Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

REVIEW ARTICLE

Glycogen storage disease type Ib: role of glucose-6-


phosphate transporter in cell metabolism and function
Sang Wan Sim1, David A. Weinstein2, Young Mok Lee2 and Hyun Sik Jun1
1 Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, Korea
2 Glycogen Storage Disease Program, University of Connecticut School of Medicine, Farmington, CT, USA

Correspondence Cellular metabolism generally refers to biochemical processes that produce or


H. S. Jun, Department of Biotechnology and consume energy within the cell. Recent studies have established that aberrant
Bioinformatics, College of Science and
metabolic states caused by internal or external stresses and genetic mutations
Technology, Korea University, Sejong 339-
are intertwined with several human pathologies. Gaining insight into these
700, Korea
Tel: +82-44-860-1411 metabolic alterations is, therefore, essential for understanding the pathophysi-
E-mail: toddjun@korea.ac.kr ology of various diseases. Glycogen storage disease type Ib (GSD-Ib) is an
autosomal recessive disorder characterized by hypoglycemia, excessive glyco-
(Received 12 July 2019, revised 16 October gen accumulation in the liver and kidney, neutropenia, neutrophil dysfunction,
2019, accepted 25 October 2019, available and inflammatory bowel disease. GSD-Ib is caused by a deficiency of glu-
online 22 November 2019)
cose-6-phosphate transporter (G6PT). Recently, it was reported that defi-
ciency of G6PT also leads to the aberrant proliferation and differentiation of
doi:10.1002/1873-3468.13666
mesenchymal stem cells and impaired regulatory T-cell function. This review
Edited by Maria Papatriantafyllou describes the broad impact of altered cellular metabolism resulting from a
lack of G6PT activity on cellular function and considers the prospects of
developing novel approaches for GSD-Ib treatment.

Keywords: autoimmune disease; CD4+ T cell; function; glucose-6-


phosphate transporter; glycogen storage disease type Ib; macrophage;
mesenchymal stem cell; metabolism; monocyte; neutrophil

Introduction to glycogen storage


disease type I
Living organisms require a constant supply of energy substrates for adenosine triphosphate (ATP) produc-
to support essential processes for life. In our body, tion to maintain various cellular activities in most cell
most cells acquire this energy from glucose, which types [3]. Secondly, diverse substrates produced at var-
enters the cell through glucose transporters and is ious steps are utilized as building blocks for macro-
immediately converted to glucose-6-phosphate (G6P) molecules, such as DNA, RNA, and proteins, which
by either hexokinase or glucokinase [1]. Glucose/G6P are essential for the proliferation, differentiation, and
is metabolized through four major pathways, glycoly- effector functions of cells [1,4].
sis, pentose phosphate pathway (PPP), tricarboxylic Many studies have demonstrated that the repro-
acid (TCA) cycle, and fatty acid synthesis [1,2], which gramming of cellular metabolism drives the differentia-
produce different substrates that are utilized in two tion and effector functions of stem cells and immune
main ways. Firstly, glycolysis and oxidative phospho- cells [1,5]. In this regard, cellular metabolism is a
rylation (OXPHOS) through the TCA cycle provide dynamic biochemical process that adapts to the

Abbreviations
DC, dendritic cell; G6P, glucose-6-phosphate; G6PT, glucose-6-phosphate transporter; GSD-Ib, glycogen storage disease type Ib; HIF-1a,
hypoxia-inducible factor-1a; hMSC, human mesenchymal stem cell; OXPHOS, oxidative phosphorylation; PPP, pentose phosphate pathway;
Treg, regulatory T cell.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 3
Role of G6PT in cell metabolism and function S. W. Sim et al.

bioenergetic demands of cells and its dysregulation are now well established, other aspects of immune dis-
resulting from genetic mutations and internal or exter- orders observed in GSD-Ib patients are still poorly
nal stresses can cause diseases in humans [6]. understood. In this review, we will introduce the gen-
Glycogen storage disease type I (GSD-I), also eral metabolic and functional phenotypes of immune
known as von Gierke disease, is an autosomal reces- cells and will discuss the impact of altered cellular
sive disorder due to altered cellular metabolism [7]. metabolism, resulting from a lack of G6PT activity in
Type I GSD is divided into two subtypes, GSD-Ia and immune cells, including neutrophils, macrophages,
glycogen storage disease type Ib (GSD-Ib), caused by CD4+ T cells, and mesenchymal stem cells. Finally, the
mutations in G6PC and G6PT (also known as limitations of current therapies and the development
SLC37A4) genes, respectively [8–11]. Glucose-6-phos- of novel clinical approaches for the treatment of GSD-
phate transporter (G6PT) is localized in the ER mem- Ib will be discussed [31–33].
brane, where it facilitates the translocation of G6P
from the cytoplasm into the ER lumen and its subse-
Glucose-6-phosphate transporter
quent hydrolysis into glucose and phosphate (Pi),
thereby controlling the cytoplasmic glucose/G6P con- Glucose-6-phosphate transporter is encoded by the
centration [12,13]. Hydrolysis of G6P is catalyzed by solute carrier family 37 member 4 (SLC37A4) gene
glucose-6-phosphatase (G6Pase) located in the ER and is localized in the ER membrane. In humans,
membrane, and hydrolyzed glucose is released into the G6PT is a single-copy gene composed of nine exons
cytoplasm [8,14]. This ER-mediated cycling of glucose on chromosome 11q23 and its two spliced variants
limits cytoplasmic glucose/G6P availability, in turn contain 429 and 451 amino acids, respectively
regulating glycolysis and PPP, and ultimately glucose [10,34,35]. The former transcript is expressed ubiqui-
homeostasis [7,15]. There are two enzymatically active tously (G6PT), while the latter one is exclusively
G6Pases: G6Pase-a, encoded by G6PC, is primarily expressed in the brain, heart, and muscle (vG6PT)
expressed in gluconeogenic organs, such as the liver, [11,36]. The functional differences between these tran-
kidney, and intestine [8], and ubiquitously expressed scripts are not understood because both proteins are
G6Pase-b, encoded by G6PC3 [14]. Both GSD-Ia and catalytically active, consist of 10 transmembrane
-Ib manifest as hypoglycemia, excessive glycogen accu- helices, and have their NH2 and COOH termini
mulation, growth retardation, hyperlipidemia, and lac- located in the cytoplasm [36,37]. G6PT belongs to a
tic acidemia [7], whereas immune disorders, including sugar-phosphate/phosphate antiporter that exchanges
neutropenia and myeloid dysfunctions, are unique to cytoplasmic G6P for inorganic phosphate stored in the
GSD-Ib and lead to recurrent bacterial infections in lumen of the ER [12]. On the ER membrane, G6PT
GSD-Ib patients [16–18]. Deficiency of G6Pase-b, also transports G6P from the cytoplasm into the ER lumen
called severe congenital neutropenia syndrome type 4 and delivers it to G6Pase-a or G6Pase-b for hydrolysis
(SCN4), does not lead to GSD, even though it causes into glucose and phosphate. Thus, G6PT plays a vital
neutropenia and myeloid dysfunctions observed in role in maintaining glucose homeostasis in blood and
GSD-Ib patients [19–24]. SCN4 also shows nonhema- energy homeostasis in the cells [7,15] (Fig. 1A).
tological defects, such as a structural defect in the
heart and abnormalities in urogenital organs, suggest-
Deficiency of G6PT and Glycogen
ing that G6Pase-b plays additional roles that have not
storage disease type Ib
been fully identified yet [20].
Most recently, G6PT was found to play a critical Glycogen storage disease type Ib is a rare inherited
role in both CD4+ T-cell functions and the prolifera- disease that is caused by G6PT deficiency. GSD-Ib is
tion and differentiation of human mesenchymal stem characterized by impaired glucose homeostasis and
cells (hMSCs), suggesting that immune deficiencies in immune disorders, including neutropenia and neu-
GSD-Ib extend beyond neutropenia and neutrophil trophil dysfunction [7,15]. As a result of these immune
dysfunction [25,26]. Multipotent hMSCs are character- deficiencies, GSD-Ib patients suffer from recurrent
ized not only by their ability to differentiate into bacterial infections. The vast majority of GSD-Ib
mesodermal lineages, such as adipocytes, osteocytes, patients who manifest neutropenia also develop inflam-
and chondrocytes [27], but also by immunoregulatory matory bowel disease, which is not distinguishable
capacity on both innate and adaptive immune cells from Crohn’s disease [16,38]. A study that examined
[28–30]. The majority of research on GSD-Ib has been the hypothalamus–pituitary–thyroid axis in seven
focused on metabolic phenotypes of the liver and neu- GSD-Ib patients revealed that 57% of the patients
trophil-specific immune deficiencies. Although these showed thyroid autoimmunity and hypothyroidism

4 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

Fig. 1. Pathways for G6P metabolism in gluconeogenic organs and neutrophils. (A) Gluconeogenic organs, such as the liver, kidney, and
intestine, primarily express G6Pase-a. Interprandial glucose homeostasis is maintained by the G6PT/G6Pase-a, which is localized in the ER
membrane. G6PT transports G6P from the cytoplasm into the lumen of the ER, and then, G6Pase-a hydrolyzes G6P to glucose and
phosphate, which are ultimately released into the cytoplasm. In gluconeogenic organs, glucose can be generated from glycogenolysis and
gluconeogenesis. G6PT/G6Pase-a plays a critical role in the final step of these two pathways, regulating the blood glucose for use by
nongluconeogenic organs between meals. (B) Neutrophil glucose homeostasis is maintained by the G6PT/G6Pase-b. In contrast with
G6Pase-a, G6Pase-b is ubiquitously expressed, but functions in a similar manner as G6PT/G6Pase-a. In wild-type neutrophils, glucose is
metabolized into G6P, which can be further utilized in the PPP, glycolysis, glycogen synthesis, and ER cycling. Most recently, a new
substrate for G6Pase-b, 1,5-AG6P, was discovered. Even though the transporter on the plasma membrane for 1,5-AG is unknown, 1,5-AG is
phosphorylated into 1,5-AG6P by hexokinases or ADP-GK in the cytoplasm. G6PT can transport both G6P and 1,5-AG6P into the ER lumen,
and G6Pase-b hydrolyzes two phosphorylated forms to glucose/phosphate and 1,5-AG/phosphate, respectively. In wild-type neutrophils,
1,5-AG6P is continuously converted into 1,5-AG by ER cycling, maintaining the constant physiological concentration.

