aim of this study is to hy- both luminal and abluminal membranes of the BBB endothelial cells
pothesize a cause-effect relationship of hyperglycemia and AD based on with the ratio of 1:4 respectively, and approximately 40% of the total
GLUT transporters downregulation. Available bioinformatic data were cellular GLUT-1 resides in intracellular membrane [53–56].
used as guidance to establish the hypothesis. In order to develop a Once glucose enters the brain extracellular space, it is rapidly taken
working model, this article was organized in the following sections: i) up by the different brain cells. The neuron glucose uptake to support
The basis of glucose transport in the peripheral tissues, ii) The basis of energy metabolism is mediated by GLUT-3. GLUT-3 has higher “affi-
glucose transport in the brain, iii) Gene network analyses, iv) Genes nity” and higher glucose transporter capacity compared to GLUT-1
network model and hypothetical influence of peripheral hyperglycemia [44,57]. Interestingly, during a high cognitive demand, astrocytes’
on downregulation of GLUT transporters in the astrocytes and neurons glycolysis results in lactate production which will be delivered to ac-
and its relationship with dementia, and v) Final considerations. tivated neurons and used as energetic substrate [58]. Therefore, a
continuous supply of glucose is required for brain function.
The basis of glucose transport in the peripheral tissues
Gene network analyses
Glucose cellular uptake is an essential physiological process. The
maintenance of a relatively constant blood glucose concentration is To explore proteins-related genes associated to hyperglycemia and
necessary in order to sustain cerebral metabolism and the delivery of AD, the GeneCards database (https://www.genecards.org) was used.
glucose to peripheral tissues for energetic storage and utilization. Based on An et al. [59] who have investigated the relationship between
Indeed, glucose transport across cell membranes plays an important hyperglycemia and Alzheimer’s disease (AD), the following terms were
role in physiological regulation and control of metabolic processes [35]. inserted at GeneCards to search for genes related cellular receptors,
Cell membranes are impermeable to glucose, so the transport of hormones, proteins, and amino acids associated glucose metabolism
glucose across biological membranes must be mediated by specialized and AD. Such cellular and molecular structures searched were: GLUT-1,
protein transporters. In the 1970s, glucose uptake into liposome was GLUT-3, insulin, alanine, hexokinase, amyloid beta protein, apolipo-
reconstituted with proteins partially purified from red blood cells protein E, tau protein, and phosphofructokinase. Several genes were
[36–37]. This specific type of glucose transporter was later named as retrieved from GeneCards, as follow: SLC2A1 (GLUT-1) and SLC2A3
GLUT-1. Subsequently, glucose transporters were recognized from dif- (GLUT-3), INS (insulin), GPT (alanine), HK1/HK2/HK3 (hexokinase 1,
ferent tissues, GLUT-2, GLUT-3 and GLUT-4 [38–40]. Research devel- 2, and 3), GCK (glucokinase), APP (amyloid precursor protein), APOE
opment during the following years lead to the discovery of tissue dis- (apolipoprotein E), MAPT (microtubule-associated protein tau), PFKL/
tribution of subtypes of GLUTs, biochemical characterization of PFKP/PFKM (phosphofructokinases), and GRN (granulins). Genes
transport functions, and the formulation of the correlation between aforementioned were inserted in String database (https://string-db.
mutation or dysregulation of GLUTs and specific diseases [41–43]. org), which permits to explore genes and their interactions as a net-
Among the GLUT family, GLUT-4 is an insulin-stimulated trans- work. Our exploration of interactions was performed considering Homo
porter and it is primarily localized intracellularly in peripheral tissues sapiens as the studied species.
