Next Article in Journal
Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective
Next Article in Special Issue
Deficient Sarcolemma Repair in ALS: A Novel Mechanism with Therapeutic Potential
Previous Article in Journal
Liquid–Liquid Phase Separation in Cardiovascular Diseases
Previous Article in Special Issue
Patient-Derived Pancreatic Cancer Cells Induce C2C12 Myotube Atrophy by Releasing Hsp70 and Hsp90
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glucose 6-P Dehydrogenase—An Antioxidant Enzyme with Regulatory Functions in Skeletal Muscle during Exercise

by
Esther García-Domínguez
1,
Aitor Carretero
1,
Aurora Viña-Almunia
2,
Julio Domenech-Fernandez
3,
Gloria Olaso-Gonzalez
1,*,
Jose Viña
1 and
Mari Carmen Gomez-Cabrera
1
1
Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, 46010 Valencia, Spain
2
Centro de Salud San Isidro, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain
3
Health Care Department Arnau-Lliria, Servicio de Cirugía Ortopédica y Traumatología, Hospital Arnau de Vilanova y Hospital de Liria, 46015 Valencia, Spain
*
Author to whom correspondence should be addressed.
Submission received: 11 July 2022 / Revised: 21 September 2022 / Accepted: 23 September 2022 / Published: 28 September 2022
(This article belongs to the Special Issue Redox Control of Cell Signaling in Cardiac and Skeletal Muscle)

Abstract

:
Hypomorphic Glucose 6-P dehydrogenase (G6PD) alleles, which cause G6PD deficiency, affect around one in twenty people worldwide. The high incidence of G6PD deficiency may reflect an evolutionary adaptation to the widespread prevalence of malaria, as G6PD-deficient red blood cells (RBCs) are hostile to the malaria parasites that infect humans. Although medical interest in this enzyme deficiency has been mainly focused on RBCs, more recent evidence suggests that there are broader implications for G6PD deficiency in health, including in skeletal muscle diseases. G6PD catalyzes the rate-limiting step in the pentose phosphate pathway (PPP), which provides the precursors of nucleotide synthesis for DNA replication as well as reduced nicotinamide adenine dinucleotide phosphate (NADPH). NADPH is involved in the detoxification of cellular reactive oxygen species (ROS) and de novo lipid synthesis. An association between increased PPP activity and the stimulation of cell growth has been reported in different tissues including the skeletal muscle, liver, and kidney. PPP activity is increased in skeletal muscle during embryogenesis, denervation, ischemia, mechanical overload, the injection of myonecrotic agents, and physical exercise. In fact, the highest relative increase in the activity of skeletal muscle enzymes after one bout of exhaustive exercise is that of G6PD, suggesting that the activation of the PPP occurs in skeletal muscle to provide substrates for muscle repair. The age-associated loss in muscle mass and strength leads to a decrease in G6PD activity and protein content in skeletal muscle. G6PD overexpression in Drosophila Melanogaster and mice protects against metabolic stress, oxidative damage, and age-associated functional decline, and results in an extended median lifespan. This review discusses whether the well-known positive effects of exercise training in skeletal muscle are mediated through an increase in G6PD.

Graphical Abstract

1. The Pentose Phosphate Pathway and the Regulation of G6PD

Glucose is catabolized by two pathways: glycolysis, to generate ATP, and the pentose phosphate pathway (PPP), also known as the hexose monophosphate shunt, to generate reduced nicotinamide adenine dinucleotide phosphate (NADPH) and ribose 5-phosphate (R5P) for nucleotide synthesis.
The PPP was fully elucidated in the 1950s owing to the joint work of several researchers including Efraim Racker [1], Bernard Horecker [2], and Frank Dickens [3]. However, in the 1930s, Otto Warburg was the first to provide evidence of the existence of the PPP when he studied the oxidation of glucose 6-phosphate (G6P) to 6-phosphogluconate (6PG) and discovered NADP+ [4].
The PPP occurs entirely in the cytosol and has both anabolic and catabolic functions. It takes place through two phases: the oxidative branch, which is irreversible, and the non-oxidative branch, which is reversible [5] (See Figure 1).
The PPP competes with glycolysis for the catabolism of glucose 6-P (G6P). In the oxidative PPP branch, G6P is converted to ribulose-5P with the loss of CO2 and the formation of two NADPH molecules. Glucose 6-P dehydrogenase (G6PD) catalyzes the first committed step of this PPP branch, which involves the conversion of G6P to 6PG and the generation of the first NADPH molecule. This irreversible reaction is unique to the PPP and has a primary role in the regulation of this pathway [6]. NADPH can be also synthesized by other enzymes such as the NADPH-malic enzyme, NADPH-dependent isocitrate dehydrogenase, and transhydrogenases [7].
Across all forms of life, NADPH donates high-energy electrons for reductive biosynthesis and antioxidant defense [8]. One of the main functions of NADPH in our cells is in the maintenance of redox homeostasis [9]. NADPH is the electron donor for the antioxidant enzymes glutathione reductase (GR) and thioredoxin reductases (TrxR). Reduced glutathione (GSH) and reduced thioredoxin (Trx(SH)2) provide reducing equivalents for glutathione peroxidase (GPx), glutaredoxins (Grx), and peroxiredoxins (Prx). Thus, NADPH is located at the core of the antioxidant defense [10]. Another function of the pyridine nucleotide NADPH is to boost biosynthetic reactions in our cells. It provides the reducing power for fatty acids and cholesterol synthesis [9]. Finally, NADPH acts as the coenzyme of NADPH-oxidase enzymes (NOXs), which—through the generation of the superoxide radical—are involved in the oxidative burst and its defensive functions in our immune cells (granulocytes and macrophages) [11], and even in other cellular types [12,13]. Nitric oxide synthases (NOS), dihydrofolate reductase (DHFR), and cytochrome P450 oxidoreductase are also NADPH-dependent enzymes (Figure 2).
The non-oxidative PPP is a flexible pathway that is able to adapt to varying cellular needs through the generation of different phosphorylated carbohydrates with three, four, five, or seven carbons [14]. This branch begins with a bifurcation: the ribulose-5P obtained from the oxidative PPP after epimerization is transformed into xylulose 5-phosphate or can isomerize and form ribose-5-phosphate, which can be used for nucleotide synthesis. The main modes of the PPP depending on cellular needs are summarized in Figure 3 [15]. Although the corresponding stoichiometric reaction for each mode is shown, the carbon flux is difficult to quantify in cells [15], and a situation in which all the flux is directed to one is unlikely. On the other hand, mode 3, also known as the recycling PPP mode, uses steps from gluconeogenesis; therefore, it only can take place in cells containing fructose biphosphatase. Mode 4 represents the standard operation of the pathway.
As previously mentioned, G6PD is the key enzyme in the regulation of the PPP. Therefore, any factor able to modify the level or activity of G6PD will determine the flow of the PPP. The “coarse control” of the PPP is carried out by modifications in the levels, location, and activity of G6PD [16]. Factors such as diet composition can induce changes in the synthesis of G6PD [17,18]. An excess of carbohydrates in the diet leads to lipogenesis and the deposition of fat, which is associated with a 5–10-fold increase in G6PD activity in the liver [16]. Accordingly, the expression of the G6PD gene is upregulated by the major transcription factor sterol-responsive element binding protein [19]. High insulin and low glucagon levels, which are associated with this kind of diet, have also been described as regulators of G6PD through the control of mRNA synthesis [20]. The transcription factor Nrf2, which regulates the antioxidant cellular response, also enhances G6PD gene expression [21]. Interestingly, diets rich in polyunsaturated fatty acids (PUFAs) have the opposite effect on G6PD levels.
However, the main regulating factor for G6PD activity is the NADPH/NADP+ ratio. NADPH is a competitive inhibitor of G6PD [22], while NADP+ is required to maintain the conformation of G6PD and thus its catalytic activity [23]. If the NADPH/NADP+ ratio is increased, G6PD activity is lowered, and vice versa. In vitro experiments have shown no G6PD activity with NADPH/NADP+ ratios close to 10 [16]. The fact that the NADPH/NADP+ ratio in physiological conditions is high (70–300 depending on the tissue) [24] led Egglestone and Krebs to postulate the existence of a mechanism able to modulate the inhibition of G6PD by NADPH [16]. These authors, after testing the effect of over a hundred cell constituents, demonstrated that the physiological concentrations of oxidized glutathione (GSSG) and AMP were able to counteract the inhibition of G6PD by NADPH [16]. The inhibitory effect of GSSG on NADPH could not be attributed to GR activity since a complete inhibition of the enzyme did not abolish the GSSG effect. Recently, a similar role of GSSG in the “fine control” of the PPP has been assigned to a 100 kDa protein named CRING (cofactor that reverses the NADPH inhibition of G6PD). GSSG is required for CRING function. CRING is found in specific tissues including adipose, liver, and adrenal tissues [7]. Finally, the post-translational modification of G6PD also plays a role in its activity, as is the case with G6PD phosphorylation by the nonreceptor tyrosine kinase Src. As a consequence, the inhibition of Src causes reduced G6PD activity in endothelial cells [25].
Apart from the NADPH/NADP+ ratio, several other factors have been described as positive or negative regulators of G6PD; they are included in the last section of this manuscript.