[39]. Numerous mutations have been identified in phagocytosis, respiratory burst activity, the release of
SLC37A4; however, all GSD-Ib patients do not show granule contents, and extracellular traps [47]. These
neutropenia even though they display metabolic phe- microenvironmental and functional changes are
notypes of the disease, suggesting that other factors accompanied by metabolic changes that have a direct
could modify gene activity [15]. To date, 91 mutations role in regulating neutrophil function [48,49].
in the G6PT gene have been described, including 39 It has been known that neutrophils primarily rely
missense, 11 nonsense, 21 insertion/deletion, and 19 on glycolysis and exhibit significant low levels of
splicing mutations. These mutations are distributed OXPHOS [49–51]. To fuel their effector functions,
throughout the coding region and exon–intron junc- including the respiratory burst activity and the forma-
tions [40]. Among these genetic changes, site-directed tion of neutrophil extracellular traps, neutrophils heav-
mutagenesis and transient expression assays have con- ily utilize glucose and glycolysis [49,50,52]. Consistent
firmed the pathogenicity of 31 missense mutations and with this notion, inhibition of glycolysis by 2-
two deletions. Of note, these pathogenic mutations deoxyglucose, a glucose molecule that is taken up by
abolish or greatly reduce the microsomal uptake activ- glucose transporters but cannot undergo further gly-
ity of G6P [10,41–43]. colysis, severely reduced cellular ATP concentration
and impaired the phagocytic response of neutrophils
[49,53]. Activated neutrophils synthesize NADPH to
Cellular metabolism and function of
support their respiratory burst activity [54]. A shunt
neutrophils
into the PPP, which generates ribose for nucleotide
In humans, neutrophils account for about 50–70% of synthesis, forms the NADPH cofactor required for
circulating leukocytes and serve as the first line of reactive oxygen species (ROS) production via the
defense against foreign antigens, such as bacteria and NADPH oxidase 2 activity [55]. In addition, neu-
fungi [44]. Once terminally differentiated, neutrophils trophils exploit glutaminolysis, which uses several steps
are relatively short-lived and about 1011 new neu- from the TCA cycle and produces NADPH from the
trophils are produced daily [45]. Neutrophils respond oxidation of glutamine-derived malate to pyruvate [1]
to immunological challenges, migrating to injured or (Fig. 1B).
infected tissues where the oxygen is limited [46]. At the Compared with most other mammalian cell types,
infected sites, they destroy invading pathogens through neutrophils contain very few mitochondria [51]. This

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 5
Role of G6PT in cell metabolism and function S. W. Sim et al.

observation has led to the impression that mitochon- gluconeogenesis; thus, they are highly dependent on
dria would lack the capacity for mitochondrial respira- the continuous supply of glucose from the blood to
tion in neutrophils. However, a few studies have function and survive [49]. Intracellular glucose/G6P in
revealed the unrecognized importance of mitochondria neutrophils is metabolized via three primary pathways:
in the function of neutrophils during infection and glycolysis, PPP, and ER cycling [21]. The latter path-
inflammation, besides their well-established role in way regulates the other two cytoplasmic pathways by
apoptosis [50,56]. Fossati et al. [50] suggested that controlling the availability of cytoplasmic glucose/G6P
some effector functions of neutrophils require intact [21]. In GSD-Ib patients, neutrophils showed reduced
mitochondrial functionality. Bao et al. [56] revealed glucose uptake [13]. In keeping with this finding,
that stimulation with f-Met-Leu-Phe (fMLP) rapidly impaired energy homeostasis, including decreased
increases the mitochondrial membrane potential and levels of G6P, lactate, and ATP, is also observed in
ATP synthesis, using a method that can observe ATP G6PT / neutrophils. In addition, this study also
release from living cells in real time. The released ATP demonstrated that both ER and the mitochondrial
is produced by mitochondria and is essential for the stress-induced hypoxia-inducible factor-1a (HIF-1a)/
activation of neutrophils. In addition, the initial burst peroxisome proliferator-activated receptor-c (PPAR-c)
of ATP release is followed by a metabolic switch to pathway downregulate respiratory burst, chemotaxis,
glycolysis for further ATP production that contributes and calcium mobilization activities, leading to neu-
to sustained intracellular Ca2+ levels and maintenance trophil dysfunction [13]. Neutrophils of SCN4 patients
of functional cell responses [56]. Taken together, mito- also showed reduced glucose uptake and decreased
chondria appear to have an unexpected role in the levels of energy homeostasis which lead to neutrophil
function of neutrophils. dysfunction [21]. However, whether the HIF-1a/
PPAR-c pathway is involved in neutrophil dysfunction
in SCN4 patients or mice remains to be further exam-
Neutrophils and G6PT
ined.
The mechanism of immune deficiency in GSD-Ib and Most recently, Veiga-da-Cunha et al. [60] proposed
its relationship with G6P metabolism has long been the interesting underlying mechanism for neutropenia
unclear. In 2003, Kuijpers et al. [57] suggested a rela- and neutrophil dysfunction observed in GSD-Ib and
tionship between apoptosis and neutropenia, showing SCN4 patients. To investigate the exact role of G6PT
that the neutrophils of GSD-Ib patients exhibit and G6Pase-b, they compared the substrate prefer-
enhanced apoptosis, but the underlying mechanism ences and kinetic properties of G6Pase-a and G6Pase-
remained uncertain. In this regard, the discovery of b. As a result, they found that the G6Pase-b acts much
G6Pase-b and its expression in neutrophils have led to more efficiently on 1,5-anhydroglucitol-6-phosphate
a better understanding of the role of the G6PT/ (1,5-AG6P) than on G6P, whereas G6Pase-a prefers
G6Pase-b complex in neutrophil homeostasis and func- G6P to 1,5-AG6P [60]. The 1,5-AG6P is a G6P struc-
tion [19]. tural analog produced by the phosphorylation of 1,5-
Neutrophils from G6pt / mice show enhanced ER anhydroglucitol (1,5-AG). 1,5-AG is a glucose analog
stress, as identified by the increased expression of the present in the blood and food-derived polyol, although
molecular chaperones GRP78 and GRP170, and pro- some of the 1,5-AG in our body is produced from glu-
tein disulfide isomerase [58]. Similarly, neutrophils cose [61,62]. The 1,5-AG has been used as a bioche-
from G6pc3 / mice also exhibit increased ER stress mical marker for diabetes mellitus because its
and apoptosis [19]. Consistent with these results, char- concentration in the plasma is constant under the nor-
acterization of neutrophils from GSD-Ib and SCN4 moglycemic condition, while its reabsorption and the
patients demonstrated that defects in G6PT or plasma level dramatically decrease under the hyper-
G6Pase-b cause ER stress, leading to an increased rate glycemic conditions [63–65]. Although the metabolic
of apoptosis [20,21,23,58]. Furthermore, additional evi- pathway and the function of 1,5-AG are not fully
dence proved that mitochondrial oxidative stress partly understood, the structural similarity between glucose
contributes to the apoptosis of neutrophils in both and 1,5-AG raised the possibility that 1,5-AG could be
G6pt / and G6pc3 / mice [58,59]. taken up into the cells through the glucose trans-
In addition to neutropenia, both GSD-Ib and SCN4 porters [66].
patients also exhibited neutrophil dysfunctions, such as Once glucose and 1,5-AG enter the cell, they are
impaired respiratory burst activity, chemotaxis, cal- phosphorylated by glucose-phosphorylating enzymes,
cium mobilization, phagocytic activities, and migration such as hexokinase. It is well known that glucose is
[17,19]. Neutrophils cannot produce glucose via phosphorylated into G6P by hexokinase in most cells