when not stimulated and can be acutely redistributed to the plasma Many specific and non-specific (unclear) significant interactions
membrane in response to insulin and other stimuli. GLUT-4 is primarily (PPI Enrichment p-value < 0.01) [60] were found among genes in the
expressed in adipose tissues, skeletal and cardiac muscle, and it facil- network displayed at String (Fig. 1). In String, each protein–protein
itates the diffusion of circulating glucose across its gradient into muscle interaction is annotated with one or more 'scores'. The scores indicate
and fat cells [44]. Upon binding of insulin, the insulin receptor kinase is how likely String judges an interaction to be true, given the available
activated, and it promotes the activation of downstream targets and evidence. Interactions were explored when a combined score above 0.4,
progression of signaling cascade leading to the translocation of GLUT-4 determined as medium confidence, was found. As the main question of
from intracellular pool to cell surface where it facilitates glucose entry this study was related to downregulation of GLUTs, only direct inter-
inside the cell [45,46]. The central role of GLUT-4 in glucose actions with genes associated with these transporters (SLC2A1 and

2
L.L. Leão, et al. Medical Hypotheses 139 (2020) 109614

Fig. 1. Genes network found at String. Direct binding among genes GRN, HK3, HK2, and SLC2A1 is shown. Color lines show molecular interactions, while grey lines
show no molecular interactions but associations co-mentioned in PubMed abstracts. Arrows: activation. Circles: unspecified interaction.

not known [67]. However, according to our genes network model,


bindings among granulin, HK3 and HK2 exist. In addition, a post-
translational interaction between HK2 and SLC2A1 is present in the
network. Hence, dysfunctional granulin, which is an important cell
growth factor associate to wound repair and inflammation, could affect
HK3 and HK2 interactions and the synthesis of GLUT-1. Dysfunctional
HK3 and HK2 and attenuated synthesis of GLUT-1 could result in glu-
cose excess in the extracellular space in the brain, causing a down-
Fig. 2. Specific network among genes related GLUT-1 found at String. Blue lines
mean direct binding among genes. Pink line: posttranslational modification. regulation of GLUT-1 and GLUT-3 transporters. In summary, these
Circle: unspecified interaction. mechanisms would affect astrocytes and neurons metabolism.
Evidence suggests that chronic hyperglycemia reduces intracellular
glucose transport in diabetes [35]. As a protective mechanism, the
SLC2A3) in the network were analyzed. Then, the following genes re-
decreased glucose influx can diminish the cytotoxic effect of the high
mained in the model: GRN, HK3, HK2, and SLC2A1 (Fig. 2).
sugar content. It has been suggested that the reduction of glucose
transport is related to a downregulation of 55 kDa isoform GLUT-1
Genes network model and hypothetical influence of peripheral (presented in microvessels) and GLUT-3 in patients with AD, which are
hyperglycemia on downregulation of GLUT transporters in the the principal factors affecting glucose transport and metabolism in the
astrocytes and neurons, and its relationship with dementia brain [41,68]. In a study with diabetic rat model, chronic hypergly-
cemia downregulated GLUT-1 and GLUT-3 gene expression levels in the
Diabetes is associated with hyperglycemia and it is caused by partial brain [61]. The downregulation of GLUT-1 and GLUT-3 expression
or total insulin insufficiency. Clinically, the rapid decline in blood might be the adaptive reaction of the body to prevent excessive glucose
glucose in patients with diabetes, even when the blood glucose is above entering the cell that may lead to cell damage [61].
normal, can lead to “hypoglycemia phenomenon”, including neu- It is well known that glucose transport from the peripheral circu-
roglycopenia symptoms. The exact mechanism remains unclear, but it is lation across the BBB and capillary endothelial cells into the interstitial
possibly related to the downregulation of GLUT-1 and GLUT-3 expres- fluid and brain tissue are largely insulin-independent processes. Many
sion in diabetes [61]. epidemiological studies indicate that peripheral insulin resistance and
GLUT-1, which is expressed and localized at the endothelial cells of diabetes are risk factors for AD [69–74], however, it is not known
the BBB, is the first step in the transport of glucose from the blood into whether brain glucose dysregulation is a key feature of AD and is re-
the tissue layers. Then, the glucose transport from extracellular space lated to severity of AD pathology or symptom expression [75].
into neuronal cells is taken by GLUT-3, localized at the neuronal cell In patients with AD, several components of the insulin signaling
membrane. After it is within the cell, glucose is phosphorylated to pathway are abnormal, including genes encoding insulin, IGF-1, and
glucose-6-phosphate by HK, a key enzyme for glucose utilization in the IGF-2 peptides and their receptors [76,77]. Evidence suggests that ab-
cell [62,63]. The downstream cascade of glucose breakdown is mainly normal insulin signaling contributes to clinical trials targeting ab-
mediated by HK, hence it is reasonable to hypothesize that any change normalities in patients with mild cognitive impairment and AD, but
in this enzyme could affect glucose metabolism. implications of these abnormalities are yet to be elucidated [78,79].