2. Loss of Function Models for G6PD

G6PD deficiency is the most common human enzymopathy. It is very heterogeneous and was first described in humans by Marks and Gross in 1959 [26]. Approximately 400 million people worldwide carry a mutation in the G6PD gene, which causes an enzyme deficiency. Deficient alleles are prevalent in South and North America and in northern Europe [27]. However, the highest prevalence of this enzymopathy is reported in Africa, the Middle East, the central and southern Pacific Islands, southern Europe, and southeast Asia. The global distribution of the G6PD deficiency is strikingly similar to that of malaria. In areas where G6PD deficiency is common, Plasmodium Falciparum malaria is endemic, supporting the so-called malaria protection hypothesis [28]. Epidemiological evidence for the association between G6PD deficiency and a reduction in the risk of severe malaria [29] has been accompanied by the results of in vitro work showing that parasite growth is slowest in G6PD-deficient cells [28].
It has also been shown that G6PD-deficient red blood cells (RBCs) infected with parasites undergo macrophage-induced phagocytosis at an earlier stage of Plasmodium Falciparum maturation than normal RBCs. This could be a further protective mechanism against malaria [30]. The vulnerability of RBCs to mutant G6PD may reflect their lack of mitochondria and thus their inability to endogenously produce the substrates for malic enzyme and isocitrate dehydrogenase [8]. This may also reflect RBCs lack of nuclei and failure to replace the deficient G6PD protein as the cells age.
The G6PD gene is located at the telomeric region in the X chromosome. Thus, its deficiency is an X-linked hereditary defect that causes variants with different clinical phenotypes (about 140 mutations have been described). The G6PD-encoding gene has been well preserved throughout evolution [31]. As a monomer, the protein is inactive; however, as a dimer or tetramer, it is active. In its catalytic center, there is an amino acid sequence that binds to NADPH. The deficiency is caused by protein instability due to amino acid substitutions in different enzyme locations [28]. The diagnosis of G6PD deficiency is based on the spectrophotometric quantification of the enzyme’s activity [32]. There are five categories of G6PD deficiency based on clinical manifestations and enzyme activity (Table 1) [28].
The most frequent clinical manifestations of G6PD deficiency are acute and chronic hemolytic anemia and neonatal jaundice [28]. The prevention of hemolysis by avoiding oxidative stress represents the most effective management of G6PD deficiency. Oxidative stress can be triggered by agents such as drugs (primaquine, sulfonamide, or acetanilide), infections (hepatitis viruses, cytomegalovirus, or pneumonia), or the ingestion of fava beans (favism). Favism is a hemolytic response to the consumption of fava beans that takes place in some individuals with G6PD deficiency [33]. Isouramil, divicine, and convicine are thought to be the toxic constituents of fava beans that lead to the onset of the clinical manifestations of deficiency [28]. The mechanism by which increased sensitivity to oxidative damage leads to hemolysis has not been fully elucidated [34].
Several clinical disorders, such as diabetes and myocardial infarction, precipitate hemolysis in G6PD-deficient subjects [35,36].
G6PD is ubiquitously expressed in mammalian cells, with the highest expression observed in the immune cells, testes, adrenals, and brain [37]. It is often upregulated in tumors [9,38]. The enzyme is subject to tissue-specific transcriptional regulation, which in turn is correlated with the methylation of specific sites in the gene [37]. We recently found that immune cells, and especially T cells, are dependent on G6PD to maintain NADPH levels and effector functions [8]. Activated T cells do not express substantial levels of malic enzyme or isocitrate dehydrogenase and produce NADPH mainly through the PPP, which is sharply upregulated during T cell activation and is related to pro-inflammatory cytokine production [8]. Thus, severe G6PD mutations that affect the enzyme’s catalytic ability can present as immune deficiency [39].
Favism has a higher incidence in males than females [28]. Males are hemizygous for the G6PD gene and thus can have normal gene expression or be G6PD deficient. Females, with two copies of the G6PD gene on each X chromosome, can have normal gene expression, be homozygous, or be heterozygous. Heterozygous females can achieve the same degree of G6PD deficiency and can be susceptible to the same pathophysiological phenotype present in G6PD-deficient males. However, heterozygous women on average have less severe clinical manifestations than G6PD-deficient males [28].
The World Health Organization Scientific Group has emphasized the need to develop animal research models for this frequent human hereditary disorder. Genetically, G6PD knockout mice are not viable [40]. Mouse viability is dependent on G6PD activity, as evidenced by a decrease in litter size corresponding to a decrease in G6PD activity [41].
In 1988, Merkle and coworkers created the first X-linked G6PD deficient animal model using 1-ethyl-l-nitrosourea-induced chemical mutagenesis [42]. Williams’ research team reported a single point mutation (A to T transversion) at the 3′ end of exon 1 that explained the decrease in G6PD activity in the G6PD-deficient mice [43].
Heterozygous, hemizygous, and homozygous mutants have ~60%, ~15%, and ~15% of remaining precipitate activity in RBCs, respectively, when compared to wild type (WT) mice. Therefore, in comparison with the human classification of G6PD mutations, the mouse mutant falls into class III (mild mutation severity) with respect to its hematological and biochemical characteristics.
Using this model, it has been shown that mild G6PD deficiency (15% activity of WT) induces a pronounced decrease in RBC deformability and worsens erythrocyte dysfunction during sepsis. RBC dysfunction aggravates organ dysfunction and microcirculatory disturbances and may also contribute to the modulation of macrophage responses during severe infections in G6PD-deficient animals [43,44].
A significant number of studies have unveiled the roles of G6PD in various aspects of physiology other than erythrocytic pathophysiology, such as diabetes, cardiovascular disease, and neurodegeneration [45]. The association between G6PD deficiency and the development of diabetes has been supported by epidemiological studies conducted in different research groups and populations [46,47,48]. An increased risk for diabetes, and also of diabetic complications such as proliferative retinopathy [49], has been reported in G6PD-deficient subjects [50,51].
In preclinical studies, it has been shown that the liver and pancreas of diabetic rats show a reduction in G6PD activity [52]. Pancreatic islets from G6PD mutant mice are smaller than those of WT mice [53], which suggests that G6PD plays important roles in the survival and functions of pancreatic cells. Accordingly, it has been reported that mutations in the G6PD gene and the consequent drop in G6PD activity are sufficient to cause changes similar to those seen in diabetic mice [54]. Using the opposite methodological approach, we found that G6PD transgenic (Tg) mice moderately overexpressing the enzyme (2–4-fold overexpression) were more insulin sensitive and glucose tolerant than WT controls [9]. These results are in accordance with previous mouse overexpression models of NADPH-dependent ROS-detoxifying enzymes. For instance, Prx3-Tg and Prx4-Tg mice were shown to have better insulin sensitivity and glucose tolerance compared to WT mice [55]. Although the molecular mechanism underlying the association between G6PD deficiency and diabetes is not completely understood, current evidence suggests that G6PD deficiency may be a risk factor for diabetes, with higher odds among men compared to women [46,47].
The role of ROS as physiological signals as well as pathological stresses has been demonstrated repeatedly in the cardiovascular system [56,57,58,59]. However, the relation between G6PD deficiency and risk for cardiovascular disease and subsequent outcomes is unclear. The existing data indicate a complex interplay in which the adverse effects of G6PD deficiency may outweigh the potential protective effects in the context of cardiac stress [34,60,61,62].
The risk of redox-mediated damage to brain cells in G6PD deficiency has also been studied [63]. G6PD is an important enzyme in the protection against age-associated ROS neurodegenerative effects, and more specifically in the age-associated increase in oxidative DNA damage in the brain [63]. Recently, brain damage associated with ROS production in G6PD-deficient animals was also found to have functional consequences. Old G6PD-deficient male mice exhibited synaptic dysfunction in their hippocampal slices while young and old G6PD-deficient females exhibited deficits in executive functions and social dominance [64].
Taken together, these results suggest that there are broad health implications of G6PD deficiencies. Among the potential outcomes related to G6PD loss of function, birth defects, heart disease, diabetes, and neurodegeneration are highlighted.

3. G6PD and Cell Growth

The modulation of cell survival and cell growth relies on intracellular redox regulation [65]. As mentioned in the previous sections of this manuscript, NADPH—the principal intracellular reductant—is a critical modulator of redox potential. In 1999, Dr. Stanton and coworkers found that G6PD plays an important role in cell death by regulating intracellular redox levels [66]. The inhibition of G6PD by both dehydroepiandrosterone (DHEA) and 6-aminonicotinamide (6-ANAD) augmented cell death triggered by serum deprivation and oxidative stress, while the overexpression of G6PD in a cell line conferred resistance to H2O2-induced cell death. Previously, in G6PD-deficient cell lines, it was reported that these cells had decreased cloning efficiencies and growth rates and were highly sensitive to ROS when compared to cells expressing endogenous levels of the enzyme [67]. Consistent with these results, an association between the stimulation of cell growth in different tissues and increased PPP activity has also been reported [68]. Kidney hypertrophy due to unilateral nephrectomy is associated with increased G6PD activity [69], while the growth of rat liver cells stimulated by growth hormone is also associated with an increase in G6PD activity [70].
In experiments to determine if the increased G6PD activity per se is an essential component of normal cell growth, it was found that G6PD activity was directly correlated with cell growth, that the inhibition of G6PD activity prevented growth, and that the overexpression of G6PD alone stimulated [3H]-thymidine incorporation [65].
As previously mentioned, cancers and cultured tumor cells exhibit large increases in G6PD activity [71]. To test the potential tumorigenic risk of G6PD overexpression, we crossed G6PD-Tg mice with several genetically modified tumor-prone animals, including ATM-KO (that develop T-cell lymphomas), Eμ-myc (that develop B-cell lymphomas), p53-KO (that develop T-cell lymphomas and sarcomas), and MMTV-PyMT (that develop mammary tumors) [9,10]. In all of these combinations, mice carrying a G6PD-Tg allele showed the same tumor latency and incidence as the WT mice. These data indicate that a moderate and regulated increase in NADPH levels or G6PD expression and activity does not result in increased tumor incidence [9]. On the contrary, G6PD-Tg mice showed improved lifespan and health parameters as they grew old: (i) the transgenic animals were more insulin sensitive and glucose tolerant; (ii) old G6PD-Tg mice tended to gain less weight and exhibited improved motor coordination; and (iii) G6PD-Tg females showed a ~14% increase in medium lifespan. At the molecular level, we found a reduction in age-associated lipid peroxidation and DNA oxidation in different tissues [9]. We related the decrease in age-associated oxidative damage to macromolecules, a result of the modulation of cellular NADPH levels, to the improvements in health and lifespan in the G6PD-Tg animals.
The treatment of both animals and humans with antioxidant vitamins and other supplements [9], specially at high doses, has not been shown to increase lifespan and has failed to protect against age-induced pathologies. Studies on the biological roles of ROS have uncovered the beneficial signaling functions of these highly reactive molecules to explain these contradictory results [10]. The overexpression of antioxidant enzymes vs. the administration of exogenous antioxidants are very different approaches to test the importance of redox balance both in aging and age-associated diseases with very different outcomes [10,55].

4. G6PD in the Regeneration of Skeletal Muscle after Damage

The hexose monophosphate shunt is considered an almost negligible pathway in normal muscle. For this reason, the function of G6PD in skeletal muscle has been poorly investigated.
In vitro studies have shown that, under normal conditions, glucose breakdown takes place via both the Embden–Meyerhof pathway and the PPP in the liver, pancreas, arterial wall, kidney, spleen, and adrenals. However, in the central nervous system and cardiac and striated muscle, it is metabolized mainly via the glycolytic route [72]. In addition, several conditions increase the activity of the PPP in skeletal muscle: (i) embryogenesis [73]; (ii) denervation; (iii) ischemia; (iv) hypertrophy; (v) the injection of myonecrotic agents with local degeneration effects [74,75]; and (vi) physical exercise [32].
The injection of myonecrotic agents (bupivacaine, Marcaine, or cardiotoxin) induces a rapid (8 h) and dramatic (6–9-fold) increase in the activities of G6PD and 6PGD during regeneration after muscle destruction. By using histological techniques [76,77], it has been shown that G6PD is localized within muscle cells in regenerating muscle; thus, the enhanced enzyme activity resides in the muscle fibers themselves for at least the first 6–8 h after Marcaine injection. After that time, phagocytic cells contribute to the increase in enzyme activity [74]. The enhanced activities of G6PD and 6PGD likely reflect accelerated glucose utilization for the production of nucleic acids and lipids [75,78,79,80]. In this regard, increased quantities of RNA have been noted in a number of studies on muscle regeneration [81,82,83]. The enhancement of the PPP is important for anabolic processes in the initial stages of skeletal muscle regeneration; however, the role of G6PD in skeletal muscle goes beyond biosynthetic processes. In 2016, Febbraio and coworkers found that one mechanism linking an altered cellular redox state to insulin resistance is NOS [84]. S G6PD activity in skeletal muscle is linked to nitric oxide (NO) bioavailability; thus, an impairment in the NOS isozyme (nNOSμ) in insulin resistant states in rodents and humans leads to an increase in G6PD activity [84].
The consequences of G6PD deficiency in skeletal muscle have been studied in clinical cases of rhabdomyolysis [85] and myopathies [86]. In fact, a statistically significant relationship has been found with regard to the activity of G6PD between RBCs and muscle in humans [87].