6 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

or glucokinase in certain cells, including liver cells [15]. bone marrow into the blood, monocytes circulate in
In the case of 1,5-AG, it has been proposed that it the blood to survey the body for sites of inflammation
would be phosphorylated by at least two distinct [70]. When they encounter inflammatory stress signals
enzymes [62]. Vegia-da-Cunha et al. [60]also described including chemokines, complement components, and
that 1,5-AG is accidentally phosphorylated by glucose- the products of tissue matrix degradation, monocytes
phosphorylating enzymes, such as hexokinases and differentiate into the proinflammatory (M1) or anti-in-
ADP-glucokinase (ADP-GK). 1,5-AG6P is then trans- flammatory (M2) macrophages [67].
ported into the ER lumen by G6PT and dephosphory- Macrophages are found in almost every tissue in
lated by G6Pase-b, maintaining the physiological our body and show great functional diversity, from
intracellular level of 1,5-AG6P [60]. When 1,5-AG immune responses to pathogens to development,
administered to control, G6pt / , and G6pc3 / mice, homeostasis, and repair [68]. Tissue homeostasis is reg-
1,5-AG6P accumulated massively in the knockout neu- ulated by resident macrophages, which act as sentinels
trophils compared with the control ones. The accumu- and respond to changes in physiology as well as exter-
lation of 1,5-AG6P was shown to inhibit the nal challenges, such as injury or infection [68]. As a
enzymatic activities of hexokinases, resulting in response to environmental challenges, monocytes from
decreased glucose phosphorylation and reduced glu- blood, spleen, and bone marrow can be also recruited
cose metabolism. These results suggest that regulation and differentiated into macrophages in injured or
of 1,5-AG6P by G6PT/G6Pase-b is a critical factor for infected tissues [71].
the function and survival of neutrophils (Fig. 2). Interestingly, recent studies have shown that innate
immune cells, such as monocytes and macrophages,
can remember a previous encounter with foreign anti-
Cellular metabolism and function of
gens and contribute to the immunological memory of
monocytes and macrophages
our immune system whereby it mounts a faster and
Monocytes are members of the mononuclear phago- greater response when rechallenged by the same anti-
cytes, which comprise monocytes and two other major gens. These adaptive characteristics of the innate
subtypes: dendritic cells (DCs) and macrophages [67]. immune system have been called trained immunity [72]
They are characterized by a unilobular nucleus and because it enables innate immune cells to respond
have an important role in the innate immune system, more effectively to secondary stimuli after they are
like the neutrophils [68]. Monocytes and neutrophils stimulated with a pathogen [73,74]. This requires that
share some similar physiological roles, but marrow monocytes and macrophages undergo intracellular
and blood monocytes can also proliferate and differen- metabolic and epigenetic changes, the latter involving
tiate into macrophages [69]. Once released from the histone modifications [75].

Fig. 2. Proposed explanation for neutropenia and neutrophil dysfunction in GSD-Ib. In the G6PT / neutrophils, glucose and 1,5-AG that
have entered the cell are metabolized by hexokinase and hexokinase or ADP-GK, respectively. Loss of G6PT causes the massive
accumulation of cytosolic 1,5-AG6P, which inhibits the hexose-phosphorylating enzymes, such as hexokinases or ADP-GK. Even though
which hexose-phosphorylating enzymes are inhibited is unknown, this inhibition results in the reduction of further glucose utilization. This is
likely responsible for impaired energy homeostasis and functionality, as well as increased ER and oxidative stress.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 7
Role of G6PT in cell metabolism and function S. W. Sim et al.

M1 and M2 macrophages have recognized [17,18]. GSD-Ib monocytes exhibit


different metabolic phenotypes decreased PPP activities and respiratory burst, which
includes oxygen consumption, production of superox-
In the M1 inflammatory phenotype, activated macro- ide anion, and calcium mobilization when they are
phages have high microbicidal activity, as well as stimulated by fMLP or phorbol 12-myristate 13-ac-
enhanced production of proinflammatory cytokines etate [17]. However, the magnitude of impairment of
and ROS [76]. These macrophages exhibit a metabolic respiratory burst in monocytes is not as severe as that
shift from OXPHOS to glycolysis and PPP, enabling observed in G6PT-deficient neutrophils. Kilpatrick
them to generate enough ATP and biosynthetic materi- et al. described these differences as being due to the
als, which include amino acids for protein synthesis, different metabolic capabilities of neutrophils and
ribose for nucleotides, and NADPH for the produc- monocytes; neutrophils are highly dependent on gly-
tion of ROS by NADPH oxidase [77,78]. Pharmaco- colysis, while monocytes use mitochondria more than
logical inhibition of glycolysis blunted many effector neutrophils to produce ATP via OXPHOS [17].
functions of the M1 inflammatory phenotypes, includ- There has been no reported case of GSD-Ib macro-
ing phagocytosis, secretion of proinflammatory cytoki- phage dysfunction. However, G6pc3 / macrophages
nes, and the generation of ROS [79,80]. As with exhibit impaired respiratory burst, chemotaxis, calcium
glycolysis, some intermediates of the TCA cycle serve mobilization, and phagocytosis activities which result
as key biosynthetic materials for the effector functions from impaired energy homeostasis [22]. This suggests
of M1 macrophages [81,82]. The upregulation of gly- that G6PT deficiency can also cause macrophage dys-
colytic metabolism also generates pyruvate to fuel the function, although further research is required to clar-
TCA cycle, even though the M1 inflammatory macro- ify this point.
phages show impaired mitochondrial OXPHOS which
is disrupted in two places, after citrate and succinate,
respectively [83]. The citrate synthesis is increased to Metabolism and function of CD4+ T
produce fatty acids, lipids, and prostaglandins which cells
act as a proinflammatory mediator [82]. In addition to CD4+ T cells organize diverse aspects of adaptive
the accumulation of citrate, the M1 macrophages exhi- immunity, including responses to pathogens, allergens,
bit increased accumulation of succinate, which results and tumors, and it is accepted that establishment and
in the stabilization of HIF-1a, thereby promoting maintenance of immune responses, homeostasis, mem-
expression of LPS-induced IL-1b [84]. ory, and self-tolerance rely on them [88]. T-cell pro-
In contrast with the functional characteristics of the genitors in the thymus are derived from common
M1 macrophages, alternatively activated M2 macro- lymphoid progenitor present in the bone marrow [89].
phages are associated with tissue remodeling, resolution T-cell progenitors undergo several changes during T-
of inflammation, anti-inflammatory responses, and anti- cell maturation: generation of CD4+CD8+ double-posi-
gen presentation [77,78]. These M2 macrophages utilize tive thymocytes, positive and negative selection of dou-
an intact TCA cycle and enhanced mitochondrial ble-positive thymocytes, interaction of positively
OXPHOS to generate ATP [77]. They are involved in selected thymocytes with medullary thymic epithelial
time-consuming tissue repair and defense against extra- cells to complete T-cell development, and the export of
cellular parasites, suggesting that OXPHOS could be na€ıve mature T cells from the thymus [90]. Upon
more suitable for the role of M2 macrophage [85]. M2 recognition of a cognate antigen presented by compe-
macrophages also exhibit upregulation of the entire pro- tent antigen-presenting cells, na€ıve CD4+ T cells differ-
gram for fatty acid metabolism, including fatty acid entiate into distinct CD4+ effector T cells (Th1, Th2,
uptake, fatty acid oxidation, and mitochondrial biogen- and Th17 cells), CD4+ regulatory T cells (Tregs), and
esis [86,87]. These metabolic changes are regulated by memory T cells [91].
PPAR-c co-activator-1b, which is a transcriptional co- Na€ıve T cells have little energy demands and require
activator that upregulates expression of genes involved only basal biosynthesis for immune surveillance of sec-
in fatty acid oxidation [86]. ondary lymphoid tissues prior to activation [92]. To
meet this need, they rely on the fatty acid b-oxidation,
and pyruvate and glutamine oxidation through the
Role of G6PT in monocytes and
TCA cycle, which yield high energy [93]. Upon activa-
macrophages
tion, more energy and various substrates are necessary
Glycogen storage disease type Ib patients show mono- for cellular proliferation, which requires biosynthesis
cyte dysfunction, even if signs are not clearly of DNA, RNA, and protein [94]. These energy

8 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

requirements are fulfilled with a metabolic change been reported that chronic lymphopenia might affect
toward aerobic glycolysis and glutamine oxidation that the proliferation of immune cells, which react with
fuels mitochondrial OXPHOS through the TCA cycle self-antigens thereby promoting autoimmune diseases
[95,96]. Hereafter, T cells differentiate into various [102]. This is supported by other studies describing the
subsets, which includes effector, memory, and Treg relationship between autoimmune diseases and
[93]. Effector T cells maintain their metabolic pheno- immune deficiencies such as lymphopenia [103,104]. In
types in response to various cytokines, while the mem- lymphopenia conditions, T cells undergo peripheral
ory and Treg undergo metabolic changes [97]. Tregs proliferation even in the absence of foreign-antigen
switch their metabolism toward fatty acid oxidation to stimulation [104]. This compensatory homeostatic pro-
promote their growth and regulatory functions [98]. At liferation of T cells is known as lymphopenia-induced
the end of an immune response, memory T cells are proliferation (LIP), causing the restoration of the
produced and they showed similar cellular metabolism peripheral T-cell pool without the thymic output of
with na€ıve T cells [99] (Fig. 3A). na€ıve T cells [105,106]. Although the mechanism
underlying LIP is not fully understood, it has been
suggested that chronic lymphopenia might lead to the
Role of G6PT in CD4+ T cells
proliferation of highly self-reactive T cells, conse-
In addition to neutropenia and neutrophil dysfunction, quently causing autoimmunity [107,108].
GSD-Ib patients also show an increased risk for developing The higher frequency of autoimmune diseases in
autoimmune disorders, including chronic inflammatory GSD-Ib can also be attributed to the reduced suppres-
bowel disease, Crohn’s disease, thyroid autoimmunity, sive capacity of CD4+CD25+FOXP3+ Tregs and
and myasthenia gravis [16,38,39,100,101]. abnormal induction of FOXP3 in CD4+CD25
Recently, Melis et al. [25] found that lymphopenia Tconvs, which are caused by impaired glycolysis [25].
in GSD-Ib patients results from the aberrant glycolysis Tregs can be broadly classified into two groups: thy-
in CD4+CD25 conventional T cells (Tconvs). It has mic Tregs, also known as natural Tregs, which develop