Granulin (GRN) is a protein related to inflammation and wound Some studies have shown reduced brain glucose uptake in regions
repair originated from progranulin (PGRN). Mutations in PGRN genes vulnerable to AD pathology [80–82]. It is unclear whether an overall
are associated with neurodegenerative diseases, such as frontotemporal failure in the regulation of brain glucose metabolism is a key aetio-
dementia [64]. PGRN was demonstrated to interact with the HK3. pathogenic factor in AD and whether abnormalities of brain glucose
Autopsy studies had shown associations of PGRN and beta-amyloid homeostasis in AD are related to peripheral glucose concentration.
plaques [65,66]. PGRN plaques were most dense in medial temporal Since dysfunctional granulin could affect HK3 and HK2 interactions,
and frontal regions and predominated over aggregated amyloid beta it could also result in glucose excess in extracellular space in the brain.
protein. At this moment, physiological significance of this interaction is These mechanisms would lead to GLUT-1 downregulation affecting

3
L.L. Leão, et al. Medical Hypotheses 139 (2020) 109614

astrocytes and neurons. Furthermore, changes in HK3 and HK2 also and late-onset Alzheimer’s disease. Dement Geriatr Cogn Disord
seem to affect GLUT-1 expression through SLC2A1 posttranslational 2011;31(6):424–30.
[14] Cichosz SL, Jensen MH, Hejlesen O. Cognitive impairment in elderly people with
modification. Therefore, it makes sense to develop a hypothetical model prediabetes or diabetes: A cross-sectional study of the NHANES population. Primary
which peripheral hyperglycemia triggers a downregulation of GLUT Care Diabetes 2019.
transporters in the brain. This hypothetical model highlights two con- [15] Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC. Increased risk of
type 2 diabetes in Alzheimer’s disease. Diabetes 2004;53(2):474–81.
sequences: 1) hyperglycemia in the brain could trigger neuroin- [16] Ballesteros S, Redondo MT. Alzheimer’s disease and Type 2 Diabetes Mellitus:
flammation, especially influenced by pro-inflammatory cytokines [83], Similar Memory and Executive Functions Impairments? J Alzheimer Dis
downregulating GLUT; and 2) fuel to neurons could be limited due to Parkinsonism 2017;7(6):389.
[17] Cheng G, Huang C, Deng H, Wang H. Diabetes as a risk factor for dementia and mild
few GLUT-1 transporters, leading them to apoptosis. cognitive impairment: a meta-analysis of longitudinal studies. J Intern Med
2012;42(5):484–91.
Final considerations [18] Cerasuolo J, Izzo A. Persistent impairment in working memory following severe
hyperglycemia in newly diagnosed type 2 diabetes. Endocrinol Diabetes Metab Case
Rep 2017;1:1–4.
Glucose is the primary energetic fuel of brain tissue, the availability [19] Reitz C. Dyslipidemia and dementia: current epidemiology, genetic evidence and
of glucose and its transport into the brain across the BBB and into brain mechanisms behind the associations. J Alzheimers Dis 2012;30(2):S127–45.
cells plays a key role in normal physiological function and energy [20] Refolo LM, Malester B, LaFrancois J, Bryant-Thomas T, Wang R, Tint GS, et al.
Hypercholesterolemia accelerates the Alzheimer's amyloid pathology in a trans-
metabolism. This mechanism is mediated by two GLUT transporters: genic mouse model. Neurobiol Dis 2000;7(4):321–31.