5. Positive Regulators of G6PD Activity in Skeletal Muscle—Role of Exercise

As previously mentioned, G6PD overexpression in Drosophila Melanogaster and mice protects against metabolic stress [9,88] and oxidative damage [9]. Very recently, we found that it also delays the onset of frailty by protecting against muscle damage [32].
As shown in Table 2, G6PD can be regulated by pharmacological, nutritional, and physiological interventions, such as physical exercise [68].
G6PD activity has been studied in both skeletal muscle and erythrocytes after one bout of exhaustive exercise. Surprisingly, contradictory results were found in the literature.
G6PD activity in erythrocytes is reduced in humans after one bout of high intensity exercise (~40%), likely due to ROS generation [89]. Accordingly, supplementation with L-cysteine for a week (0.5 g/24 h) [89] or with α-Tocopherol for a month (200 mg/24 h) [90] leads to the maintenance of the enzyme activity. These results have also been verified in long distance runners [91] and soccer players [91,92]. To the contrary, the highest relative increase in enzyme activities, both for mitochondrial and extramitochondrial enzymes, after exhaustive swimming in rat skeletal muscle was shown for G6PD and 6PGD, which increased by 115% and 40%, respectively, 1 and 3 days after an acute bout of exercise [93]. Similarly, an increase in muscle G6PD activity of ~100–350% was observed after a downhill running protocol in rats [94], suggesting that the activation of the PPP occurs in skeletal muscle to provide substrates for muscle repair.
In one study, the changes in G6PD expression in skeletal muscle associated with different exercise intensities were investigated [95]. Based on the lactate threshold, it was shown that low-intensity aerobic treadmill running induced higher increases in the mRNA levels of G6PD in rat soleus muscle when compared to high-intensity anaerobic running [95]. Exercise duration is also a critical factor in the activation of G6PD in skeletal muscle. A significant linear correlation has been reported between the duration of downhill running (0, 30, or 90 min) and G6PD activity in different muscle groups in untrained rats [94].
G6PD activity also shows a susceptibility to exercise training in skeletal muscle. The exercise-induced elevation in muscle G6PD activity after one bout of downhill running was shown to be significantly reduced with only 5 days of either level or downhill training in rats [94]. This is the reason why changes in skeletal muscle G6PD activity have been widely used to study the “repeated bout effect”, which refers to an adaptation whereby a single bout of eccentric exercise protects against muscle damage from subsequent eccentric bouts [94,96].
The activity of G6PD and 6PDG increases pronouncedly by a factor of three in the gastrocnemius muscle after 5 days of repeated ischemia [97]. A similar increase in the activities of PPP enzymes has also been found in the heart after myocardial infarction [98]. Again, these results suggest that the increase in G6PD activity is important for repair purposes, as it increases the production of NADPH and the pentoses necessary for biosynthetic processes.
The PPP has been proven to be a fundamental metabolic pathway that allows for rapid and robust hypertrophic growth in muscle cells in response to mechanical overload [99]. For example, the denervation of one half of the diaphragm was shown to induce transient hypertrophy in the muscle on the other side [100]. In this model, the activities of G6PD and 6PDG increased immediately after denervation, reaching a maximum after 3 days [100]. More recently, the importance of G6PD in the regulation of skeletal muscle metabolism during hypertrophy was highlighted in a study analyzing gene expression from a transcriptomic microarray of specific metabolic pathways in mechanically overloaded plantaris muscle-induced hypertrophy [99]. A robust increase in G6PD mRNA expression was found in the overloaded muscle throughout the whole analyzed time course (1, 3, 5, and 7 days), consistent with an increase in NADPH levels to support nucleotide biosynthesis and to boost the muscle antioxidant defense [99]. It was also shown that the abundance of the G6PD protein significantly increased (~140%) in response to 5 days of mechanical overload in muscle [101].
The “MyoMouse” is a conditional mice model that inducibly expresses an activated form of Akt1 specifically in skeletal muscle [102]. The induction of the Akt1 signaling pathway leads to selective hypertrophy in type II fibers and an increase in muscle strength [102,103]. A combination of metabolomic and transcriptomic analyses has shown that Akt1-induced muscle growth is accompanied by a robust upregulation of biosynthetic metabolic pathways, such as the PPP, and the downregulation of catabolic pathways, such as glycolysis and oxidative phosphorylation [103]. Specifically, the “MyoMouse” shows a 3.5-fold increase in G6PD and a 2.3-fold increase in 6PDG in the hypertrophied muscles. Consistent with an increase in metabolite flux through the PPP, a 1.8-fold accumulation of R5P, an increase in total RNA, and an increase in purines and pyrimidine metabolites, including 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) and xanthosine, were also reported in the muscle tissue [103].
A potential limitation of the studies discussed above is the fact that all the analyses encompassed the whole muscle and could not distinguish the contribution of non-muscle cell types to the observed changes in G6PD expression, protein levels, or activity.
It has been suggested that the accumulation of cells in the connective tissue rather than changes in the activity within muscle fibers may explain the increase in the activity of the PPP enzymes in skeletal muscle following injury [104]. Macrophage, neutrophil, and mast cell levels are elevated after exercise and in mechanically overloaded muscles [104,105], which may influence the reported activity of G6PD.
The development of in vitro studies in C2C12 myoblasts has helped to overcome this concern. The overexpression of G6PD in C2C12 G6PD cells promotes their proliferation and significantly increases the percentage of EdU-positive cells. To the contrary, G6PD inhibition in myoblasts induces cell cycle arrest in the G0/G1 phase and suppresses muscle cell proliferation [106]. Moreover, the high-frequency electrical stimulation of C2C12 myotubes—mimicking muscle contraction—increases the expression of genes encoding the enzymes of the PPP [107].
The results reported by McCarthy’s research team also provide evidence that most of the observed changes in gene expression reflected in skeletal muscle occur within muscle fibers themselves [108]. These authors showed that myofibers are overwhelmingly the most transcriptionally active cell type in skeletal muscle at rest and during muscle hypertrophy. Approximately 90% of nascent RNA is associated with myonuclei during a mechanical overload induced by synergist ablation.
The results reported by Shimokawa and coworkers also support the idea that the exercise-induced increase G6PD activity is muscle specific and independent of inflammatory cells [95]. They found an increase in the mRNA expression of G6PD with aerobic exercise in rat soleus muscle, while this increment was absent in the animals following an anaerobic protocol. Macrophage invasion and injured and regenerating fibers were observed after anaerobic exercise, while neither of these signs of damage were found after the aerobic protocol [95]
Finally, testosterone [109] and growth hormone-induced muscle fiber hypertrophy in aging [110], two treatments that are independent of inflammatory signals, have been associated with an increase in G6PD protein levels in skeletal muscle.
These data suggest that muscle adaptations to exercise training or to mechanical overload require enhanced redox metabolism via the production of NADPH through the PPP and an increase in the expression of G6PD [99,111].
Physical exercise acutely increases ROS generation; however, if practiced regularly, it induces positive adaptations in mitochondrial density [112,113,114,115] and antioxidant defenses, including increased G6PD enzymatic activity [115,116,117]. Growing evidence suggests that physical training upregulates the level of antioxidant enzymes in the tissues actively involved in exercise [118,119]. For instance, eccentric exercise training in mice lasting 5 days was shown to be enough to induce an increase in G6PD mRNA levels and activity in skeletal muscle in young animals [32], similar to that found in a transgenic mouse model moderately overexpressing G6PD [9].
The age-associated loss in muscle mass and strength (i.e., sarcopenia) leads to a decrease in G6PD activity and protein content in skeletal muscle [120]. Whether the well-known positive effects of exercise training in old individuals are mediated through an increase in G6PD activity should be further studied in depth. The results published to date are contradictory and do not allow definitive conclusions to be drawn [121,122,123,124].
Finally, the question of whether exercise training is a safe and useful intervention in G6PD-deficient patients is something that has been an object of debate. G6PD-deficient individuals, as previously mentioned, are less protected against oxidative stress and could be predisposed to oxidative damage when they perform high-intensity physical training [125]. However, several studies have shown that exercise intensity does not cause oxidative stress or hemolysis above those levels expected in people without G6PD deficiency [126,127,128,129]. Therefore, despite the limited published studies, it seems that G6PD-deficient patients can safely participate in physical exercise programs with different intensities and durations.
Table 2. Cellular signals regulating G6PD and the PPP.
Table 2. Cellular signals regulating G6PD and the PPP.
Positive RegulatorsNegative Regulators
Acetylation [130]5′ adenosine monophosphate-activated protein kinase (AMPK) [131]
G6PD activator AG1 [132]Aldosterone [133]
AKT [134]Angiotensin II [120]
ATM serine/threonine kinase (ATM) [135]Arachidonic acid [136]
Benfotiamine (vitamin B1 analog) [110,137]Cyclic adenosine monophosphate (cAMP) [138]
Proto-oncogene tyrosine-protein kinase Src (c-Src) [25]cAMP-dependent protein kinase A [138]
cGMP-dependent protein kinase G [139]cAMP response element modulator (CREM) [133]
Cyclin D3-CDK6 [140]Dehydroepiandrosterone (DHEA) [141]
Epidermal growth factor (EGF) [142]miR-122 and miR-1 [143]
Estrogens [110]p38 mitogen-activated protein kinase [136]
Exercise [32]p53 [144]
Glycosylation [145]Phosphatase and tensin homolog (PTEN) [146]
Growth hormone [110]TP53 [144]
Hepatocyte growth factor (HGF) [147]Tumor necrosis factor-α (TNFα) [68]
Heat shock protein 27 (Hsp27) [148]
Hypoxia inducible factor (HIF) [149]
Inhibitor of DNA binding 1 (ID1) [150]
Insulin [151]
Mammalian target of rapamycin (mTOR) [152]
Nuclear-factor-E2-related factor (Nrf2) [153]
Ribosomal protein S6 kinase beta-1 (p70S6K) [110]
Serine/threonine-protein kinase PAK 4 (PAK4) [154]
Protein disulfide isomerase family A, member 3 pseudogene (PDIA3P) [155]
Phosphatidylinositol-3-kinase (PI-3K) [134]
Phospholipase C [110]
Phospholipase C-γ [156]
Platelet-derived growth factor (PDGF) [156]
Polo-like kinase 1 (PLK-1) [157]
Ras-GTPase [68]
S6 kinase [158]
Snail [159]
Sterol-responsive element bindingprotein (SREBP) 1 [68]
Stobadine [160]
TAp73 [161]
Testosterone [110]
Transforming growth factor beta 1 (TGF-β1) [162]
TP53-induced glycolysis and apoptosis regulator (TIGAR) [163]
Vascular endothelial cell growth factor (VEGF) [25]
Vitamin D [164]
Vitamin E [160]

Author Contributions

J.V., M.C.G.-C. and G.O.-G. contributed to design the structure of this manuscript. E.G.-D., A.C., G.O.-G., A.V.-A. and M.C.G.-C. write the manuscript first draft. Figures and tables were created by E.G.-D. and G.O.-G.; J.D.-F., J.V. and M.C.G.-C. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Instituto de Salud Carlos III CB16/10/00435 (CIBERFES); (PID2019-110906RB-I00/AEI/10.13039/501100011033) from the Spanish Ministry of Innovation and Science; FGCSIC/PSLINTERREG/FEDER; PROMETEO/2019/097 de “Consellería, de Sanitat de la Generalitat Valenciana”; EU Funded H2020- DIABFRAIL-LATAM (Ref: 825546); European Joint Programming Initiative “A Healthy Diet for a Healthy Life” (JPI HDHL) and the ERA-NET Cofund ERA-HDHL (GA N 696295 of the EU Horizon 2020 Research and Innovation Programme). Part of the equipment employed in this work was funded by Generalitat Valenciana and co-financed with ERDF funds (OP ERDF of Comunitat Valenciana 2014-2020). We would also like to acknowledge the support from the Ramón Areces Foundation and Soria Melguizo Foundation. In addition, E.G.-D. was a recipient of a predoctoral grant financed by the Spanish Ministry (FPU18/05350).