Fig. 3. Cellular metabolism of various CD4+ T-cell subsets and G6PT / T cells. (A) Na€ıve or memory T cells use glucose-derived pyruvate to
fuel the OXPHOS pathway, along with fatty acids and glutamine. This metabolic program facilitates their survival and migration through the
lymphatic system for immune surveillance. Upon activation, glycolysis and glutaminolysis are increased to generate biosynthetic materials,
which are required for rapid proliferation, while fatty acid oxidation is decreased. Following the activation of T cells, CD4+ T cells
differentiate into the effector subsets (Th1, Th2, and Th17 cells) and FOXP3+ Treg. FOXP3+ Tregs preferentially utilize exogenously derived
fatty acids through fatty acid oxidation to fuel their regulatory function. (B) In the G6PT / T cells, reduced glycolysis upon TCR stimulation
of CD4+CD25 conventional T cells (Tconvs) and lymphopenia were observed. These phenotypes are associated with decreased expression
of transcription factor FOXP3 in CD4+CD25 Tconvs and reduced suppressive function of CD4+CD25+FOXP3+ Treg, which can explain an
increased risk of autoimmune disorders in GSD-Ib patients.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 9
Role of G6PT in cell metabolism and function S. W. Sim et al.

in the thymus, and induced Tregs (iTregs) generated characteristics were supported by a metabolic shift
from conventional CD4+ T cells in the periphery after toward glycolysis rather than OXPHOS, along with
antigen encounter [109]. Transcription factor FOXP3 the differential expressions of the glycolytic enzyme
is key for the development, and suppressive functions hexokinase 2 (HK2) and the mitochondrial anion car-
of both Tregs and mutations in this gene are strongly rier uncoupling protein 2 (UCP2) [26]. Furthermore,
linked to immune dysregulation, autoimmune polyen- G6PT deficiency in hMSC induced ER stress and
docrinopathy, and enteropathy [110–112]. Further- mitochondrial oxidative stress, which in turn led to
more, it was reported that the induction of FOXP3 cyclooxygenase-2 (COX-2)-induced prostaglandin E2
splicing variants containing exon 2 (FOXP3-E2) in (PGE2) production [26]. Considering that PGE2 pro-
human iTreg cells depends on glycolysis leading to the duction by MSCs increases cell proliferation through
development of iTregs from Tconvs following subopti- E-prostanoid 2 receptor in an autocrine manner as
mal stimulation via the TCR [113]. Collectively, the well as its immunoregulatory function [123], it is rea-
hyperactivity of autoreactive T cells and the failure of sonable to suggest that increased PGE2 production
local regulatory mechanisms by Tregs could be results in increased proliferation of G6PT / hMSC.
involved in the immunopathogenesis of autoimmune Taken together, G6PT plays an important role in regu-
diseases that are found in GSD-Ib patients. This lating the proliferation and differentiation of hMSC
strongly suggests that G6PT plays a vital role in the by regulating glucose homeostasis.
energy homeostasis and the function of lymphocytes, In addition to their differentiation capacity, the
as shown in myeloid cells [25,114,115] (Fig. 3B). immunomodulatory potential of MSCs has been
reported to regulate the immune reaction in various
diseases [30,124]. For example, MSCs inhibit the prolif-
Potential impact of G6PT /
eration and cytotoxic activity of the natural killer (NK)
mesenchymal stem cells on immune
cells, as well as cytokine production [125,126]. They
cells
also block the differentiation of monocytes into DCs,
Mesenchymal stem cells are a type of adult stem cells, alter mature DCs into immature DC states, and induce
characterized by fibroblast-like multipotency and the interleukin-10 (IL-10) production by plasmacytoid DCs
ability to differentiate into mesodermal tissues, such as (pDCs) [127–129]. In addition, they can inhibit the dif-
adipocytes, osteoblasts, and chondrocytes [27]. They ferentiation of Treg [130,131]. Considering that these
can be isolated from several tissues including bone effects are mainly mediated by PGE2 and its secretion
marrow and adipose tissue [116]. Throughout our is upregulated in G6PT / hMSCs, the inactivation of
body, MSCs home to sites of tissue injury and repair G6PT might affect not only immune deficiencies but
the tissue by either two processes: differentiation into also the regulation of the immune responses including
tissue-specific cell types, or creation of microenviron- autoimmune diseases by MSCs (Fig. 4).
ment that enhances tissue repair of the endogenous
cells and modulates the immune responses
Clinical therapies for immune
[30,117,118]. Therefore, cell-based regenerative thera-
deficiencies in GSD-Ib patients
pies that exploit MSCs are considered as a promising
approach in a variety of diseases [119]. The current treatment for GSD-Ib patients involves
Mesenchymal stem cells reside in hypoxic environ- dietary therapy involving frequent oral administration
ments in vivo, such as the bone marrow and other of uncooked cornstarch (a slow-releasing glucose
niches [120]. Compared to differentiated osteoblasts, preparation) [132]. In addition to this, administration
MSCs exhibit more glycolytic and lower levels of of granulocyte colony-stimulating factor (G-CSF) is
OXPHOS metabolism, indicating that MSCs depend commonly used to increase absolute neutrophil counts
more on glycolysis than do osteoblasts [121]. This and to alleviate the severity of recurrent bacterial
metabolic state may minimize ROS production to pre- infections in GSD-Ib and SCN4 patients [31,38,133].
vent oxidative stress-induced senescence and enable The combined dietary and G-CSF therapy significantly
MSCs to preserve long-term self-renewal [122]. During improves the abnormal metabolic state and neutrope-
adipogenesis and osteogenesis, MSCs require increased nia of GSD-Ib patients [132,133]. Above the ameliora-
mitochondrial biogenesis and OXPHOS, while glycoly- tion of neutropenia, numerous studies have examined
sis is upregulated during chondrogenesis [5]. We previ- whether G-CSF can also rescue neutrophil function
ously reported that a G6PT / hMSC line exhibits with contrasting outcomes [32,134–136]. Furthermore,
increased proliferation and decreased differentiation the toxicity of G-CSF therapy must also be considered
into adipocytes and osteoblasts [26]. These aberrant because it has been reported that long-term treatment

10 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

Fig. 4. The effect of G6PT deficiency on hMSC metabolism and immunomodulation capacities of MSCs by PGE2 secretion. (A) G6PT
deficiency in hMSCs caused increased proliferation and impaired differentiation into adipocytes and osteocytes. These phenotypes were
associated with a metabolic shift toward glycolysis and stress-induced PGE2 secretion. Metabolic changes encompass increased glycolysis
and decreased OXPHOS, which was explained with increased expression of HK2 including Akt/GSK-3b signaling pathway and decreased
expression of UCP2. Furthermore, disruption of G6PT triggers an ER and mitochondrial oxidative stress that lead to COX-2-induced PGE2
production. (B) PGE2 secretion by normal MSCs can modulate various functions of some immune cells. MSCs can suppress the
proliferation, cytotoxicity, and cytokine production of resting NK cells. MSCs can also inhibit the differentiation into DC from monocytes, and
maturation and tumor necrosis factor (TNF) production of DCs s and upregulate the expression of interleukin-10 (IL-10) by pDCs. Finally, the
effect of MSC-derived PGE2 secretion includes defective activation of CD4+ T cells.

with G-CSF can induce myelodysplasia or acute mye- patients [146,147]. Although studies on the long-term
loid leukemia [137–140]. Concerning the improvement efficacy, safety, and accessibility to donor cells are
of the neutrophil functions, Jun et al. [13] showed that underway, this would be one of the most promising
the addition of the PPAR-c antagonist GW9662 signif- options for future treatments for GSD-Ib patients.
icantly enhanced fMLP-mediated chemotaxis, calcium
mobilization, and superoxide production. Therefore,
Summary and perspectives
the combinational use of both G-CSF and GW9662
could be considered to improve neutropenia and neu- Glycogen storage disease type Ib is an autosomal
trophil dysfunctions of GSD-Ib patients. genetic disorder that leads to metabolic and myeloid
As a viable therapeutic option, the potential of gene abnormalities. G6PT is a ubiquitously expressed ER-
therapy for GSD-Ib has been evaluated [141]. This resident G6P transporter that transfers cytoplasmic
study revealed that an adenoviral vector carrying a G6P into the lumen of the ER. In immune cells, G6PT
human G6PT infusion transiently corrects both abnor- and G6Pase-b are essential for the hydrolysis of G6P
mal metabolism and myeloid abnormalities in G6pt / into glucose, regulating endogenous glucose produc-
mice. Although studies on the long-term efficacy and tion. This metabolic regulatory role of G6PT/G6Pase-
safety of this therapy are still necessary, genetic correc- b becomes more important as the unrecognized role of
tion seems to be a promising treatment option for GSD- G6Pase-b is uncovered. G6PT / neutrophils exhibit
Ib patients. Most recent gene therapy development increased ER-induced apoptosis, mitochondrial oxida-
using adeno-associated virus vectors (AAV-hG6PT) on tive stress, and decreased energy metabolism, which
G6pt / mice showed improved survival rate and glu- drives functional impairment through the HIF-1a/
cose homeostasis profiles. However, AAV-hG6PT was PPAR-c pathway. In addition, G6PT / monocytes
not sufficient to correct the immune deficiency (neu- show impaired respiratory burst, suggesting that the
tropenia and neutrophil dysfunction) and further inves- function of G6PT in glucose homeostasis can influence
tigation is needed to achieve effective viral vector- other immune cell types. In concordance with this
mediated delivery of therapeutic genes into cell types observation, it was reported that G6PT plays a critical
other than hepatocytes [142]. The deficiency of ubiqui- role in regulating the suppressive activity of Treg, pro-
tously expressed G6PT may need a multidirectional viding evidence for an increased autoimmune risk in
approach [143]. Stem cell transplantation is emerging as GSD-Ib. In addition to immune cells, G6PT deficiency
an attractive option for inheritable genetic deficiencies in hMSCs leads to a metabolic shift toward glycolysis
[144,145], and there are documented cases of successful and defects in differentiation capacities into adipocytes
hematopoietic stem cell transplantations in GSD-Ib and osteoblasts, upregulating COX-2-induced PGE2