GLUT-1 and GLUT-3. GLUT-1 transporter presents on both the luminal [21] Grasso G, Pietropaolo A, Spoto G, Pappalardo G, Tundo GR, Ciaccio C, et al. Copper
and abluminal membranes of the BBB endothelial cells, also in astro- (I) and copper(II) inhibit Ab peptides proteolysis by insulin-degrading enzyme
differently: implications for metallostasis alteration in Alzheimer’s disease. Chem
cytes. GLUT-3 is located in neurons. Studies showed that chronic hy- 2011;17:2752–62.
poglycemia enhances the GLUT-1 and GLUT-3 gene expression. Also, [22] Hölscher C. Diabetes as a risk factor for Alzheimer’s disease: insulin signalling
the expression levels of both transporters are downregulated in chronic impairment in the brain as an alternative model of Alzheimer’s disease. Biochem
Soc Trans 2011;39(4):891–7.
peripheral hyperglycemia, suggesting that GLUT-1 and GLUT-3 ex- [23] Shinohara M, Sato N. Bidirectional interactions between diabetes and Alzheimer's
pression might be the adaptive reaction of the body to prevent cell disease. Neurochem Int 2017;108:296–302.
damage. However, molecular mechanisms that lead to GLUT-1 and [24] Vieira MNN, Lima-Filho RAS, De Felice FG. Connecting Alzheimer's disease to
diabetes: Underlying mechanisms and potential therapeutic targets.
GLUT-3 reductions in AD remain unknown. The present hypothesis is Neuropharmacol 2018;136:160–71.
based on molecular and clinical findings and presents the consequences [25] Lebovitz HE, Banerji MA. Treatment of insulin resistance in diabetes mellitus. Eur J
of GLUTs downregulation that could be related events leading to neu- Pharmacol 2004;490:135–46.
[26] Kuzawa CW, Chugani HT, Grossman LI, Lipovich L, Muzik O, Hof PR, et al.
ronal damage, neurodegeneration, cognitive decline and AD.
Metabolic costs and evolutionary implications of human brain development. Proc
Natl Acad Sci USA 2014;111(36):13010–5.
Declaration of Competing Interest [27] Shah K, DeSilva S, Abbruscato T. The Role of Glucose Transporters in Brain Disease:
diabetes and Alzheimer’s Disease. Int J Mol Sci 2012;13(10):12629–55.
[28] Dick AP, Harik SI, Klip A, Walker DM. Identification and characterization of the
The authors declare that they have no known competing financial glucose transporter of the blood-brain barrier by cytochalasin B binding and im-
interests or personal relationships that could have appeared to influ- munological reactivity. Proc Natl Acad Sci USA 1984;81(22):7233–7.
ence the work reported in this paper. [29] Duelli R, Kuschinsky W. Brain glucose transporters: relationship to local energy
demand. News Physiol Sci 2001;16:71–6.
[30] Wood IS, Trayhurn P. Glucose transporters (GLUT and SGLT): expanded families of
Acknowledgements sugar transport proteins. Br J Nutr 2003;89(1):3–9.
[31] Dwyer KJ, Boado RJ, Pardridge WM. Cis-element/cytoplasmic protein interaction
within the 3'-untranslated region of the GLUT1 glucose transporter mRNA. J
Coordenação de Aperfeiçoamento de Pessoal de Nível Superior Neurochem 1996;66(2):449–58.
(CAPES), and Conselho Nacional de Desenvolvimento Científico e [32] Lee DH, Chung MY, Lee JU, Kang DG, Paek YW. Changes of glucose transporters in
Tecnológico (CNPq). the cerebral adaptation to hypoglycemia. Diabetes Res Clin Pract
2000;47(1):15–23.
[33] Alpert E, Gruzman A, Totary H, Kaiser N, Reich R, Sasson S. A natural protective
References mechanism against hyperglycemia in vascular endothelial and smooth-muscle cells:
role of glucose and 12-hydroxyeicosatetraenoic acid. Biochem J 2002;362:413–22.
[34] Riahi Y, Sin-Malia Y, Cohen G, Alpert E, Gruzman A, Eckel J, et al. The natural
[1] World Health Organization. Global Report on Diabetes. 2016. Accessed online at.
protective mechanism against hyperglycemia in vascular endothelial cells: roles of
https://www.who.int/diabetes/global-report/en/.
the lipid peroxidation product 4-hydroxydodecadienal and peroxisome proliferator-
[2] World Health Organization. Diabetes. 2018. Accessed online at. https://www.who.
activated receptor delta. Diabetes 2010;59(4):808–18.
int/news-room/fact-sheets/detail/diabetes.