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. Couri, D.; Racker, E. The oxidative pentose phosphate cycle. V. Complete oxidation of glucose 6-phosphate in a reconstructed system of the oxidative pentose phosphate cycle. Arch. Biochem. Biophys. 1959, 83, 195–205. [Google Scholar] [CrossRef]
  2. Horecker, B.L. The pentose phosphate pathway. J. Biol. Chem. 2002, 277, 47965–47971. [Google Scholar] [CrossRef] [PubMed]
  3. Dickens, F.; Williamson, D.H. Pentose phosphate isomerase and epimerase from animal tissues. Biochem. J. 1956, 64, 567–578. [Google Scholar] [CrossRef] [PubMed]
  4. Sulek, K. Nobel prize in 1931 for Otto Warburg for discovery of the respiratory enzyme. Wiad. Lek. 1968, 21, 329. [Google Scholar]
  5. Stincone, A.; Prigione, A.; Cramer, T.; Wamelink, M.M.; Campbell, K.; Cheung, E.; Olin-Sandoval, V.; Grüning, N.M.; Krüger, A.; Tauqeer Alam, M.; et al. The return of metabolism: Biochemistry and physiology of the pentose phosphate pathway. Biol. Rev. Camb. Philos. Soc. 2015, 90, 927–963. [Google Scholar] [CrossRef]
  6. Baquer, N.Z.; Hothersall, J.S.; McLean, P. Function and regulation of the pentose phosphate pathway in brain. Curr. Top. Cell. Regul. 1988, 29, 265–289. [Google Scholar]
  7. Barcia-Vieitez, R.; Ramos-Martínez, J.I. The regulation of the oxidative phase of the pentose phosphate pathway: New answers to old problems. IUBMB Life 2014, 66, 775–779. [Google Scholar] [CrossRef]
  8. Ghergurovich, J.M.; García-Cañaveras, J.C.; Wang, J.; Schmidt, E.; Zhang, Z.; TeSlaa, T.; Patel, H.; Chen, L.; Britt, E.C.; Piqueras-Nebot, M.; et al. A small molecule G6PD inhibitor reveals immune dependence on pentose phosphate pathway. Nat. Chem. Biol. 2020, 16, 731–739. [Google Scholar] [CrossRef]
  9. Nóbrega-Pereira, S.; Fernandez-Marcos, P.J.; Brioche, T.; Gomez-Cabrera, M.C.; Salvador-Pascual, A.; Flores, J.M.; Viña, J.; Serrano, M. G6PD protects from oxidative damage and improves healthspan in mice. Nat. Commun. 2016, 7, 10894. [Google Scholar] [CrossRef]
  10. Fernandez-Marcos, P.J.; Nobrega-Pereira, S. NADPH: New oxygen for the ROS theory of aging. Oncotarget 2016, 7, 50814–50815. [Google Scholar] [CrossRef]
  11. Panday, A.; Sahoo, M.K.; Osorio, D.; Batra, S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cell. Mol. Immunol. 2015, 12, 5–23. [Google Scholar] [CrossRef]
  12. Brown, D.I.; Griendling, K.K. Nox proteins in signal transduction. Free Radic. Biol. Med. 2009, 47, 1239–1253. [Google Scholar] [CrossRef] [Green Version]
  13. Ferreira, L.F.; Laitano, O. Regulation of NADPH oxidases in skeletal muscle. Free Radic. Biol. Med. 2016, 98, 18–28. [Google Scholar] [CrossRef]
  14. Berg, J.M.; Tymoczko, J.L.; Gatto, G.J., Jr.; Stryer, L.; Held, A.T.; Maxam, G.T.; Seidler, L.T.; Hacker, B.R.T.; Jarosch, B.T. Biochemistry, 8th ed.; Springer: Berlin/Heidelberg, Germany, 2017. [Google Scholar]
  15. Cho, E.S.; Cha, Y.H.; Kim, H.S.; Kim, N.H.; Yook, J.I. The Pentose Phosphate Pathway as a Potential Target for Cancer Therapy. Biomol. Ther. 2018, 26, 29–38. [Google Scholar] [CrossRef]
  16. Eggleston, L.V.; Krebs, H.A. Regulation of the pentose phosphate cycle. Biochem. J. 1974, 138, 425–435. [Google Scholar] [CrossRef]
  17. Salati, L.M.; Amir-Ahmady, B. Dietary regulation of expression of glucose-6-phosphate dehydrogenase. Annu. Rev. Nutr. 2001, 21, 121–140. [Google Scholar] [CrossRef]
  18. Jiang, A.; Guo, H.; Jiang, X.; Tao, J.; Wu, W.; Liu, H. G6PD Deficiency is Crucial for Insulin Signaling Activation in Skeletal Muscle. Int. J. Mol. Sci. 2022, 23, 7425. [Google Scholar] [CrossRef]
  19. Horton, J.D. Sterol regulatory element-binding proteins: Transcriptional activators of lipid synthesis. Biochem. Soc. Trans. 2002, 30, 1091–1095. [Google Scholar] [CrossRef]
  20. Salati, L.M.; Szeszel-Fedorowicz, W.; Tao, H.; Gibson, M.A.; Amir-Ahmady, B.; Stabile, L.P.; Hodge, D.L. Nutritional regulation of mRNA processing. J. Nutr. 2004, 134, 2437S–2443S. [Google Scholar] [CrossRef]
  21. Zhang, H.S.; Wang, S.Q. Nrf2 is involved in the effect of tanshinone IIA on intracellular redox status in human aortic smooth muscle cells. Biochem. Pharmacol. 2007, 73, 1358–1366. [Google Scholar] [CrossRef]
  22. Holten, D.; Procsal, D.; Chang, H.L. Regulation of pentose phosphate pathway dehydrogenases by NADP+/NADPH ratios. Biochem. Biophys. Res. Commun. 1976, 68, 436–441. [Google Scholar] [CrossRef]
  23. Garcia, A.A.; Mathews, I.I.; Horikoshi, N.; Matsui, T.; Kaur, M.; Wakatsuki, S.; Mochly-Rosen, D. Stabilization of glucose-6-phosphate dehydrogenase oligomers enhances catalytic activity and stability of clinical variants. J. Biol. Chem. 2022, 298, 101610. [Google Scholar] [CrossRef]
  24. Veech, R.L.; Eggleston, L.V.; Krebs, H.A. The redox state of free nicotinamide-adenine dinucleotide phosphate in the cytoplasm of rat liver. Biochem. J. 1969, 115, 609–619. [Google Scholar] [CrossRef]
  25. Pan, S.; World, C.J.; Kovacs, C.J.; Berk, B.C. Glucose 6-phosphate dehydrogenase is regulated through c-Src-mediated tyrosine phosphorylation in endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 895–901. [Google Scholar] [CrossRef]
  26. Marks, P.A.; Gross, R.T. Erythrocyte glucose-6-phosphate dehydrogenase deficiency: Evidence of differences between Negroes and Caucasians with respect to this genetically determined trait. J. Clin. Investig. 1959, 38, 2253–2262. [Google Scholar] [CrossRef]
  27. Frank, J.E. Diagnosis and management of G6PD deficiency. Am. Fam. Physician 2005, 72, 1277–1282. [Google Scholar]
  28. Cappellini, M.D.; Fiorelli, G. Glucose-6-phosphate dehydrogenase deficiency. Lancet 2008, 371, 64–74. [Google Scholar] [CrossRef]
  29. Ruwende, C.; Khoo, S.C.; Snow, R.W.; Yates, S.N.; Kwiatkowski, D.; Gupta, S.; Warn, P.; Allsopp, C.E.; Gilbert, S.C.; Peschu, N. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 1995, 376, 246–249. [Google Scholar] [CrossRef]
  30. Cappadoro, M.; Giribaldi, G.; O’Brien, E.; Turrini, F.; Mannu, F.; Ulliers, D.; Simula, G.; Luzzatto, L.; Arese, P. Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood 1998, 92, 2527–2534. [Google Scholar] [CrossRef]
  31. Kletzien, R.F.; Harris, P.K.; Foellmi, L.A. Glucose-6-phosphate dehydrogenase: A “housekeeping” enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress. FASEB J. 1994, 8, 174–181. [Google Scholar] [CrossRef]
  32. Arc-Chagnaud, C.; Salvador-Pascual, A.; Garcia-Dominguez, E.; Olaso-Gonzalez, G.; Correas, A.G.; Serna, E.; Brioche, T.; Chopard, A.; Fernandez-Marcos, P.J.; Serrano, M.; et al. Glucose 6-P dehydrogenase delays the onset of frailty by protecting against muscle damage. J. Cachexia Sarcopenia Muscle 2021, 12, 1879–1896. [Google Scholar] [CrossRef] [PubMed]
  33. Luzzatto, L.; Arese, P. Favism and Glucose-6-Phosphate Dehydrogenase Deficiency. N. Engl. J. Med. 2018, 378, 1068–1069. [Google Scholar] [CrossRef] [PubMed]
  34. Hecker, P.A.; Lionetti, V.; Ribeiro, R.F.; Rastogi, S.; Brown, B.H.; O’Connell, K.A.; Cox, J.W.; Shekar, K.C.; Gamble, D.M.; Sabbah, H.N.; et al. Glucose 6-phosphate dehydrogenase deficiency increases redox stress and moderately accelerates the development of heart failure. Circ. Heart Fail. 2013, 6, 118–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Shalev, O.; Wollner, A.; Menczel, J. Diabetic ketoacidosis does not precipitate haemolysis in patients with the Mediterranean variant of glucose-6-phosphate dehydrogenase deficiency. Br. Med. J. 1984, 288, 179–180. [Google Scholar] [CrossRef]
  36. Lee, D.H.; Warkentin, T.E.; Neame, P.B.; Ali, M.A. Acute hemolytic anemia precipitated by myocardial infarction and pericardial tamponade in G6PD deficiency. Am. J. Hematol. 1996, 51, 174–175. [Google Scholar] [CrossRef]
  37. Battistuzzi, G.; D’Urso, M.; Toniolo, D.; Persico, G.M.; Luzzatto, L. Tissue-specific levels of human glucose-6-phosphate dehydrogenase correlate with methylation of specific sites at the 3′ end of the gene. Proc. Natl. Acad. Sci. USA 1985, 82, 1465–1469. [Google Scholar] [CrossRef]
  38. Nagashio, R.; Oikawa, S.; Yanagita, K.; Hagiuda, D.; Kuchitsu, Y.; Igawa, S.; Naoki, K.; Satoh, Y.; Ichinoe, M.; Murakumo, Y.; et al. Prognostic significance of G6PD expression and localization in lung adenocarcinoma. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 38–46. [Google Scholar] [CrossRef]
  39. Vives Corrons, J.L.; Feliu, E.; Pujades, M.A.; Cardellach, F.; Rozman, C.; Carreras, A.; Jou, J.M.; Vallespí, M.T.; Zuazu, F.J. Severe-glucose-6-phosphate dehydrogenase (G6PD) deficiency associated with chronic hemolytic anemia, granulocyte dysfunction, and increased susceptibility to infections: Description of a new molecular variant (G6PD Barcelona). Blood 1982, 59, 428–434. [Google Scholar] [CrossRef]
  40. Longo, L.; Vanegas, O.C.; Patel, M.; Rosti, V.; Li, H.; Waka, J.; Merghoub, T.; Pandolfi, P.P.; Notaro, R.; Manova, K.; et al. Maternally transmitted severe glucose 6-phosphate dehydrogenase deficiency is an embryonic lethal. EMBO J. 2002, 21, 4229–4239. [Google Scholar] [CrossRef]
  41. Nicol, C.J.; Zielenski, J.; Tsui, L.C.; Wells, P.G. An embryoprotective role for glucose-6-phosphate dehydrogenase in developmental oxidative stress and chemical teratogenesis. FASEB J. 2000, 14, 111–127. [Google Scholar] [CrossRef]