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 11
Role of G6PT in cell metabolism and function S. W. Sim et al.

secretion. With an immunomodulatory activity, G6PT- 4 Pearce EL and Pearce EJ (2013) Metabolic pathways
deficient hMSCs might also have an impact on in immune cell activation and quiescence. Immunity 38,
immune regulation. 633–643.
The biological role of G6PT in diverse cell types is 5 Shyh-Chang N, Daley GQ and Cantley LC (2013)
being elucidated; G6PT-mediated metabolism and Stem cell metabolism in tissue development and aging.
intermediate metabolic products are critical for cell Development 140, 2535–2547.
proliferation, differentiation, functions, and cell death. 6 Zmora N, Bashiardes S, Levy M and Elinav E (2017)
However, molecular mechanisms underlying the com- The role of the immune system in metabolic health and
disease. Cell Metab 25, 506–521.
plex phenotypes observed in cells of GSD-Ib remain to
7 Chou JY, Matern D, Mansfield BC and Chen YT
be investigated. Moreover, considering that cells from
(2002) Type I glycogen storage diseases: disorders of
different tissues or organs are confronted with a large
the glucose-6-phosphatase complex. Curr Mol Med 2,
variety of microenvironments and diverse metabolic
121–143.
fuels, it is possible that the role of G6PT may be sensi-
8 Lei KJ, Shelly LL, Pan CJ, Sidbury JB and Chou JY
tive to different microenvironments. G6PT-deficient (1993) Mutations in the glucose-6-phosphatase gene
mouse model and cell lines are available, and their that cause glycogen storage disease type 1a. Science
phenotypes should be characterized in detail, focusing 262, 580–583.
particularly on metabolism and metabolic products. 9 Lei KJ, Pan CJ, Shelly LL, Liu JL and Chou JY
These studies may give valuable insights as to the (1994) Identification of mutations in the gene for
development of novel therapies for metabolic and glucose-6-phosphatase, the enzyme deficient in
functional correction of cells, such as neutrophils and glycogen storage disease type 1a. J Clin Invest 93,
macrophages in GSD-Ib. 1994–1999.
10 Hiraiwa H, Pan CJ, Lin B, Moses SW and Chou JY
(1999) Inactivation of the glucose 6-phosphate
Acknowledgements
transporter causes glycogen storage disease type 1b. J
This work was supported by Basic Science Research Biol Chem 274, 5532–5536.
Program through the National Research Foundation 11 Gerin I, Veiga-da-Cunha M, Achouri Y, Collet JF and
of Korea (NRF) funded by the Ministry of Education, Van Schaftingen E (1997) Sequence of a putative
Science and Technology (2016R1D1A3B03931840). glucose 6-phosphate translocase, mutated in glycogen
Additional support was provided by Cooperative storage disease type Ib. FEBS Lett 419, 235–238.
Research Program for Agriculture Science & Technol- 12 Chen SY, Pan CJ, Nandigama K, Mansfield BC,
ogy Development (Project No. PJ01323004) and the Ambudkar SV and Chou JY (2008) The glucose-6-
phosphate transporter is a phosphate-linked antiporter
Global Center for GSD for grant support along with
deficient in glycogen storage disease type Ib and Ic.
the following GSD-Ib philanthropy funds managed by
FASEB J 22, 2206–2213.
the University of Connecticut Foundation: the Jonah
13 Jun HS, Weinstein DA, Lee YM, Mansfield BC and
Pournazarian fund and the McMillan GSD-Ib research
Chou JY (2014) Molecular mechanisms of neutrophil
fund.
dysfunction in glycogen storage disease type Ib. Blood
123, 2843–2853.
Declaration of financial disclosure 14 Shieh JJ, Pan CJ, Mansfield BC and Chou JY (2003)
A glucose-6-phosphate hydrolase, widely expressed
The authors have no financial interest to disclose. outside the liver, can explain age-dependent resolution
of hypoglycemia in glycogen storage disease type Ia. J
References Biol Chem 278, 47098–47103.
15 Chou JY, Jun HS and Mansfield BC (2010) Glycogen
1 Ganeshan K and Chawla A (2014) Metabolic storage disease type I and G6Pase-beta deficiency:
regulation of immune responses. Annu Rev Immunol etiology and therapy. Nat Rev Endocrinol 6, 676–688.
32, 609–634. 16 Visser G, Rake JP, Fernandes J, Labrune P, Leonard
2 DeBerardinis RJ, Lum JJ, Hatzivassiliou G and JV, Moses S, Ullrich K and Smit GP (2000)
Thompson CB (2008) The biology of cancer: metabolic Neutropenia, neutrophil dysfunction, and
reprogramming fuels cell growth and proliferation. Cell inflammatory bowel disease in glycogen storage disease
Metab 7, 11–20. type Ib: results of the European Study on Glycogen
3 Fox CJ, Hammerman PS and Thompson CB (2005) Storage Disease type I. J Pediatr 137, 187–191.
Fuel feeds function: energy metabolism and the T-cell 17 Kilpatrick L, Garty BZ, Lundquist KF, Hunter K,
response. Nat Rev Immunol 5, 844–852. Stanley CA, Baker L, Douglas SD and Korchak HM

12 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

(1990) Impaired metabolic function and signaling potential of adult human mesenchymal stem cells.
defects in phagocytic cells in glycogen storage disease Science 284, 143–147.
type 1b. J Clin Invest 86, 196–202. 28 Bernardo ME and Fibbe WE (2013) Mesenchymal
18 McCawley LJ, Korchak HM, Cutilli JR, Stanley CA, stromal cells: sensors and switchers of inflammation.
Baker L, Douglas SD and Kilpatrick L (1993) Cell Stem Cell 13, 392–402.
Interferon-gamma corrects the respiratory burst defect 29 Le Blanc K and Mougiakakos D (2012) Multipotent
in vitro in monocyte-derived macrophages from mesenchymal stromal cells and the innate immune
glycogen storage disease type 1b patients. Pediatr Res system. Nat Rev Immunol 12, 383–396.
34, 265–269. 30 Uccelli A, Moretta L and Pistoia V (2008)
19 Cheung YY, Kim SY, Yiu WH, Pan CJ, Jun HS, Ruef Mesenchymal stem cells in health and disease. Nat Rev
RA, Lee EJ, Westphal H, Mansfield BC and Chou JY Immunol 8, 726–736.
(2007) Impaired neutrophil activity and increased 31 Visser G, Rake JP, Labrune P, Leonard JV, Moses S,
susceptibility to bacterial infection in mice lacking Ullrich K, Wendel U, Groenier KH and Smit GP
glucose-6-phosphatase-beta. J Clin Invest 117, 784–793. (2002) Granulocyte colony-stimulating factor in
20 Boztug K,Appaswamy G, Ashikov A, Sch€affer AA, glycogen storage disease type 1b. Results of the
Salzer U, Diestelhorst J, Germeshausen M, Brandes G, European Study on Glycogen Storage Disease Type 1.
Lee-Gossler J, Noyan F et al. (2009) A syndrome with Eur J Pediatr 161 (Suppl 1), S83–S87.
congenital neutropenia and mutations in G6PC3. N 32 McCawley LJ, Korchak HM, Douglas SD, Campbell
Engl J Med 360, 32–43. DE, Thornton PS, Stanley CA, Baker L and
21 Jun HS, Lee YM, Cheung YY, McDermott DH, Kilpatrick L (1994) In vitro and in vivo effects of
Murphy PM, De Ravin SS, Mansfield BC and Chou granulocyte colony-stimulating factor on neutrophils in
JY (2010) Lack of glucose recycling between glycogen storage disease type 1B: granulocyte colony-
endoplasmic reticulum and cytoplasm underlies cellular stimulating factor therapy corrects the neutropenia and
dysfunction in glucose-6-phosphatase-beta-deficient the defects in respiratory burst activity and Ca2+
neutrophils in a congenital neutropenia syndrome. mobilization. Pediatr Res 35, 84–90.
Blood 116, 2783–2792. 33 Li AM, Thyagu S, Maze D, Schreiber R, Sirrs S,
22 Jun HS, Cheung YY, Lee YM, Mansfield BC and Stockler-Ipsiroglu S, Sutherland H, Vercauteren S and
Chou JY (2012) Glucose-6-phosphatase-beta, Schultz KR (2018) Prolonged granulocyte colony
implicated in a congenital neutropenia syndrome, is stimulating factor use in glycogen storage disease type
essential for macrophage energy homeostasis and 1b associated with acute myeloid leukemia and with
functionality. Blood 119, 4047–4055. shortened telomere length. Pediatr Hematol Oncol 35,
23 McDermott DH, De Ravin SS, Jun HS, Liu Q, Priel 45–51.
DAL, Noel P, Takemoto CM, Ojode T, Paul SM, 34 Annabi B, Mansfield BC, Hiraiwa H, Lei K-J, Ubagai
Dunsmore KP et al. (2010) Severe congenital T, Polymeropoulos MH, Moses SW, Parvari R,
neutropenia resulting from G6PC3 deficiency with Hershkovitz E, Mandel H et al. (1998) The gene for
increased neutrophil CXCR4 expression and glycogen-storage disease type 1b maps to chromosome
myelokathexis. Blood 116, 2793–2802. 11q23. Am J Hum Genet 62, 400–405.
24 Banka S and Newman WG (2013) A clinical and 35 Gerin I, Veiga-da-Cunha M, Noel G and Van
molecular review of ubiquitous glucose-6-phosphatase Schaftingen E (1999) Structure of the gene mutated in
deficiency caused by G6PC3 mutations. Orphanet J glycogen storage disease type Ib. Gene 227, 189–195.
Rare Dis 8, 84. 36 Lin B, Pan CJ and Chou JY (2000) Human variant
25 Melis D, Carbone F, Minopoli G, La Rocca C, Perna glucose-6-phosphate transporter is active in
F, De Rosa V, Galgani M, Andria G, Parenti G and microsomal transport. Hum Genet 107, 526–529.
Matarese G (2017) Cutting edge: increased 37 Pan CJ, Lin B and Chou JY (1999) Transmembrane
autoimmunity risk in glycogen storage disease type 1b topology of human glucose 6-phosphate transporter. J
Is associated with a reduced engagement of glycolysis Biol Chem 274, 13865–13869.
in T cells and an impaired regulatory T cell function. J 38 Dieckgraefe BK, Korzenik JR, Husain A and Dieruf L
Immunol 198, 3803–3808. (2002) Association of glycogen storage disease 1b and
26 Sim SW, Park TS, Kim SJ, Park BC, Weinstein DA, Crohn disease: results of a North American survey.
Lee YM and Jun HS (2018) Aberrant proliferation Eur J Pediatr 161 (Suppl 1), S88–92.
and differentiation of glycogen storage disease type Ib 39 Melis D, Pivonello R, Parenti G, Casa RD, Salerno
mesenchymal stem cells. FEBS Lett 592, 162–171. M, Lombardi G, Sebastio G, Colao A and Andria G
27 Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, (2007) Increased prevalence of thyroid autoimmunity
Douglas R, Mosca JD, Moorman MA, Simonetti DW, and hypothyroidism in patients with glycogen storage
Craig S and Marshak DR (1999) Multilineage disease type I. J Pediatr 150, 300–305.e1.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 13
Role of G6PT in cell metabolism and function S. W. Sim et al.