[35] Brown GK. Glucose transporters: structure, function and consequences of defi-
[3] Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, et al. Global and
ciency. J Inherit Metab Dis 2000;23(3):237–46.
regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045:
[36] Enerson BE, Drewes LR. The rat blood-brain barrier transcriptome. J Cereb Blood
Results from the International Diabetes Federation Diabetes Atlas, 9th edition.
Flow Metab 2006;26(7):959–73.
Diabetes Res Clin Pract 2019;157:107843.
[37] Gerhart DZ, Broderius MA, Borson ND, Drewes LR. Neurons and microvessels ex-
[4] Moini J. Epidemiology of Diabetes, 1st edition. 2019;75–90. doi:10.1016/b978-0-
press the brain glucose transporter protein glut3. Proc Natl Acad Sci USA
12-816864-6.00006-7.
1992:733–7.
[5] van Duinkerken E, Ryan CM. Diabetes mellitus in the young and the old: Effects on
[38] Ngarmukos C, Baur EL, Kumagai AK. Co-localization of glut1 and glut4 in the blood-
cognitive functioning across the life span. Neurobiol Dis 2020;134:104608.
brain barrier of the rat ventromedial hypothalamus. Brain Res 2001;900(1):1–8.
[6] Rendraa E, Riabovb V, Mosselb DM, Sevastyanovab T, Harmsena MC, Kzhyshkowsk
[39] Nishizaki T, Matsuoka T. Low glucose enhances na+/glucose transport in bovine
J. Reactive oxygen species (ROS) in macrophage activation and function in diabetes.
brain artery endothelial cells. Stroke 1998;29:844–9.
Immunobiology 2019;224(2):242–53.
[40] Wang D, Pascual JM, Yang H, Engelstad K, Mao X, Cheng J, et al. A mouse model for
[7] Wright EM. Glucose transport families SLC5 and SLC50. Mol Aspects Med
Glut-1 haploinsufficiency. Hum Mol Genet 2006;15(7):1169–79.
2013;34(2–3):183–96.
[41] Mooradian AD, Chung HC, Shah GN. GLUT-1 expression in the cerebra of patients
[8] Cura AJ, Carruthers A. Role of monosaccharide transport proteins in carbohydrate
with Alzheimer's disease. Neurobiol Aging 1997;18(5):469–74.
assimilation, distribution, metabolism, and homeostasis. Comprehens Physiol
[42] Winkler EA, Yoichiro Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D, et al.
2012;2(2):863–914.
GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction
[9] Szablewski L. Glucose Transporters in Brain. In Health and in Alzheimer's Disease. J
and degeneration. Nat Neurosci 2015;18:521–30.
Alzheimers Dis 2017;55(4):1307–20.
[43] Vemula S, Roder KE, Yang T, Bhat GJ, Thekkumkara TJ. A functional role for so-
[10] Thorens B, Mueckler M. Glucose transporters in the 21st Century. Am J Physiol
dium-dependent glucose transport across the blood-brain barrier during oxygen
Endocrinol Metab 2010;298(2):141–5.
glucose deprivation. J Pharmacol Exp Ther 2009;328(2):487–95.
[11] Huang S, Czech MP. The GLUT4 glucose transporter. Cell Metab 2007;5(4):237–52.
[44] Simpson IA, Carruthers A, Vannucci SJ. Supply and demand in cerebral energy
[12] Moraes-Vieira PM, Saghatelian A, Kahn BB. GLUT4 expression in adipocytes reg-
metabolism: The role of nutrient transporters. J Cereb Blood Flow Metab
ulates de novo lipogenesis and levels of a novel class of lipids with antidiabetic and
2007;27(11):1766–91.
anti-inflammatory effects. Diabetes 2016;65(7):1808–15.
[45] Lowe AG, Walmsley AR. The kinetics of glucose transport in human red blood cells.
[13] Cheng D, Noble J, Tang MX, Schupf N, Mayeux R, Luchsinger JA. Type 2 diabetes
Biochim Biophys Acta 1986;857(2):146–54.