  42. Pretsch, W.; Charles, D.J.; Merkle, S. X-linked glucose-6-phosphate dehydrogenase deficiency in Mus musculus. Biochem. Genet. 1988, 26, 89–103. [Google Scholar] [CrossRef]
  43. Sanders, S.; Smith, D.P.; Thomas, G.A.; Williams, E.D. A glucose-6-phosphate dehydrogenase (G6PD) splice site consensus sequence mutation associated with G6PD enzyme deficiency. Mutat. Res. 1997, 374, 79–87. [Google Scholar] [CrossRef]
  44. Spolarics, Z.; Condon, M.R.; Siddiqi, M.; Machiedo, G.W.; Deitch, E.A. Red blood cell dysfunction in septic glucose-6-phosphate dehydrogenase-deficient mice. Am. J. Physiol. Heart Circ. Physiol. 2004, 286, H2118–H2126. [Google Scholar] [CrossRef]
  45. Ho, H.Y.; Cheng, M.L.; Chiu, D.T. Glucose-6-phosphate dehydrogenase—Beyond the realm of red cell biology. Free Radic. Res. 2014, 48, 1028–1048. [Google Scholar] [CrossRef]
  46. Wan, G.H.; Tsai, S.C.; Chiu, D.T. Decreased blood activity of glucose-6-phosphate dehydrogenase associates with increased risk for diabetes mellitus. Endocrine 2002, 19, 191–195. [Google Scholar] [CrossRef]
  47. Lai, Y.K.; Lai, N.M.; Lee, S.W. Glucose-6-phosphate dehydrogenase deficiency and risk of diabetes: A systematic review and meta-analysis. Ann. Hematol. 2017, 96, 839–845. [Google Scholar] [CrossRef]
  48. Heymann, A.D.; Cohen, Y.; Chodick, G. Glucose-6-phosphate dehydrogenase deficiency and type 2 diabetes. Diabetes Care 2012, 35, e58. [Google Scholar] [CrossRef]
  49. Cappai, G.; Songini, M.; Doria, A.; Cavallerano, J.D.; Lorenzi, M. Increased prevalence of proliferative retinopathy in patients with type 1 diabetes who are deficient in glucose-6-phosphate dehydrogenase. Diabetologia 2011, 54, 1539–1542. [Google Scholar] [CrossRef]
  50. Niazi, G.A. Glucose-6-phosphate dehydrogenase deficiency and diabetes mellitus. Int. J. Hematol. 1991, 54, 295–298. [Google Scholar]
  51. Carette, C.; Dubois-Laforgue, D.; Gautier, J.F.; Timsit, J. Diabetes mellitus and glucose-6-phosphate dehydrogenase deficiency: From one crisis to another. Diabetes Metab. 2011, 37, 79–82. [Google Scholar] [CrossRef]
  52. Díaz-Flores, M.; Ibáñez-Hernández, M.A.; Galván, R.E.; Gutiérrez, M.; Durán-Reyes, G.; Medina-Navarro, R.; Pascoe-Lira, D.; Ortega-Camarillo, C.; Vilar-Rojas, C.; Cruz, M.; et al. Glucose-6-phosphate dehydrogenase activity and NADPH/NADP+ ratio in liver and pancreas are dependent on the severity of hyperglycemia in rat. Life Sci. 2006, 78, 2601–2607. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Z.; Liew, C.W.; Handy, D.E.; Zhang, Y.; Leopold, J.A.; Hu, J.; Guo, L.; Kulkarni, R.N.; Loscalzo, J.; Stanton, R.C. High glucose inhibits glucose-6-phosphate dehydrogenase, leading to increased oxidative stress and beta-cell apoptosis. FASEB J. 2010, 24, 1497–1505. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, Y.; Zhang, Z.; Hu, J.; Stillman, I.E.; Leopold, J.A.; Handy, D.E.; Loscalzo, J.; Stanton, R.C. Glucose-6-phosphate dehydrogenase-deficient mice have increased renal oxidative stress and increased albuminuria. FASEB J. 2010, 24, 609–616. [Google Scholar] [CrossRef] [PubMed]
  55. Hamilton, R.T.; Walsh, M.E.; Van Remmen, H. Mouse Models of Oxidative Stress Indicate a Role for Modulating Healthy Aging. J. Clin. Exp. Pathol. 2012, 4, 5. [Google Scholar] [CrossRef] [Green Version]
  56. Brandes, R.P.; Weissmann, N.; Schroder, K. NADPH oxidases in cardiovascular disease. Free. Radic. Biol. Med. 2010, 49, 687–706. [Google Scholar] [CrossRef]
  57. Murphy, E.; Steenbergen, C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol. Rev. 2008, 88, 581–609. [Google Scholar] [CrossRef]
  58. Penna, C.; Mancardi, D.; Rastaldo, R.; Pagliaro, P. Cardioprotection: A radical view Free radicals in pre and postconditioning. Biochim. Biophys. Acta 2009, 1787, 781–793. [Google Scholar] [CrossRef]
  59. Carretero, A.; Gomez-Cabrera, M.C.; Rios-Navarro, C.; Salvador-Pascual, A.; Bodi, V.; Viña, J. Early reductive stress and late onset overexpression of antioxidant enzymes in experimental myocardial infarction. Free Radic. Res. 2020, 54, 173–184. [Google Scholar] [CrossRef]
  60. Hecker, P.A.; Leopold, J.A.; Gupte, S.A.; Recchia, F.A.; Stanley, W.C. Impact of glucose-6-phosphate dehydrogenase deficiency on the pathophysiology of cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H491–H500. [Google Scholar] [CrossRef]
  61. Matsui, R.; Xu, S.; Maitland, K.A.; Mastroianni, R.; Leopold, J.A.; Handy, D.E.; Loscalzo, J.; Cohen, R.A. Glucose-6-phosphate dehydrogenase deficiency decreases vascular superoxide and atherosclerotic lesions in apolipoprotein E(−/−) mice. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 910–916. [Google Scholar] [CrossRef]
  62. Cocco, P.; Todde, P.; Fornera, S.; Manca, M.B.; Manca, P.; Sias, A.R. Mortality in a cohort of men expressing the glucose-6-phosphate dehydrogenase deficiency. Blood 1998, 91, 706–709. [Google Scholar] [CrossRef]
  63. Jeng, W.; Loniewska, M.M.; Wells, P.G. Brain glucose-6-phosphate dehydrogenase protects against endogenous oxidative DNA damage and neurodegeneration in aged mice. ACS Chem. Neurosci. 2013, 4, 1123–1132. [Google Scholar] [CrossRef]
  64. Loniewska, M.M.; Gupta, A.; Bhatia, S.; MacKay-Clackett, I.; Jia, Z.; Wells, P.G. DNA damage and synaptic and behavioural disorders in glucose-6-phosphate dehydrogenase-deficient mice. Redox Biol. 2020, 28, 101332. [Google Scholar] [CrossRef]
  65. Tian, W.N.; Braunstein, L.D.; Pang, J.; Stuhlmeier, K.M.; Xi, Q.C.; Tian, X.; Stanton, R.C. Importance of glucose-6-phosphate dehydrogenase activity for cell growth. J. Biol. Chem. 1998, 273, 10609–10617. [Google Scholar] [CrossRef] [Green Version]
  66. Tian, W.N.; Braunstein, L.D.; Apse, K.; Pang, J.; Rose, M.; Tian, X.; Stanton, R.C. Importance of glucose-6-phosphate dehydrogenase activity in cell death. Am. J. Physiol. 1999, 276, C1121–C1131. [Google Scholar] [CrossRef]
  67. Pandolfi, P.P.; Sonati, F.; Rivi, R.; Mason, P.; Grosveld, F.; Luzzatto, L. Targeted disruption of the housekeeping gene encoding glucose 6-phosphate dehydrogenase (G6PD): G6PD is dispensable for pentose synthesis but essential for defense against oxidative stress. EMBO J. 1995, 14, 5209–5215. [Google Scholar] [CrossRef]
  68. Stanton, R.C. Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life 2012, 64, 362–369. [Google Scholar] [CrossRef]
  69. Farquhar, J.K.; Scott, W.N.; Coe, F.L. Hexose monophosphate shunt activity in compensatory renal hypertrophy. Proc. Soc. Exp. Biol. Med. 1968, 129, 809–812. [Google Scholar] [CrossRef]
  70. Schaffer, W.T. Effects of growth hormone on lipogenic enzyme activities in cultured rat hepatocytes. Am. J. Physiol. 1985, 248, E719–E725. [Google Scholar] [CrossRef]
  71. Sulis, E. G.-6-PD deficiency and cancer. Lancet 1972, 1, 1185. [Google Scholar] [CrossRef]
  72. Beaconsfield, P. Local metabolic response to physio-pathological demands: The pentose phosphate pathway. Experientia 1963, 19, 437–438. [Google Scholar] [CrossRef]
  73. Beatty, C.H.; Peterson, R.D.; Basinger, G.M.; Bocek, R.M. Major metabolic pathways for carbohydrate metabolism of voluntary skeletal muscle. Am. J. Physiol. 1966, 210, 404–410. [Google Scholar] [CrossRef]
  74. Wagner, K.R.; Kauffman, F.C.; Max, S.R. The pentose phosphate pathway in regenerating skeletal muscle. Biochem. J. 1978, 170, 17–22. [Google Scholar] [CrossRef]
  75. Rifenberick, D.H.; Koski, C.L.; Max, S.R. Metabolic studies of skeletal muscle regeneration. Exp. Neurol. 1974, 45, 527–540. [Google Scholar] [CrossRef]
  76. Smith, B. Histochemical changes in muscle necrosis and regeneration. J. Pathol. Bacteriol. 1965, 89, 139–143. [Google Scholar] [CrossRef] [PubMed]
  77. Snow, M.H. Metabolic activity during the degenerative and early regenerative stages of minced skeletal muscle. Anat. Rec. 1973, 176, 185–203. [Google Scholar] [CrossRef]
  78. Beaconsfield, P.; Carpi, A. Localization of an infectious lesion and glucose metabolism via the pentose phosphate pathway. Nature 1964, 201, 825–827. [Google Scholar] [CrossRef]
  79. Boveris, A.; Erecinska, M.; Wagner, M. Reduction kinetics of cytochromes b. Biochim. Biophys. Acta 1972, 256, 223–242. [Google Scholar] [CrossRef]
  80. Beaconsfield, P.; Reading, H.W. Pathways of glucose metabolism and nucleic acid synthesis. Nature 1964, 202, 464–466. [Google Scholar] [CrossRef]
  81. Susheela, A.K.; Hudgson, P.; Walton, J.N. Murine muscular dystrophy. Some histochemical and biochemical observations. J. Neurol. Sci. 1968, 7, 437–463. [Google Scholar] [CrossRef]
  82. Carlson, B.M. Regeneration of the rat gastrocnemius muscle from sibling and non-sibling muscle fragments. Am. J. Anat. 1970, 128, 21–31. [Google Scholar] [CrossRef] [PubMed]
  83. Carlson, B.M. Relationship between the tissue and epimorphic regeneration of muscles. Am. Zool. 1970, 10, 175–186. [Google Scholar] [CrossRef] [PubMed]
  84. Lee-Young, R.S.; Hoffman, N.J.; Murphy, K.T.; Henstridge, D.C.; Samocha-Bonet, D.; Siebel, A.L.; Iliades, P.; Zivanovic, B.; Hong, Y.H.; Colgan, T.D.; et al. Glucose-6-phosphate dehydrogenase contributes to the regulation of glucose uptake in skeletal muscle. Mol. Metab. 2016, 5, 1083–1091. [Google Scholar] [CrossRef] [PubMed]