40 Chou JY and Mansfield BC (2014) The SLC37 family 55 Stanton RC (2012) Glucose-6-phosphate
of sugar-phosphate/phosphate exchangers. Curr Top dehydrogenase, NADPH, and cell survival. IUBMB
Membr 73, 357–382. Life 64, 362–369.
41 Chen LY, Lin B, Pan CJ, Hiraiwa H and Chou JY 56 Bao Y, Ledderose C, Seier T, Graf AF, Brix B, Chong
(2000) Structural requirements for the stability and E and Junger WG (2014) Mitochondria regulate
microsomal transport activity of the human glucose 6- neutrophil activation by generating ATP for autocrine
phosphate transporter. J Biol Chem 275, 34280–34286. purinergic signaling. J Biol Chem 289, 26794–26803.
42 Chen LY, Pan CJ, Shieh JJ and Chou JY (2002) 57 Kuijpers TW, Maianski NA, Tool AT, Smit GP, Rake
Structure-function analysis of the glucose-6-phosphate JP, Roos D and Visser G (2003) Apoptotic neutrophils
transporter deficient in glycogen storage disease type in the circulation of patients with glycogen storage
Ib. Hum Mol Genet 11, 3199–3207. disease type 1b (GSD1b). Blood 101, 5021–5024.
43 Pan CJ, Chen LY, Mansfield BC, Salani B, Varesio L and 58 Kim SY, Jun HS, Mead PA, Mansfield BC and Chou
Chou JY (2003) The signature motif in human glucose-6- JY (2008) Neutrophil stress and apoptosis underlie
phosphate transporter is essential for microsomal transport myeloid dysfunction in glycogen storage disease type
of glucose-6-phosphate. Hum Genet 112, 430–433. Ib. Blood 111, 5704–5711.
44 Mishalian I, Granot Z and Fridlender ZG (2017) The 59 Jun HS, Lee YM, Song KD, Mansfield BC and Chou
diversity of circulating neutrophils in cancer. JY (2011) G-CSF improves murine G6PC3-deficient
Immunobiology 222, 82–88. neutrophil function by modulating apoptosis and
45 Borregaard N (2010) Neutrophils, from marrow to energy homeostasis. Blood 117, 3881–3892.
microbes. Immunity 33, 657–670. 60 Veiga-da-Cunha M, Chevalier N, Stephenne X, Defour
46 Walmsley SR, Print C, Farahi N, Peyssonnaux C, J-P, Paczia N, Ferster A, Achouri Y, Dewulf JP,
Johnson RS, Cramer T, Sobolewski A, Condliffe AM, Linster CL, Bommer GT et al. (2019) Failure to
Cowburn AS, Johnson N et al. (2005). Hypoxia-induced eliminate a phosphorylated glucose analog leads to
neutrophil survival is mediated by HIF-1alpha-dependent neutropenia in patients with G6PT and G6PC3
NF-kappaB activity. J Exp Med 201, 105–115. deficiency. Proc Natl Acad Sci USA 116, 1241–1250.
47 Mayadas TN, Cullere X and Lowell CA (2014) The 61 Yu S (2008) The anhydrofructose pathway of glycogen
multifaceted functions of neutrophils. Annu Rev Pathol catabolism. IUBMB Life 60, 798–809.
9, 181–218. 62 Mizuno H, Morita M and Akanuma H (1995)
48 Rane D, Patil T, More V, Patra SS, Bodhale N, Phosphorylation of 1,5-anhydro-D-glucitol in
Dandapat J and Sarkar A (2018) Neutrophils: mammalian cells. J Biochem 118, 411–417.
interplay between host defense, cellular metabolism 63 Akanuma H, Ogawa K, Lee Y and Akanuma Y (1981)
and intracellular infection. Cytokine 112, 44–51. Reduced levels of plasma 1,5-anhydroglucitol in
49 Borregaard N and Herlin T (1982) Energy metabolism diabetic patients. J Biochem 90, 157–162.
of human neutrophils during phagocytosis. J Clin 64 Yamanouchi T, Akanuma H, Asano T, Konishi C,
Invest 70, 550–557. Akaoka I and Akanuma Y (1987) Reduction and
50 Fossati G, Moulding DA, Spiller DG, Moots RJ, recovery of plasma 1,5-anhydro-D-glucitol level in
White MR and Edwards SW (2003) The mitochondrial diabetes mellitus. Diabetes 36, 709–715.
network of human neutrophils: role in chemotaxis, 65 Kim WJ and Park CY (2013) 1,5-Anhydroglucitol in
phagocytosis, respiratory burst activation, and diabetes mellitus. Endocrine 43, 33–40.
commitment to apoptosis. J Immunol 170, 1964–1972. 66 Yamanouchi T, Tachibana Y, Sekino N, Akanuma H,
51 Maianski NA, Geissler J, Srinivasula SM, Alnemri ES, Akaoka I and Miyashita H (1994) Transport and
Roos D and Kuijpers TW (2004) Functional accumulation of 1,5-anhydro-D-glucitol in the human
characterization of mitochondria in neutrophils: a role erythroleukemia cell line K-562. J Biol Chem 269,
restricted to apoptosis. Cell Death Differ 11, 143–153. 9664–9668.
52 Tan AS, Ahmed N and Berridge MV (1998) Acute 67 Jakubzick CV, Randolph GJ and Henson PM (2017)
regulation of glucose transport after activation of Monocyte differentiation and antigen-presenting
human peripheral blood neutrophils by phorbol functions. Nat Rev Immunol 17, 349–362.
myristate acetate, fMLP, and granulocyte-macrophage 68 Wynn TA, Chawla A and Pollard JW (2013)
colony-stimulating factor. Blood 91, 649–655. Macrophage biology in development, homeostasis and
53 Boxer LA, Baehner RL and Davis J (1977) The effect disease. Nature 496, 445–455.
of 2-deoxyglucose on guinea pig polymorphonuclear 69 Dale DC, Boxer L and Liles WC (2008) The
leukocyte phagocytosis. J Cell Physiol 91, 89–102. phagocytes: neutrophils and monocytes. Blood 112,
54 Iyer GY and Questel JH (1963) NADPH and NADH 935–945.
oxidation by guinea pig polymorphonuclear leucocytes. 70 Kramer PA, Ravi S, Chacko B, Johnson MS and
Can J Biochem Physiol 41, 427–434. Darley-Usmar VM (2014) A review of the