4
L.L. Leão, et al. Medical Hypotheses 139 (2020) 109614

[46] Arha D, Pandeti S, Mishra A, Srivastava SP, Srivastava AK, Narender T, et al. [65] Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C,
Deoxyandrographolide promotes glucose uptake through glucose transporter-4 et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked
translocation to plasma membrane in L6 myotubes and exerts antihyperglycemic to chromosome 17. Nature 2006;442(7105):916–9.
effect in vivo. Eur J Pharmacol 2015;768:207–16. [66] Gliebus G, Rosso A, Lippa CF. Progranulin and beta-amyloid distribution: a case
[47] Nicholson T, Church C, Baker DJ, Jones SW. The role of adipokines in skeletal report of the brain from preclinical PS-1 mutation carrier. Am J Alzheimer's Dis
muscle inflammation and insulin sensitivity. J Inflamm 2018;9(15):9. Dementias 2009;24(6):456–60.
[48] García-Cáceres C, Quarta C, Varela L, Gao Y, Gruber T, Legutko B, et al. Astrocytic [67] Sui D, Wilson JE. Interaction of insulin-like growth factor binding protein-4, Miz-1,
Insulin Signaling Couples Brain Glucose Uptake with Nutrient Availability. Cell leptin, lipocalin-type prostaglandin D synthase, and granulin precursor with the N-
2016;166(6):867–80. terminal half of type III hexokinase. Arch Biochem Biophys 2000;382(2):262–74.
[49] Shawahna R, Uchida Y, Decleves X, Ohtsuki S, Yousif S, Dauchy S, et al. [68] Choeiri C, Staines W, Messier C. Immunohistochemical localization and quantifi-
Transcriptomic and quantitative proteomic analysis of transporters and drug me- cation of glucose transporters in the mouse brain. Neuroscience 2002;111(1):19–34.
tabolizing enzymes in freshly isolated human brain microvessels. Mol Pharm [69] Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, O’Brien PC, et al. Risk of
2011;8(4):1332–41. dementia among persons with diabetes mellitus: a population-based cohort study.
[50] Uchida Y, Ohtsuki S, Katsukura T, Ikeda C, Suzuki T, Kamiie J, et al. Quantitative Am J Epidemiol 1997;145(4):301–8.
targeted absolute proteomics of human blood-brain barrier transporters and re- [70] Luchsinger JA, Tang MX, Stern Y, Shea S, Mayeux R. Diabetes mellitus and risk of
ceptors. J Neurochem 2011;117(2):333–45. Alzheimer’s disease and dementia with stroke in a multiethnic cohort. Am J
[51] Ito K, Uchida Y, Ohtsuki S, Aizawa S, Kawakami H, Katsukura Y, et al. Quantitative Epidemiol 2001;154(7):635–41.
membrane protein expression at the blood-brain barrier of adult and younger cy- [71] Chen Z, Zhong C. Decoding Alzheimer's disease from perturbed cerebral glucose
nomolgus monkeys. J Pharm Sci 2011;100(9):3939–50. metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol
[52] Forsyth R, Fray A, Boutelle M, Fillenz M, Middleditch C, Burchell A. A role for 2013;108:21–43.
astrocytes in glucose delivery to neurons? Dev Neurosci 1996;18(5–6):360–70. [72] Ahtiluoto S, Polvikoski T, Peltonen M, Solomon A, Tuomilehto J, Winblad B, et al.
[53] Gerhart DZ, LeVasseur RJ, Broderius MA, Drewes LR. Glucose transporter locali- Diabetes, Alzheimer disease, and vascular dementia: a population-based neuro-
zation in brain using light and electron immunocytochemistry. J Neurosci Res pathologic study. Neurology 2010;75(13):1195–202.
1989;22(4):464–72. [73] Hsu CC, Wahlqvist ML, Lee MS, Tsai HN. Incidence of dementia is increased in type
[54] Farrell CL, Pardridge WM. Ultrastructural localization of blood-brain barrier-spe- 2 diabetes and reduced by the use of sulfonylureas and metformin. J Alzheimers Dis
cific antibodies using immunogold-silver enhancement techniques. J Neurosci 2011;24(3):485–93.