  85. Kimmick, G.; Owen, J. Rhabdomyolysis and hemolysis associated with sickle cell trait and glucose-6-phosphate dehydrogenase deficiency. South Med. J. 1996, 89, 1097–1098. [Google Scholar] [CrossRef]
  86. Meijer, A.E.; Elias, E.A. The inhibitory effect of actinomycin D and cycloheximide on the increase in activity of glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase in experimentally induced diseased skeletal muscles. Histochem. J. 1984, 16, 971–982. [Google Scholar] [CrossRef]
  87. Ninfali, P.; Baronciani, L.; Bardoni, A.; Bresolin, N. Muscle expression of glucose-6-phosphate dehydrogenase deficiency in different variants. Clin. Genet. 1995, 48, 232–237. [Google Scholar] [CrossRef]
  88. Legan, S.K.; Rebrin, I.; Mockett, R.J.; Radyuk, S.N.; Klichko, V.I.; Sohal, R.S.; Orr, W.C. Overexpression of glucose-6-phosphate dehydrogenase extends the life span of Drosophila melanogaster. J. Biol. Chem. 2008, 283, 32492–32499. [Google Scholar] [CrossRef]
  89. Schulpis, K.H.; Reclos, G.J.; Parthimos, T.; Parthimos, N.; Gavriilidis, A.; Tsakiris, S. L-cysteine supplementation protects the erythrocyte glucose-6-phosphate dehydrogenase activity from reduction induced by forced training. Clin. Biochem. 2006, 39, 1002–1006. [Google Scholar] [CrossRef]
  90. Tsakiris, S.; Reclos, G.J.; Parthimos, T.; Tsakiris, T.; Parthimos, N.; Schulpis, K.H. α-Tocopherol supplementation restores the reduction of erythrocyte glucose-6-phosphate dehydrogenase activity induced by forced training. Pharmacol. Res. 2006, 54, 373–379. [Google Scholar] [CrossRef]
  91. Schulpis, K.H.; Tsironi, M.; Skenderi, K.; Lazaropoulou, C.; Parthimos, N.; Reclos, G.; Goussetis, E.; Tsakiris, S.; Papassotiriou, I. Dramatic reduction of erythrocyte glucose-6-phosphate dehydrogenase activity in athletes participating in the ultradistance foot race “Spartathlon”. Scand. J. Clin. Lab. Investig. 2008, 68, 228–232. [Google Scholar] [CrossRef]
  92. Tsakiris, S.; Parthimos, T.; Reclos, G.J.; Parthimos, N.; Tsakiris, T.; Schulpis, K.H. Significant reduction of erythrocyte glucose-6-phosphate dehydrogenase activity in soccer-players during play. Evidence for catecholamine mediated enzyme inhibition. Clin. Chem. Lab. Med. 2009, 47, 621–624. [Google Scholar] [CrossRef]
  93. Boström, S.; Fahlén, M.; Hjalmarson, A.; Johansson, R. Activities of rat muscle enzymes after acute exercise. Acta Physiol. Scand. 1974, 90, 544–554. [Google Scholar] [CrossRef]
  94. Schwane, J.A.; Armstrong, R.B. Effect of training on skeletal muscle injury from downhill running in rats. J. Appl. Physiol. Respir. Environ. Exerc. Physiol. 1983, 55, 969–975. [Google Scholar] [CrossRef]
  95. Farenia, R.; Lesmana, R.; Uchida, K.; Iwasaki, T.; Koibuchi, N.; Shimokawa, N. Changes in biomarker levels and myofiber constitution in rat soleus muscle at different exercise intensities. Mol. Cell. Biochem. 2019, 458, 79–87. [Google Scholar] [CrossRef]
  96. Armstrong, R.B.; Ogilvie, R.W.; Schwane, J.A. Eccentric exercise-induced injury to rat skeletal muscle. J. Appl. Physiol. 1983, 54, 80–93. [Google Scholar] [CrossRef]
  97. Boström, S.; Fahlen, M.; Hjalmarsson, A.; Johansson, R.G. Muscle enzyme activities after repeated ischemia. Int. J. Biochem. 1974, 5, 359–363. [Google Scholar] [CrossRef]
  98. Gudbjarnason, S.; Braasch, W.; Cowan, C.; Bing, R.J. Metabolism of infarcted heart muscle during tissue repair. Am. J. Cardiol. 1968, 22, 360–369. [Google Scholar] [CrossRef]
  99. Valentino, T.; Figueiredo, V.C.; Mobley, C.B.; McCarthy, J.J.; Vechetti, I.J. Evidence of myomiR regulation of the pentose phosphate pathway during mechanical load-induced hypertrophy. Physiol. Rep. 2021, 9, e15137. [Google Scholar] [CrossRef]
  100. Turner, L.V.; Manchester, K.L. Glucose and glycogen metabolism in hypertrophied denervated rat hemidiaphragm. Biochem. J. 1970, 117, 33P. [Google Scholar] [CrossRef]
  101. Weyrauch, L.A.; McMillin, S.L.; Witczak, C.A. Insulin Resistance Does Not Impair Mechanical Overload-Stimulated Glucose Uptake, but Does Alter the Metabolic Fate of Glucose in Mouse Muscle. Int. J. Mol. Sci. 2020, 21, 4715. [Google Scholar] [CrossRef]
  102. Izumiya, Y.; Hopkins, T.; Morris, C.; Sato, K.; Zeng, L.; Viereck, J.; Hamilton, J.A.; Ouchi, N.; LeBrasseur, N.K.; Walsh, K. Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metab. 2008, 7, 159–172. [Google Scholar] [CrossRef]
  103. Wu, C.L.; Satomi, Y.; Walsh, K. RNA-seq and metabolomic analyses of Akt1-mediated muscle growth reveals regulation of regenerative pathways and changes in the muscle secretome. BMC Genom. 2017, 18, 181. [Google Scholar] [CrossRef]
  104. Tullson, P.; Armstrong, R.B. Muscle hexose monophosphate shunt activity following exercise. Experientia 1981, 37, 1311–1312. [Google Scholar] [CrossRef]
  105. Novak, M.L.; Billich, W.; Smith, S.M.; Sukhija, K.B.; McLoughlin, T.J.; Hornberger, T.A.; Koh, T.J. COX-2 inhibitor reduces skeletal muscle hypertrophy in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 296, R1132–R1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Jiang, A.; Dong, C.; Li, B.; Zhang, Z.; Chen, Y.; Ning, C.; Wu, W.; Liu, H. MicroRNA-206 regulates cell proliferation by targeting G6PD in skeletal muscle. FASEB J. 2019, 33, 14083–14094. [Google Scholar] [CrossRef] [PubMed]
  107. Hoshino, D.; Kawata, K.; Kunida, K.; Hatano, A.; Yugi, K.; Wada, T.; Fujii, M.; Sano, T.; Ito, Y.; Furuichi, Y.; et al. Trans-omic Analysis Reveals ROS-Dependent Pentose Phosphate Pathway Activation after High-Frequency Electrical Stimulation in C2C12 Myotubes. iScience 2020, 23, 101558. [Google Scholar] [CrossRef] [PubMed]
  108. Kirby, T.J.; Patel, R.M.; McClintock, T.S.; Dupont-Versteegden, E.E.; Peterson, C.A.; McCarthy, J.J. Myonuclear transcription is responsive to mechanical load and DNA content but uncoupled from cell size during hypertrophy. Mol. Biol. Cell 2016, 27, 788–798. [Google Scholar] [CrossRef] [PubMed]
  109. Kovacheva, E.L.; Hikim, A.P.; Shen, R.; Sinha, I.; Sinha-Hikim, I. Testosterone supplementation reverses sarcopenia in aging through regulation of myostatin, c-Jun NH2-terminal kinase, Notch, and Akt signaling pathways. Endocrinology 2010, 151, 628–638. [Google Scholar] [CrossRef]
  110. Brioche, T.; Kireev, R.A.; Cuesta, S.; Gratas-Delamarche, A.; Tresguerres, J.A.; Gomez-Cabrera, M.C.; Viña, J. Growth hormone replacement therapy prevents sarcopenia by a dual mechanism: Improvement of protein balance and of antioxidant defenses. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2014, 69, 1186–1198. [Google Scholar] [CrossRef]
  111. Laguens, R.P.; Gómez Dumm, C.L. Deoxyribonucleic acid synthesis in the heart mitochondria after acute and exhaustive exercise. Experientia 1968, 24, 163–164. [Google Scholar] [CrossRef]
  112. Place, N.; Ivarsson, N.; Venckunas, T.; Neyroud, D.; Brazaitis, M.; Cheng, A.J.; Ochala, J.; Kamandulis, S.; Girard, S.; Volungevicius, G.; et al. Ryanodine receptor fragmentation and sarcoplasmic reticulum Ca2+ leak after one session of high-intensity interval exercise. Proc. Natl. Acad. Sci. USA 2015, 112, 15492–15497. [Google Scholar] [CrossRef]
  113. Ristow, M.; Zarse, K.; Oberbach, A.; Kloting, N.; Birringer, M.; Kiehntopf, M.; Stumvoll, M.; Kahn, C.R.; Bluher, M. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc. Natl. Acad. Sci. USA 2009, 106, 8665–8670. [Google Scholar] [CrossRef]
  114. Paulsen, G.; Cumming, K.T.; Holden, G.; Hallen, J.; Ronnestad, B.R.; Sveen, O.; Skaug, A.; Paur, I.; Bastani, N.E.; Ostgaard, H.N.; et al. Vitamin C and E supplementation hampers cellular adaptation to endurance training in humans: A double-blind, randomised, controlled trial. J. Physiol. 2014, 592, 1887–1901. [Google Scholar] [CrossRef]
  115. Gomez-Cabrera, M.C.; Domenech, E.; Romagnoli, M.; Arduini, A.; Borras, C.; Pallardo, F.V.; Sastre, J.; Vina, J. Oral administration of vitamin C decreases muscle mitochondrial biogenesis and hampers training-induced adaptations in endurance performance. Am. J. Clin. Nutr. 2008, 87, 142–149. [Google Scholar] [CrossRef] [Green Version]
  116. Melikoglu, M.A.; Kaldirimci, M.; Katkat, D.; Sen, I.; Kaplan, I.; Senel, K. The effect of regular long term training on antioxidant enzymatic activities. J. Sports Med. Phys. Fit. 2008, 48, 388–390. [Google Scholar]
  117. Spodaryk, K.; Szyguła, Z.; Dabrowski, Z.; Miszta, H. The activity of erythrocyte enzymes in rats subjected to running exercises. Eur. J. Appl. Physiol. Occup. Physiol. 1985, 54, 533–537. [Google Scholar] [CrossRef]
  118. Gomez-Cabrera, M.C.; Borrás, C.; Pallardó, F.V.; Sastre, J.; Ji, L.L.; Viña, J. Decreasing xanthine oxidase-mediated oxidative stress prevents useful cellular adaptations to exercise in rats. J. Physiol. 2005, 567, 113–120. [Google Scholar] [CrossRef]
  119. Gomez-Cabrera, M.C.; Domenech, E.; Viña, J. Moderate exercise is an antioxidant: Upregulation of antioxidant genes by training. Free Radic. Biol. Med. 2008, 44, 126–131. [Google Scholar] [CrossRef]
  120. Brioche, T.; Pagano, A.F.; Py, G.; Chopard, A. Muscle wasting and aging: Experimental models, fatty infiltrations, and prevention. Mol. Aspects Med. 2016, 50, 56–87. [Google Scholar] [CrossRef]