14 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

mitochondrial and glycolytic metabolism in 82 Wei X, Song H, Yin L, Rizzo MG, Sidhu R, Covey
human platelets and leukocytes: implications for DF, Ory DS and Semenkovich CF (2016) Fatty acid
their use as bioenergetic biomarkers. Redox Biol synthesis configures the plasma membrane for
2, 206–210. inflammation in diabetes. Nature 539, 294–298.
71 Geissmann F, Manz MG, Jung S, Sieweke MH, 83 O’Neill LA (2015) A broken krebs cycle in
Merad M and Ley K (2010) Development of macrophages. Immunity 42, 393–394.
monocytes, macrophages, and dendritic cells. Science 84 Tannahill GM,Curtis AM, Adamik J, Palsson-
327, 656–661. McDermott EM, McGettrick AF, Goel G, Frezza C,
72 Netea MG, Quintin J and van der Meer JW (2011) Bernard NJ, Kelly B, Foley NH et al. (2013) Succinate
Trained immunity: a memory for innate host defense. is an inflammatory signal that induces IL-1beta
Cell Host Microbe 9, 355–361. through HIF-1alpha. Nature 496, 238–242.
73 Kleinnijenhuis J, Quintin J, Preijers F, Joosten LAB, 85 O’Neill LA and Hardie DG (2013) Metabolism of
Ifrim DC, Saeed S, Jacobs C, van Loenhout J, de Jong inflammation limited by AMPK and pseudo-
D, Stunnenberg HG et al. (2012) Bacille Calmette- starvation. Nature 493, 346–355.
Guerin induces NOD2-dependent nonspecific 86 Vats D, Mukundan L, Odegaard JI, Zhang L, Smith
protection from reinfection via epigenetic KL, Morel CR, Wagner RA, Greaves DR, Murray PJ
reprogramming of monocytes. Proc Natl Acad Sci and Chawla A (2006) Oxidative metabolism and PGC-
USA 109, 17537–17542. 1beta attenuate macrophage-mediated inflammation.
74 Quintin J, Saeed S, Martens JHA, Giamarellos- Cell Metab 4, 13–24.
Bourboulis EJ, Ifrim DC, Logie C, Jacobs L, Jansen 87 Huang SC, Everts B, Ivanova Y, O’Sullivan D,
T, Kullberg B-J, Wijmenga C (2012) Candida albicans Nascimento M, Smith AM, Beatty W, Love-Gregory
infection affords protection against reinfection via L, Lam WY, O’Neill CM et al. (2014) Cell-intrinsic
functional reprogramming of monocytes. Cell Host lysosomal lipolysis is essential for alternative activation
Microbe 12, 223–232. of macrophages. Nat Immunol 15, 846–855.
75 van der Heijden C, Noz MP, Joosten LAB, Netea 88 Kumar BV, Connors TJ and Farber DL (2018)
MG, Riksen NP and Keating ST (2018) Epigenetics Human T cell development, localization, and function
and trained immunity. Antioxid Redox Signal 29, throughout life. Immunity 48, 202–213.
1023–1040. 89 Koch U and Radtke F (2011) Mechanisms of T cell
76 Rodriguez-Prados JC, Traves PG, Cuenca J, Rico D, development and transformation. Annu Rev Cell Dev
Aragones J, Martin-Sanz P, Cascante M and Bosca L Biol 27, 539–562.
(2010) Substrate fate in activated macrophages: a 90 Takahama Y (2006) Journey through the thymus:
comparison between innate, classic, and alternative stromal guides for T-cell development and selection.
activation. J Immunol 185, 605–614. Nat Rev Immunol 6, 127–135.
77 Van den Bossche J, O’Neill LA and Menon D (2017) 91 Zhu J, Yamane H and Paul WE (2010) Differentiation
Macrophage immunometabolism: where are we of effector CD4 T cell populations (*). Annu Rev
(going)? Trends Immunol 38, 395–406. Immunol 28, 445–489.
78 Diskin C and Palsson-McDermott EM (2018) 92 Newton R, Priyadharshini B and Turka LA (2016)
Metabolic modulation in macrophage effector Immunometabolism of regulatory T cells. Nat Immunol
function. Front Immunol 9, 270. 17, 618–625.
79 Michl J, Ohlbaum DJ and Silverstein SC (1976) 2- 93 MacIver NJ, Michalek RD and Rathmell JC (2013)
Deoxyglucose selectively inhibits Fc and complement Metabolic regulation of T lymphocytes. Annu Rev
receptor-mediated phagocytosis in mouse peritoneal Immunol 31, 259–283.
macrophages. I. Description of the inhibitory effect. J 94 Michalek RD and Rathmell JC (2010) The metabolic
Exp Med 144, 1465–1483. life and times of a T-cell. Immunol Rev 236, 190–202.
80 Freemerman AJ, Johnson AR, Sacks GN, Milner JJ, 95 Hume DA, Radik JL, Ferber E and Weidemann MJ
Kirk EL, Troester MA, Macintyre AN, Goraksha- (1978) Aerobic glycolysis and lymphocyte
Hicks P, Rathmell JC and Makowski L (2014) transformation. Biochem J 174, 703–709.
Metabolic reprogramming of macrophages: glucose 96 Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch
transporter 1 (GLUT1)-mediated glucose metabolism E, Aghvanyan A, Turay AM and Frauwirth KA
drives a proinflammatory phenotype. J Biol Chem 289, (2010) Glutamine uptake and metabolism are
7884–7896. coordinately regulated by ERK/MAPK during T
81 Infantino V, Iacobazzi V, Palmieri F and Menga A lymphocyte activation. J Immunol 185, 1037–1044.
(2013) ATP-citrate lyase is essential for macrophage 97 Gerriets VA and Rathmell JC (2012) Metabolic
inflammatory response. Biochem Biophys Res Commun pathways in T cell fate and function. Trends Immunol
440, 105–111. 33, 168–173.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 15
Role of G6PT in cell metabolism and function S. W. Sim et al.

98 Michalek RD, Gerriets VA, Jacobs SR, Macintyre from a FoxP3-centered view to an epigenome-defined
AN, MacIver NJ, Mason EF, Sullivan SA, Nichols view of natural Treg cells. Immunol Rev 259, 192–205.
AG and Rathmell JC (2011) Cutting edge: distinct 112 Wildin RS, Smyk-Pearson S and Filipovich AH (2002)
glycolytic and lipid oxidative metabolic programs are Clinical and molecular features of the
essential for effector and regulatory CD4+ T cell immunodysregulation, polyendocrinopathy,
subsets. J Immunol 186, 3299–3303. enteropathy, X linked (IPEX) syndrome. J Med Genet
99 Loftus RM and Finlay DK (2016) 39, 537–545.
Immunometabolism: cellular metabolism turns immune 113 De Rosa V, Galgani M, Porcellini A, Colamatteo A,
regulator. J Biol Chem 291, 1–10. Santopaolo Ma, Zuchegna C, Romano A, De Simone
100 Melis D, Parenti G, Della Casa R, Sibilio M, Berni S, Procaccini C, La Rocca C et al. (2015) Glycolysis
Canani R, Terrin G, Cucchiara S and Andria G controls the induction of human regulatory T cells by
(2003) Crohn’s-like ileo-colitis in patients affected by modulating the expression of FOXP3 exon 2 splicing
glycogen storage disease Ib: two years’ follow-up of variants. Nat Immunol 16, 1174–1184.
patients with a wide spectrum of gastrointestinal signs. 114 Dejaco C, Duftner C, Grubeck-Loebenstein B and
Acta Paediatr 92, 1415–1421. Schirmer M (2006) Imbalance of regulatory T cells in
101 Melis D, Balivo F, Della Casa R, Romano A, human autoimmune diseases. Immunology 117, 289–
Taurisano R, Capaldo B, Riccardi G, Monsurr o M r, 300.
Parenti G and Andria G (2008) Myasthenia gravis in a 115 Dornmair K, Goebels N, Weltzien HU, Wekerle H
patient affected by glycogen storage disease type Ib: a and Hohlfeld R (2003) T-cell-mediated autoimmunity:
further manifestation of an increased risk for novel techniques to characterize autoreactive T-cell
autoimmune disorders? J Inherit Metab Dis 31 (Suppl receptors. Am J Pathol 163, 1215–1226.
2), S227–S231. 116 Hass R, Kasper C, Bohm S and Jacobs R (2011)
102 King C, Ilic A, Koelsch K and Sarvetnick N (2004) Different populations and sources of human
Homeostatic expansion of T cells during immune mesenchymal stem cells (MSC): a comparison of adult
insufficiency generates autoimmunity. Cell 117, 265– and neonatal tissue-derived MSC. Cell Commun Signal
277. 9, 12.
103 Gleeson PA, Toh BH and van Driel IR (1996) Organ- 117 Liechty KW, MacKenzie TC, Shaaban AF, Radu
specific autoimmunity induced by lymphopenia. A, Moseley AM, Deans R, Marshak DR and
Immunol Rev 149, 97–125. Flake AW (2000) Human mesenchymal stem cells
104 Merayo-Chalico J, Rajme-Lopez S, Barrera-Vargas A, engraft and demonstrate site-specific differentiation
Alcocer-Varela J, Diaz-Zamudio M and Gomez- after in utero transplantation in sheep. Nat Med 6,
Martin D (2016) Lymphopenia and autoimmunity: a 1282–1286.
double-edged sword. Hum Immunol 77, 921–929. 118 Hofstetter CP, Schwarz EJ, Hess D, Widenfalk J, El
105 Jameson SC (2002) Maintaining the norm: T-cell Manira A, Prockop DJ and Olson L (2002) Marrow
homeostasis. Nat Rev Immunol 2, 547–556. stromal cells form guiding strands in the injured spinal
106 Khoruts A and Fraser JM (2005) A causal link cord and promote recovery. Proc Natl Acad Sci USA
between lymphopenia and autoimmunity. Immunol 99, 2199–2204.
Lett 98, 23–31. 119 Liu Y and Ma T (2015) Metabolic regulation of
107 Ge Q, Rao VP, Cho BK, Eisen HN and Chen J (2001) mesenchymal stem cell in expansion and therapeutic
Dependence of lymphopenia-induced T cell application. Biotechnol Prog 31, 468–481.
proliferation on the abundance of peptide/ MHC 120 Muguruma Y, Yahata T, Miyatake H, Sato T, Uno T,
epitopes and strength of their interaction with T cell Itoh J, Kato S, Ito M, Hotta T and Ando K (2006)
receptors. Proc Natl Acad Sci USA 98, 1728–1733. Reconstitution of the functional human hematopoietic
108 Baccala R and Theofilopoulos AN (2005) The new microenvironment derived from human mesenchymal
paradigm of T-cell homeostatic proliferation-induced stem cells in the murine bone marrow compartment.
autoimmunity. Trends Immunol 26, 5–8. Blood 107, 1878–1887.
109 Dominguez-Villar M and Hafler DA (2018) 121 Chen CT, Shih YR, Kuo TK, Lee OK and Wei YH
Regulatory T cells in autoimmune disease. Nat (2008) Coordinated changes of mitochondrial
Immunol 19, 665–673. biogenesis and antioxidant enzymes during osteogenic
110 Li MO and Rudensky AY (2016) T cell receptor differentiation of human mesenchymal stem cells. Stem
signalling in the control of regulatory T cell Cells 26, 960–968.
differentiation and function. Nat Rev Immunol 16, 122 Pattappa G, Thorpe SD, Jegard NC, Heywood HK,
220–233. de Bruijn JD and Lee DA (2013) Continuous and
111 Morikawa H and Sakaguchi S (2014) Genetic and uninterrupted oxygen tension influences the colony
epigenetic basis of Treg cell development and function: formation and oxidative metabolism of human