Methods 1991;37(2):103–10. [74] Domínguez RO, Marschoff ER, González SE, Repetto MG, Serra JA. Type 2 diabetes
[55] Cornford EM, Hyman S, Landaw EM. Developmental modulation of blood-brain- and/or its treatment leads to less cognitive impairment in Alzheimer’s disease pa-
barrier glucose transport in the rabbit. Brain Res 1994;663(1):7–18. tients. Diabetes Res Clin Pract 2012;98(1):68–74.
[56] Farrell CL, Pardridge WM. Blood-brain barrier glucose transporter is asymmetrically [75] Brain Hoyer S. glucose and energy metabolism abnormalities in sporadic Alzheimer
distributed on brain capillary endothelial lumenal and ablumenal membranes: An disease. Causes and consequences: an update. Exp Gerontol
electron microscopic immunogold study. Proc Natl Acad Sci USA 2000;35(9–10):1363–72.
1991;88(13):5779–83. [76] Werner H, LeRoith D. Insulin and insulin-like growth factor receptors in the brain:
[57] Maher F, Davies-Hill TM, Simpson IA. Substrate specificity and kinetic parameters physiological and pathological aspects. Eur Neuropsychopharmacol
of glut3 in rat cerebellar granule neurons. Biochem J 1996;315:827–31. 2014;24(4):1947–53.
[58] Sobral-Monteiro-Junior R, Maillot P, Gatica-Rojas V, Ávila WRM, de Paula AMB, [77] Frater J, Lie D, Bartlett P, McGrath JJ. Insulin-like Growth Factor 1 (IGF-1) as a
Guimarães ALS, et al. Is the “lactormone” a key-factor for exercise-related neuro- marker of cognitive decline in normal ageing: A review. Age Res Rev
plasticity? A hypothesis based on an alternative lactate neurobiological pathway. 2018;42:14–27.
Med Hypotheses 2019;123:63–6. [78] Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al. Demonstrated brain
[59] An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, et al. Evidence for insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance,
brain glucose dysregulation in Alzheimer's disease. Alzheimer's Dementia IRS-1 dysregulation, and cognitive decline. J Clin Investig 2012;122(4):1316–38.
2018;14(3):318–29. [79] Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, Craft S. Insulin resistance
[60] Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, et al. and Alzheimer-like reductions in regional cerebral glucose metabolism for cogni-
STRING v9.1: protein-protein interaction networks, with increased coverage and tively normal adults with prediabetes or early type 2 diabetes. Arch Neurol
integration. Nucleic Acids Res 2013;41:808–15. 2011;68(1):51–7.
[61] Hou WK, Xian YX, Zhang L, Lai H, Hou XG, Xu YX, et al. Influence of blood glucose [80] Herholz K. Cerebral glucose metabolism in preclinical and prodromal Alzheimer’s
on the expression of glucose trans-porter proteins 1 and 3 in the brain of diabetic disease. Expert Rev Neurother 2010;10(11):1667–73.
rats. Chin Med J 2007;120(19):1704–9. [81] Hunt A, Schonknecht P, Henze M, Seidl U, Haberkorn U, Schroder J. Reduced
[62] Pardridge WM, Boado RJ, Farrell CR. Brain-type glucose transporter (GLUT-1) is cerebral glucose metabolism in patients at risk for Alzheimer’s disease. Psychiatry
selectively localized to the blood-brain barrier. Studies with quantitative western Res 2007;155(2):147–54.
blotting and in situ hybridization. J Bio Chem 1990;15(29):18035–40. [82] Mosconi L. Brain glucose metabolism in the early and specific diagnosis of
[63] Maher F, Vannucci SJ, Simpson IA. Glucose transporter proteins in brain. FASEB J Alzheimer’s disease. FDG-PET studies in MCI and AD. Eur J Nucl Med Mol Imaging
1994;8(13):1003–11. 2005;32(4):486–510.
[64] Gass J, Cannon A, Mackenzie IR, Boeve B, Baker M, Adamson J, et al. Mutations in [83] Lee HJ, Seo HI, Cha HY, Yang YJ, Kwon SH, Yang SJ. Diabetes and Alzheimer's
progranulin are a major cause of ubiquitin-positive frontotemporal lobar degen- Disease: Mechanisms and Nutritional Aspects. Clin Nutr Res 2018;7(4):229–40.
eration. Hum Mol Genet 2006;15(20):2988–3001.

You might also like