  121. Herscovich, S.; Gershon, D. Effects of aging and physical training on the neuromuscular junction of the mouse. Gerontology 1987, 33, 7–13. [Google Scholar] [CrossRef]
  122. Griffiths, M.A.; Baker, D.H.; Novakofski, J.E.; Ji, L.L. Effects of exercise training on diet-induced lipogenic enzymes and body composition in rats. J. Am Coll. Nutr. 1993, 12, 155–161. [Google Scholar] [CrossRef]
  123. Pereira, B.; Costa Rosa, L.F.; Safi, D.A.; Medeiros, M.H.; Curi, R.; Bechara, E.J. Superoxide dismutase, catalase, and glutathione peroxidase activities in muscle and lymphoid organs of sedentary and exercise-trained rats. Physiol. Behav. 1994, 56, 1095–1099. [Google Scholar] [CrossRef]
  124. Borges-Silva, C.N.; Fonseca-Alaniz, M.H.; Alonso-Vale, M.I.; Takada, J.; Andreotti, S.; Peres, S.B.; Cipolla-Neto, J.; Pithon-Curi, T.C.; Lima, F.B. Reduced lipolysis and increased lipogenesis in adipose tissue from pinealectomized rats adapted to training. J. Pineal. Res. 2005, 39, 178–184. [Google Scholar] [CrossRef] [PubMed]
  125. Ninfali, P.; Bresolin, N. Muscle glucose 6-phosphate dehydrogenase (G6PD) deficiency and oxidant stress during physical exercise. Cell Biochem. Funct. 1995, 13, 297–298. [Google Scholar] [CrossRef]
  126. Jamurtas, A.Z.; Fatouros, I.G.; Deli, C.K.; Georgakouli, K.; Poulios, A.; Draganidis, D.; Papanikolaou, K.; Tsimeas, P.; Chatzinikolaou, A.; Avloniti, A.; et al. The Effects of Acute Low-Volume HIIT and Aerobic Exercise on Leukocyte Count and Redox Status. J. Sports Sci. Med. 2018, 17, 501–508. [Google Scholar]
  127. Georgakouli, K.; Fatouros, I.G.; Draganidis, D.; Papanikolaou, K.; Tsimeas, P.; Deli, C.K.; Jamurtas, A.Z. Exercise in Glucose-6-Phosphate Dehydrogenase Deficiency: Harmful or Harmless? A Narrative Review. Oxid. Med. Cell. Longev. 2019, 2019, 8060193. [Google Scholar] [CrossRef] [Green Version]
  128. Jamurtas, A.Z.; Fatouros, I.G.; Koukosias, N.; Manthou, E.; Tofas, T.; Yfanti, C.; Nikolaidis, M.G.; Koutedakis, Y. Effect of exercise on oxidative stress in individuals with glucose-6-phosphate dehydrogenase deficiency. In Vivo 2006, 20, 875–880. [Google Scholar]
  129. Theodorou, A.A.; Nikolaidis, M.G.; Paschalis, V.; Sakellariou, G.K.; Fatouros, I.G.; Koutedakis, Y.; Jamurtas, A.Z. Comparison between glucose-6-phosphate dehydrogenase-deficient and normal individuals after eccentric exercise. Med. Sci. Sports Exerc. 2010, 42, 1113–1121. [Google Scholar] [CrossRef]
  130. Makarona, K.; Caputo, V.S.; Costa, J.R.; Liu, B.; O’Connor, D.; Iskander, D.; Roper, D.; Robertson, L.; Bhatnagar, N.; Terpos, E.; et al. Transcriptional and epigenetic basis for restoration of G6PD enzymatic activity in human G6PD-deficient cells. Blood 2014, 124, 134–141. [Google Scholar] [CrossRef] [PubMed]
  131. Yang, L.; He, Z.; Yao, J.; Tan, R.; Zhu, Y.; Li, Z.; Guo, Q.; Wei, L. Regulation of AMPK-related glycolipid metabolism imbalances redox homeostasis and inhibits anchorage independent growth in human breast cancer cells. Redox Biol. 2018, 17, 180–191. [Google Scholar] [CrossRef] [PubMed]
  132. Hwang, S.; Mruk, K.; Rahighi, S.; Raub, A.G.; Chen, C.H.; Dorn, L.E.; Horikoshi, N.; Wakatsuki, S.; Chen, J.K.; Mochly-Rosen, D. Correcting glucose-6-phosphate dehydrogenase deficiency with a small-molecule activator. Nat. Commun. 2018, 9, 4045. [Google Scholar] [CrossRef]
  133. Leopold, J.A.; Dam, A.; Maron, B.A.; Scribner, A.W.; Liao, R.; Handy, D.E.; Stanton, R.C.; Pitt, B.; Loscalzo, J. Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nat. Med. 2007, 13, 189–197. [Google Scholar] [CrossRef]
  134. Wagle, A.; Jivraj, S.; Garlock, G.L.; Stapleton, S.R. Insulin regulation of glucose-6-phosphate dehydrogenase gene expression is rapamycin-sensitive and requires phosphatidylinositol 3-kinase. J. Biol. Chem. 1998, 273, 14968–14974. [Google Scholar] [CrossRef]
  135. Cosentino, C.; Grieco, D.; Costanzo, V. ATM activates the pentose phosphate pathway promoting anti-oxidant defence and DNA repair. EMBO J. 2011, 30, 546–555. [Google Scholar] [CrossRef]
  136. Talukdar, I.; Szeszel-Fedorowicz, W.; Salati, L.M. Arachidonic acid inhibits the insulin induction of glucose-6-phosphate dehydrogenase via p38 MAP kinase. J. Biol. Chem. 2005, 280, 40660–40667. [Google Scholar] [CrossRef]
  137. Katare, R.; Caporali, A.; Emanueli, C.; Madeddu, P. Benfotiamine improves functional recovery of the infarcted heart via activation of pro-survival G6PD/Akt signaling pathway and modulation of neurohormonal response. J. Mol. Cell. Cardiol. 2010, 49, 625–638. [Google Scholar] [CrossRef]
  138. Zhang, Z.; Apse, K.; Pang, J.; Stanton, R.C. High glucose inhibits glucose-6-phosphate dehydrogenase via cAMP in aortic endothelial cells. J. Biol. Chem. 2000, 275, 40042–40047. [Google Scholar] [CrossRef] [Green Version]
  139. Patel, D.; Kandhi, S.; Kelly, M.; Neo, B.H.; Wolin, M.S. Dehydroepiandrosterone promotes pulmonary artery relaxation by NADPH oxidation-elicited subunit dimerization of protein kinase G 1α. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 306, L383–L391. [Google Scholar] [CrossRef]
  140. Wang, H.; Nicolay, B.N.; Chick, J.M.; Gao, X.; Geng, Y.; Ren, H.; Gao, H.; Yang, G.; Williams, J.A.; Suski, J.M.; et al. The metabolic function of cyclin D3-CDK6 kinase in cancer cell survival. Nature 2017, 546, 426–430. [Google Scholar] [CrossRef]
  141. Schwartz, A.G.; Pashko, L.L. Dehydroepiandrosterone, glucose-6-phosphate dehydrogenase, and longevity. Ageing Res. Rev. 2004, 3, 171–187. [Google Scholar] [CrossRef]
  142. Tsao, M.S.; Earp, H.S.; Grisham, J.W. The effects of epidermal growth factor and the state of confluence on enzymatic activities of cultured rat liver epithelial cells. J. Cell. Physiol. 1986, 126, 167–173. [Google Scholar] [CrossRef]
  143. Köberle, V.; Kronenberger, B.; Pleli, T.; Trojan, J.; Imelmann, E.; Peveling-Oberhag, J.; Welker, M.W.; Elhendawy, M.; Zeuzem, S.; Piiper, A.; et al. Serum microRNA-1 and microRNA-122 are prognostic markers in patients with hepatocellular carcinoma. Eur. J. Cancer 2013, 49, 3442–3449. [Google Scholar] [CrossRef]
  144. Jiang, P.; Du, W.; Wang, X.; Mancuso, A.; Gao, X.; Wu, M.; Yang, X. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 2011, 13, 310–316. [Google Scholar] [CrossRef] [PubMed]
  145. Rao, X.; Duan, X.; Mao, W.; Li, X.; Li, Z.; Li, Q.; Zheng, Z.; Xu, H.; Chen, M.; Wang, P.G.; et al. O-GlcNAcylation of G6PD promotes the pentose phosphate pathway and tumor growth. Nat. Commun. 2015, 6, 8468. [Google Scholar] [CrossRef] [PubMed]
  146. Hong, X.; Song, R.; Song, H.; Zheng, T.; Wang, J.; Liang, Y.; Qi, S.; Lu, Z.; Song, X.; Jiang, H.; et al. PTEN antagonises Tcl1/hnRNPK-mediated G6PD pre-mRNA splicing which contributes to hepatocarcinogenesis. Gut 2014, 63, 1635–1647. [Google Scholar] [CrossRef] [PubMed]
  147. Aird, K.M.; Worth, A.J.; Snyder, N.W.; Lee, J.V.; Sivanand, S.; Liu, Q.; Blair, I.A.; Wellen, K.E.; Zhang, R. ATM couples replication stress and metabolic reprogramming during cellular senescence. Cell Rep. 2015, 11, 893–901. [Google Scholar] [CrossRef] [PubMed]
  148. Préville, X.; Salvemini, F.; Giraud, S.; Chaufour, S.; Paul, C.; Stepien, G.; Ursini, M.V.; Arrigo, A.P. Mammalian small stress proteins protect against oxidative stress through their ability to increase glucose-6-phosphate dehydrogenase activity and by maintaining optimal cellular detoxifying machinery. Exp. Cell Res. 1999, 247, 61–78. [Google Scholar] [CrossRef] [PubMed]
  149. Yi, W.; Clark, P.M.; Mason, D.E.; Keenan, M.C.; Hill, C.; Goddard, W.A.; Peters, E.C.; Driggers, E.M.; Hsieh-Wilson, L.C. Phosphofructokinase 1 glycosylation regulates cell growth and metabolism. Science 2012, 337, 975–980. [Google Scholar] [CrossRef]
  150. Yin, X.; Tang, B.; Li, J.H.; Wang, Y.; Zhang, L.; Xie, X.Y.; Zhang, B.H.; Qiu, S.J.; Wu, W.Z.; Ren, Z.G. ID1 promotes hepatocellular carcinoma proliferation and confers chemoresistance to oxaliplatin by activating pentose phosphate pathway. J. Exp. Clin. Cancer Res. 2017, 36, 166. [Google Scholar] [CrossRef]
  151. Nakamura, T.; Yoshimoto, K.; Aoyama, K.; Ichihara, A. Hormonal regulations of glucose-6-phosphate dehydrogenase and lipogenesis in primary cultures of rat hepatocytes. J. Biochem. 1982, 91, 681–693. [Google Scholar] [CrossRef]
  152. Tsouko, E.; Khan, A.S.; White, M.A.; Han, J.J.; Shi, Y.; Merchant, F.A.; Sharpe, M.A.; Xin, L.; Frigo, D.E. Regulation of the pentose phosphate pathway by an androgen receptor-mTOR-mediated mechanism and its role in prostate cancer cell growth. Oncogenesis 2014, 3, e103. [Google Scholar] [CrossRef]
  153. Zimta, A.A.; Cenariu, D.; Irimie, A.; Magdo, L.; Nabavi, S.M.; Atanasov, A.G.; Berindan-Neagoe, I. The Role of Nrf2 Activity in Cancer Development and Progression. Cancers 2019, 11, 1755. [Google Scholar] [CrossRef]
  154. Zhang, X.; Li, Y.; Shao, Y.; Xiao, J.; Zhu, G.; Li, F. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017, 8, e2820. [Google Scholar] [CrossRef]
  155. Yang, X.; Ye, H.; He, M.; Zhou, X.; Sun, N.; Guo, W.; Lin, X.; Huang, H.; Lin, Y.; Yao, R.; et al. LncRNA PDIA3P interacts with c-Myc to regulate cell proliferation via induction of pentose phosphate pathway in multiple myeloma. Biochem. Biophys. Res. Commun. 2018, 498, 207–213. [Google Scholar] [CrossRef]