16 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies
S. W. Sim et al. Role of G6PT in cell metabolism and function

mesenchymal stem cells. Tissue Eng Part C Methods Granulocyte and granulocyte-macrophage colony-
19, 68–79. stimulating factors for treatment of neutropenia in
123 Lee BC, Kim H-S, Shin T-H, Kang I, Lee JY, Kim J- glycogen storage disease type Ib. J Pediatr 119,
J, Kang HK, Seo Y, Lee S, Yu K-R et al. (2016) 748–754.
PGE2 maintains self-renewal of human adult stem 134 Buckle AM and Hogg N (1989) The effect of IFN-
cells via EP2-mediated autocrine signaling and its gamma and colony-stimulating factors on the
production is regulated by cell-to-cell contact. Sci Rep expression of neutrophil cell membrane receptors. J
6, 26298. Immunol 143, 2295–2301.
124 Gao F, Chiu SM, Motan DAl, Zhang Z, Chen L, Ji 135 Ishiguro A, Nakahata T, Shimbo T, Amano Y, Yasui
H-l, Tse H-F, Fu Q-l and Lian Q (2016) Mesenchymal K, Koike K and Komiyama A (1993) Improvement of
stem cells and immunomodulation: current status and neutropenia and neutrophil dysfunction by
future prospects. Cell Death Dis 7, e2062. granulocyte colony-stimulating factor in a patient with
125 Spaggiari GM, Capobianco A, Becchetti S, Mingari glycogen storage disease type Ib. Eur J Pediatr 152,
MC and Moretta L (2006) Mesenchymal stem cell- 18–20.
natural killer cell interactions: evidence that activated 136 Kim GY, Lee YM, Kwon JH, Jun HS and Chou J
NK cells are capable of killing MSCs, whereas MSCs (2017) Glycogen storage disease type Ib neutrophils
can inhibit IL-2-induced NK-cell proliferation. Blood exhibit impaired cell adhesion and migration. Biochem
107, 1484–1490. Biophys Res Commun 482, 569–574.
126 Spaggiari GM, Capobianco A, Abdelrazik H, 137 Donadieu J, Leblanc T, Bader Meunier B, Barkaoui
Becchetti F, Mingari MC and Moretta L (2008) M, Fenneteau O, Bertrand Y, Maier-Redelsperger M,
Mesenchymal stem cells inhibit natural killer-cell Micheau M, Stephan JL, Phillipe N et al. (2005)
proliferation, cytotoxicity, and cytokine production: Analysis of risk factors for myelodysplasias, leukemias
role of indoleamine 2,3-dioxygenase and prostaglandin and death from infection among patients with
E2. Blood 111, 1327–1333. congenital neutropenia. Experience of the French
127 Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD Severe Chronic Neutropenia Study Group.
and Mao N (2005) Human mesenchymal stem cells Haematologica 90, 45–53.
inhibit differentiation and function of monocyte- 138 Rosenberg PS, Alter BP, Bolyard AA, Bonilla MA,
derived dendritic cells. Blood 105, 4120–4126. Boxer LA, Cham B, Fier C, Freedman M,
128 Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R Kannourakis G, Kinsey S et al. (2006) The incidence
and Fibbe WE (2006) Mesenchymal stem cells inhibit of leukemia and mortality from sepsis in patients with
generation and function of both CD34+-derived and severe congenital neutropenia receiving long-term G-
monocyte-derived dendritic cells. J Immunol 177, CSF therapy. Blood 107, 4628–4635.
2080–2087. 139 Schroeder T, Hildebrandt B, Mayatepek E, Germing
129 Ramasamy R, Fazekasova H, Lam EW, Soeiro I, U and Haas R (2008) A patient with glycogen storage
Lombardi G and Dazzi F (2007) Mesenchymal stem disease type Ib presenting with acute myeloid leukemia
cells inhibit dendritic cell differentiation and function (AML) bearing monosomy 7 and translocation t(3;8)
by preventing entry into the cell cycle. Transplantation (q26;q24) after 14 years of treatment with granulocyte
83, 71–76. colony-stimulating factor (G-CSF): a case report. J
130 Aggarwal S and Pittenger MF (2005) Human Med Case Rep 2, 319.
mesenchymal stem cells modulate allogeneic immune 140 Dale DC, Bolyard AA, Marrero T, Kelley ML,
cell responses. Blood 105, 1815–22. Makaryan V, Tran E, Leung J, Boxer LA, Kishnani
131 Zappia E, Casazza S, Pedemonte E, Benvenuto F, PS, Austin S et al. (2019) Neutropenia in glycogen
Bonanni I, Gerdoni E, Giunti D, Ceravolo A, storage disease Ib: outcomes for patients treated with
Cazzanti F, Frassoni F et al. (2005) Mesenchymal granulocyte colony-stimulating factor. Curr Opin
stem cells ameliorate experimental autoimmune Hematol 26, 16–21.
encephalomyelitis inducing T-cell anergy. Blood 106, 141 Yiu WH, Pan CJ, Allamarvdasht M, Kim SY and
1755–1761. Chou JY (2007) Glucose-6-phosphate transporter gene
132 Correia CE, Bhattacharya K, Lee PJ, Shuster JJ, therapy corrects metabolic and myeloid abnormalities
Theriaque DW, Shankar MN, Smit GP and Weinstein in glycogen storage disease type Ib mice. Gene Ther
DA (2008) Use of modified cornstarch therapy to 14, 219–226.
extend fasting in glycogen storage disease types Ia and 142 Kwon JH, Lee YM, Cho JH, Kim GY, Anduaga
Ib. Am J Clin Nutr 88, 1272–1276. J, Starost MF, Mansfield BC and Chou JY
133 Schroten H, Roesler J, Breidenbach T, Wendel U, (2017) Liver-directed gene therapy for murine
Elsner J, Schweitzer S, Zeidler C, Burdach S, glycogen storage disease type Ib. Hum Mol Genet
Lohmann-Matthes M-L, Wahn V et al. (1991) 26, 4395–4405.

FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies 17
Role of G6PT in cell metabolism and function S. W. Sim et al.

143 Yiu WH, Pan CJ, Mead PA, Starost MF, Mansfield 146 Mehyar LS, Abu-Arja R, Rangarajan HG, Pai V,
BC and Chou JY (2009) Normoglycemia alone is Bartholomew DW, Rose MJ and Bajwa RPS (2018)
insufficient to prevent long-term complications of Matched unrelated donor transplantation in glycogen
hepatocellular adenoma in glycogen storage disease storage disease type 1b patient corrects severe
type Ib mice. J Hepatol 51, 909–917. neutropenia and recurrent infections. Bone Marrow
144 Daley GQ and Scadden DT (2008) Prospects for stem Transplant 53, 1076–1078.
cell-based therapy. Cell 132, 544–548. 147 Pierre G, Chakupurakal G, McKiernan P, Hendriksz
145 Lengerke C and Daley GQ (2010) Autologous blood C, Lawson S and Chakrapani A (2008) Bone marrow
cell therapies from pluripotent stem cells. Blood Rev transplantation in glycogen storage disease type 1b. J
24, 27–37. Pediatr 152, 286–288.

18 FEBS Letters 594 (2020) 3–18 ª 2019 Federation of European Biochemical Societies

You might also like