  156. Tian, W.N.; Pignatare, J.N.; Stanton, R.C. Signal transduction proteins that associate with the platelet-derived growth factor (PDGF) receptor mediate the PDGF-induced release of glucose-6-phosphate dehydrogenase from permeabilized cells. J. Biol. Chem. 1994, 269, 14798–14805. [Google Scholar] [CrossRef]
  157. Ma, X.; Wang, L.; Huang, D.; Li, Y.; Yang, D.; Li, T.; Li, F.; Sun, L.; Wei, H.; He, K.; et al. Polo-like kinase 1 coordinates biosynthesis during cell cycle progression by directly activating pentose phosphate pathway. Nat. Commun. 2017, 8, 1506. [Google Scholar] [CrossRef]
  158. Thakur, A.; Rahman, K.W.; Wu, J.; Bollig, A.; Biliran, H.; Lin, X.; Nassar, H.; Grignon, D.J.; Sarkar, F.H.; Liao, J.D. Aberrant expression of X-linked genes RbAp46, Rsk4, and Cldn2 in breast cancer. Mol. Cancer Res. 2007, 5, 171–181. [Google Scholar] [CrossRef] [Green Version]
  159. Kim, N.H.; Cha, Y.H.; Lee, J.; Lee, S.H.; Yang, J.H.; Yun, J.S.; Cho, E.S.; Zhang, X.; Nam, M.; Kim, N.; et al. Snail reprograms glucose metabolism by repressing phosphofructokinase PFKP allowing cancer cell survival under metabolic stress. Nat. Commun. 2017, 8, 14374. [Google Scholar] [CrossRef]
  160. Ulusu, N.N.; Sahilli, M.; Avci, A.; Canbolat, O.; Ozansoy, G.; Ari, N.; Bali, M.; Stefek, M.; Stolc, S.; Gajdosik, A.; et al. Pentose phosphate pathway, glutathione-dependent enzymes and antioxidant defense during oxidative stress in diabetic rodent brain and peripheral organs: Effects of stobadine and vitamin E. Neurochem. Res. 2003, 28, 815–823. [Google Scholar] [CrossRef]
  161. Du, W.; Jiang, P.; Mancuso, A.; Stonestrom, A.; Brewer, M.D.; Minn, A.J.; Mak, T.W.; Wu, M.; Yang, X. TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat. Cell Biol. 2013, 15, 991–1000. [Google Scholar] [CrossRef]
  162. Zhang, R.; Tao, F.; Ruan, S.; Hu, M.; Hu, Y.; Fang, Z.; Mei, L.; Gong, C. The TGFβ1-FOXM1-HMGA1-TGFβ1 positive feedback loop increases the cisplatin resistance of non-small cell lung cancer by inducing G6PD expression. Am. J. Transl. Res. 2019, 11, 6860–6876. [Google Scholar]
  163. Wang, J.; Duan, Z.; Nugent, Z.; Zou, J.X.; Borowsky, A.D.; Zhang, Y.; Tepper, C.G.; Li, J.J.; Fiehn, O.; Xu, J.; et al. Reprogramming metabolism by histone methyltransferase NSD2 drives endocrine resistance via coordinated activation of pentose phosphate pathway enzymes. Cancer Lett. 2016, 378, 69–79. [Google Scholar] [CrossRef] [Green Version]
  164. Sardar, S.; Chakraborty, A.; Chatterjee, M. Comparative effectiveness of vitamin D3 and dietary vitamin E on peroxidation of lipids and enzymes of the hepatic antioxidant system in Sprague—Dawley rats. Int. J. Vitam. Nutr. Res. 1996, 66, 39–45. [Google Scholar]
Figure 1. The pentose phosphate pathway and its interrelation with glycolysis. Blue and orange arrows show reversible and irreversible reactions, respectively. Enzyme names are shown in blue and orange boxes. Glycolysis metabolites obtained through the PPP are shown in red squares. G6P—glucose 6-phosphate; F6P—fructose 6-phosphate; F1,6P—fructose 1,6-biphosphate; DHAP—dihydroxyacetone phosphate; G3P—glyceraldehyde 3-phosphate; 1,3 BPG—1,3-bisphosphoglycerate; 3PG—3-phosphoglycerate; 2PG—2-phosphoglycerate; PEP—phosphoenolpyruvate; 6PG—6-phosphogluconate; HK—hexokinase; PGI—phosphoglucoisomerase; PFK—phosphofructokinase; FBA—fructose-1,6-bisphosphate aldolase; TPI—triose-phosphate isomerase; GAPDH—glyceraldehyde 3-phosphate dehydrogenase; PGK—phosphoglycerate kinase; PGM—phosphoglycerate mutase; G6PD—glucose 6-phosphate dehydrogenase; 6PGD—6-phosphogluconate dehydrogenase; RPE—ribulose-phosphate 3-epimerase; RPI—ribose-5-phosphate isomerase; TKL—transketolase; TAL: transaldolase.
Figure 1. The pentose phosphate pathway and its interrelation with glycolysis. Blue and orange arrows show reversible and irreversible reactions, respectively. Enzyme names are shown in blue and orange boxes. Glycolysis metabolites obtained through the PPP are shown in red squares. G6P—glucose 6-phosphate; F6P—fructose 6-phosphate; F1,6P—fructose 1,6-biphosphate; DHAP—dihydroxyacetone phosphate; G3P—glyceraldehyde 3-phosphate; 1,3 BPG—1,3-bisphosphoglycerate; 3PG—3-phosphoglycerate; 2PG—2-phosphoglycerate; PEP—phosphoenolpyruvate; 6PG—6-phosphogluconate; HK—hexokinase; PGI—phosphoglucoisomerase; PFK—phosphofructokinase; FBA—fructose-1,6-bisphosphate aldolase; TPI—triose-phosphate isomerase; GAPDH—glyceraldehyde 3-phosphate dehydrogenase; PGK—phosphoglycerate kinase; PGM—phosphoglycerate mutase; G6PD—glucose 6-phosphate dehydrogenase; 6PGD—6-phosphogluconate dehydrogenase; RPE—ribulose-phosphate 3-epimerase; RPI—ribose-5-phosphate isomerase; TKL—transketolase; TAL: transaldolase.
Cells 11 03041 g001
Figure 2. NADPH-dependent enzymes. NOs—nitric oxide synthases; NOx—NADPH-oxidase enzymes; DHFR—dihydrofolate reductase.
Figure 2. NADPH-dependent enzymes. NOs—nitric oxide synthases; NOx—NADPH-oxidase enzymes; DHFR—dihydrofolate reductase.
Cells 11 03041 g002
Figure 3. Pentose phosphate pathway regulation depends on cellular needs. MODE 1: This mode dominates when the need for R5P is higher than that for NADPH, for instance, in proliferative cells. In this situation, the glycolytic metabolites 3GP and F6P can be converted in R5P through the reversible non-oxidative PPP. The oxidative PPP and its associated NADPH formation are bypassed. MODE 2: This mode occurs when the needs for NADPH and R5P are balanced. Then, ideally, from one molecule of G6P two molecules of NADPH and a molecule of R5P can be obtained with no generation of glycolytic metabolite. MODE 3: This mode is adopted when the cellular need for NADPH exceeds that for R5P and ATP, for instance, during fatty acid synthesis in adipocytes. The non-oxidative phase of the pathway leads to the conversion of ribulose 5-phosphate to fructose 6-phosphate (F6P) and glyceraldehyde 3-phosphate (G3P). Then, these glycolytic metabolites—through gluconeogenesis reactions—form G6P, which can enter again into the PPP to produce more NADPH. MODE 4: In this scenario, the cellular need for NADPH and ATP is higher than that for R5P. As described in PPP mode 3, ribulose 5-P is transformed into G3P and F5P through the non-oxidative branch of the PPP; however, in mode 4, these molecules are metabolized to pyruvate through glycolysis, which is associated with ATP formation.
Figure 3. Pentose phosphate pathway regulation depends on cellular needs. MODE 1: This mode dominates when the need for R5P is higher than that for NADPH, for instance, in proliferative cells. In this situation, the glycolytic metabolites 3GP and F6P can be converted in R5P through the reversible non-oxidative PPP. The oxidative PPP and its associated NADPH formation are bypassed. MODE 2: This mode occurs when the needs for NADPH and R5P are balanced. Then, ideally, from one molecule of G6P two molecules of NADPH and a molecule of R5P can be obtained with no generation of glycolytic metabolite. MODE 3: This mode is adopted when the cellular need for NADPH exceeds that for R5P and ATP, for instance, during fatty acid synthesis in adipocytes. The non-oxidative phase of the pathway leads to the conversion of ribulose 5-phosphate to fructose 6-phosphate (F6P) and glyceraldehyde 3-phosphate (G3P). Then, these glycolytic metabolites—through gluconeogenesis reactions—form G6P, which can enter again into the PPP to produce more NADPH. MODE 4: In this scenario, the cellular need for NADPH and ATP is higher than that for R5P. As described in PPP mode 3, ribulose 5-P is transformed into G3P and F5P through the non-oxidative branch of the PPP; however, in mode 4, these molecules are metabolized to pyruvate through glycolysis, which is associated with ATP formation.
Cells 11 03041 g003
Table 1. Classification of G6PD mutations.
Table 1. Classification of G6PD mutations.
ClassMutation Severity% of Normal G6PD Function
Class ISevere deficiency associated with chronic non-spherocytic hemolytic anemia<1
Class IIResidual activity associated with acute hemolytic anemia1–10
Class IIIMild10–60
Class IVNormal activity60–150
Class VMore than normal activity>150
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

García-Domínguez, E.; Carretero, A.; Viña-Almunia, A.; Domenech-Fernandez, J.; Olaso-Gonzalez, G.; Viña, J.; Gomez-Cabrera, M.C. Glucose 6-P Dehydrogenase—An Antioxidant Enzyme with Regulatory Functions in Skeletal Muscle during Exercise. Cells 2022, 11, 3041. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11193041

AMA Style

García-Domínguez E, Carretero A, Viña-Almunia A, Domenech-Fernandez J, Olaso-Gonzalez G, Viña J, Gomez-Cabrera MC. Glucose 6-P Dehydrogenase—An Antioxidant Enzyme with Regulatory Functions in Skeletal Muscle during Exercise. Cells. 2022; 11(19):3041. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11193041

Chicago/Turabian Style

García-Domínguez, Esther, Aitor Carretero, Aurora Viña-Almunia, Julio Domenech-Fernandez, Gloria Olaso-Gonzalez, Jose Viña, and Mari Carmen Gomez-Cabrera. 2022. "Glucose 6-P Dehydrogenase—An Antioxidant Enzyme with Regulatory Functions in Skeletal Muscle during Exercise" Cells 11, no. 19: 3041. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11193041

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop