Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jul 16.
Published in final edited form as: Nat Metab. 2023 Aug 23;5(8):1275–1289. doi: 10.1038/s42255-023-00863-2

The pentose phosphate pathway in health and disease

Tara TeSlaa 1, Markus Ralser 2,3,4, Jing Fan 5,6, Joshua D Rabinowitz 7,8,9,
PMCID: PMC11251397  NIHMSID: NIHMS2003188  PMID: 37612403

Abstract

The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.


The PPP is a glucose-metabolizing pathway that comprises two branches. The input of the oxidative branch (oxPPP) is glucose 6-phosphate, which also feeds glycolysis. The oxPPP converts glucose 6-phosphate into ribose 5-phosphate while making two molecules of NADPH. The non-oxidative branch (non-oxPPP) reversibly interconverts ribose 5-phosphate with glycolytic intermediates. In most healthy mammalian cells in unstressed conditions, the PPP exhibits much lower flux than glycolysis, for example, by 10–100-fold1,2. Nevertheless, the PPP carries higher flux than most pathways outside central metabolism. Moreover, it can be transiently activated in response to oxidative stress or during the phagocyte oxidative burst to meet urgent NADPH demands.

One function of the PPP is to provide biosynthetic precursors, with ribose 5-phosphate an essential nucleotide building block and NADPH used for biosynthesis of fatty acids, cholesterol, proline, tetrahydrofolate and deoxyribonucleotides. From bacteria to mammals, the oxPPP is a major source of cytosolic NADPH to support these processes3,4. However, alternative routes for cytosolic NADPH production exist, including isocitrate dehydrogenase 1 (IDH1), malic enzyme 1 (ME1), methylenetetrahydrofolate dehydrogenase and formyltetrahydrofolate dehydrogenase, and can often compensate to generate the required NADPH for biosynthesis.

The PPP also supports cellular defence against oxidative stress, a driver of cardiovascular disease, neurodegeneration and aging5. To prevent damage from ROS and RNS, cells rely on antioxidant systems including glutathione and thioredoxin. Both use NADPH to regenerate reduced thiols from disulfides. The oxPPP may be uniquely suited for acute antioxidant responses compared to other NADPH-production routes due to its having excess capacity that is not used in the basal state but stands ready for times of enhanced demand6 (reserve flux capacity). Indeed, PPP flux upregulation keeps yeast cells alive when they encounter oxidative substances, protects skin cells from the consequences of UV irradiation and allows cancer cells to tolerate increased oxidant levels during detachment.

While often considered bad actors, ROS and RNS also play physiological roles in mammals and are produced by dedicated enzymes that use NADPH as the reductant: NADPH oxidase (NOX) or nitric oxide synthase (NOS)7. Modest amounts of ROS and RNS play signalling and biochemical roles. Larger quantities contribute to immune defence by killing invading pathogens and thereby preventing life-threatening bacterial and fungal infections. In activated phagocytes, the PPP fuels this process, shifting flux away from glycolysis to generate large amounts of NADPH8.

In this Review, we will cover the basic principles of the PPP and its regulation as well as current technical approaches to determine metabolic flux through this pathway. We will then discuss its implications in physiology and pathology in the context of red blood cells (RBCs), immune cells, tissue homoeostasis and cancer.

Basic principles of the PPP

Biochemical modes

The oxPPP pathway produces two NADPH molecules per glucose-6-phosphate molecule through two dehydrogenase steps, resulting in the formation of ribulose 5-phosphate (Fig. 1a). The first committed step, which is catalysed by G6PD, generates an initial NADPH molecule. Mutations in G6PD are the most common enzymatic defect in humans, leading to fragile RBCs, but also providing protection against malaria9. Next comes 6-phosphogluconolactonase (6PGL). Mammals also have an alternate enzyme, hexose-6-phosphate dehydrogenase (H6GD), localized in the endoplasmic reticulum, which can catalyse the first two steps of the oxPPP pathway to produce NADPH for processing corticosteroids. The final step of the oxPPP pathway, which produces the second molecule of NADPH, is catalysed by 6-phosphogluconate dehydrogenase (6PGD).

Fig. 1 |. The PPP and its modes of operation.

Fig. 1 |

a, Overview of the oxPPP, the non-oxPPP and their connections to glycolysis. Each glucose that goes through the PPP can generate two NADPH molecules and one ribose-5-phosphate molecule. Abbreviations: HK, hexokinase; GPI, glucose-6-phosphate isomerase; RPE, ribulose-phosphate 3-epimerase; RPI, ribose-5-phosphate isomerase; FBPase, fructose 1,6-bisphophatase; ALDO, fructose-bisphosphate aldolase. b, Modes of PPP operation. Unmet ribose demand (that is, pentose insufficiency) leads to net non-oxPPP flux toward ribose-5-phosphate synthesis. Higher NADPH demand than ribose demand (after accounting for 2:1 pathway stoichiometry) causes non-oxPPP flux in the opposite direction, from ribose 5-phosphate toward glycolysis (that is, pentose overflow). Very high NADPH demand can lead to pentose cycling, in which the glycolytic enzyme 6-phosphate isomerase runs in reverse to make additional glucose 6-phosphate to feed the oxPPP.

The non-oxPPP pathway allows for interconversion of various important molecules, including the oxPPP product ribulose 5-phosphate, the key nucleotide precursor ribose 5-phosphate and the glycolytic intermediates fructose 6-phosphate and glyceraldehyde 3-phosphate. These reversible reactions are catalysed by a series of enzymes, including ribulose-5-phosphate epimerase, ribose-5-phosphate isomerase, transaldolase (TALDO) and the thiamine-dependent enzyme transketolase (TK; Fig. 1a). Another intermediate in the non-oxPPP pathway, erythrose 4-phosphate, is used by microbes and plants to synthesize essential amino acids, but mammals are unable to produce these and instead obtain them through their diet. Accordingly, in mammals, the non-oxPPP pathway primarily functions to provide flexible carbon flow between pentoses and glycolysis.

The two branches of the PPP can function together in three different modes based on cellular requirements: pentose insufficiency, pentose overflow and pentose cycling (Fig. 1b). In pentose-insufficiency mode, the non-oxPPP pathway produces ribose 5-phosphate, as it is insufficiently supplied by the oxPPP pathway. In pentose-overflow mode, the non-oxPPP pathway consumes excess ribose 5-phosphate produced by the oxPPP pathway and feeds it back into glycolysis. In pentose-cycling mode, glycolytic intermediates made from excess ribose 5-phosphate are redirected up the glycolysis pathway to regenerate glucose 6-phosphate. This glucose 6-phosphate is then reused by the oxPPP pathway to produce more NADPH. Theoretically, if the gluconeogenic enzyme fructose 1,6-bisphosphatase is active, pentose cycling could lead to complete oxidation of glucose into six CO2 molecules, generating 12 NADPH molecules (Fig. 1). Although this total oxidation has been postulated for several decades, it has not yet been observed in mammalian systems. However, partial pentose cycling appears to be crucial for the oxidative burst of immune cells, which aids in pathogen killing. The PPP mode depends on the cellular context. In proliferating cells, both ribose and NADPH are consumed in parallel to support proliferation, with the PPP mode determined by whether cellular demand for nucleotides or lipids is greater. In non-proliferating cells, on the other hand, demand for ribose is typically low, while NADPH is still needed for redox and/or biosynthetic purposes (for example, fat synthesis in adipocytes), resulting in pentose overflow as seen in quiescent fibroblasts10 or pentose cycling as seen in immune cell oxidative burst11.

Regulation

PPP activity correlates with NADPH demand (Fig. 2) and depends on the provision of glucose 6-phosphate, for which it competes with other pathways (foremost glycolysis). Such coordinated metabolic activity is achieved substantially through levels of the pathway substrate NADP (in part relative to the product NADPH). NADPH consumption makes NADP, which promotes G6PD activity and thus oxPPP flux both (1) as a substrate and (2) via an allosteric binding site on G6PD12,13 (Fig. 3). The non-oxPPP in turn adapts to consume or produce pentose phosphates, as controlled by metabolite levels, which determine reaction thermodynamics and thus flux direction.

Fig. 2 |. Major NADPH-consuming pathways.

Fig. 2 |

Abbreviations: CoA, coenzyme A; GR, glutathione reductase; GPx, glutathione peroxidase; GSH, glutathione; GSSG, glutathione disulfide; TR, thioredoxin reductase; FASN, fatty acid synthase; HMGCR, 3-hydroxy-3-methylglutaryl coenzyme A reductase; SQLE, squalene epoxidase; P5CR, pyrroline-5-carboxylate reductase; RNR, ribonucleotide reductase; DHFR, dihydrofolate reductase; TRX, thioredoxin.

Fig. 3 |. Regulation of the PPP.

Fig. 3 |

The PPP and glycolysis compete for carbon flux. Factors that increase oxPPP flux are highlighted in yellow, and those that decrease it are in blue. Names of enzymes induced by NRF2 are in red. E4P, erythrose 4-phosphate; F6P, fructose 6-phosphate; F1,6BP, fructose 1,6-biphosphate; F2,6BP, fructose 2,6-biphosphate; G6P, glucose 6-phosphate; 6PG, 6-phosphogluconate; GAP, glyceraldehyde 3-phosphate; DHAP, dihydroxyacetone phosphate; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; S7P, sedoheptulose 7-phosphate; X5P, xylulose 5-phosphate.

Alignment between NADPH demand and oxPPP flux is also achieved via transcriptional regulation. One major demand is de novo lipogenesis (Fig. 2). OxPPP flux is much higher in lipogenic yeast than in baker’s yeast14. In mammals, expression of G6PD is high in most lipogenic tissues including the lactating mammary gland and brown adipose tissue (but not the liver, which makes NADPH in other ways)1517. Such activity is supported by a master transcription factor that promotes conversion of carbohydrates to fat: sterol regulatory element-binding protein (SREBP)1822. Another important NADPH demand is oxidative stress23. The nuclear respiratory factor 2 (NRF2) family of transcription factors activate many genes involved in responding to oxidative stress including multiple PPP enzymes (G6PD, 6PGD, TK and TALDO)24,25. Oxidative stress turns on NRF2 in part by suppression of its Kelch-like ECH-associated protein 1 (KEAP1)-mediated degradation by oxidation of KEAP1 cysteine residues26,27. ROS-induced DNA damage can also simulate G6PD expression through the combined activities of ataxia-telangiectasia mutated (ATM) serine/threonine kinase and heat shock protein 27 (HSP27)2831.

Rapid oxPPP responsiveness, on the timescale of seconds, is physiologically important for dealing with acute oxidative stress and to support oxidative burst for pathogen killing by phagocytes. Such responsiveness is enabled by reserve flux capacity, which is defined as excess enzyme relative to that required to catalyse basal pathway flux. Reserve PPP capacity has been seen in a wide variety of contexts from Escherichia coli to mammalian cells3,6,11,23,32,33. Basal activity in these cells is limited by low NADP and/or high NADPH. When NADP levels increase due to oxidative stress, the pathway nearly instantaneously turns on3,11,33.

Mechanisms that enhance the supply of glucose 6-phosphate can complement such NADP-mediated regulation. One way to enhance glucose 6-phosphate is to slow glycolysis. Oxidative stress reduces glycolytic flux by oxidation of the active site cysteine of the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase, which inactivates the enzyme within seconds of oxidative stress, or in response to endogeneous increases in oxidant levels, for instance, upon anchorage-independent growth of cancer cells23,3437. Also, cysteine oxidation of pyruvate kinase M2 (PKM2), the last enzyme in glycolysis, decreases PKM2 activity and has been reported to correlate with increased PPP flux38. In support of glycolysis and the PPP competing for glucose 6-phosphate, in bacteria, knocking out the gene encoding the upper glycolytic enzyme glucose-6-phosphate isomerase redirects carbon flux to the PPP39, and, in yeast, reducing the activity of triosephosphate isomerase (TPI) results in increased NADPH formation in the PPP and increased oxidant tolerance6. Furthermore, rendering GAPDH oxidation-resistant in cancer cells mitigates an increase in PPP flux (for instance, upon matrix detachment)37. In mammals, increased consumption of hexose phosphates by phosphofructokinase (PFK) driven by small-molecule PFK agonists can decrease NADPH levels in immune cells, while knockdown of PFK can increase NADPH40,41. Furthermore, the p53-induced phosphatase TIGAR, which supports intestinal oxidative defence and nucleotide synthesis42,43, may promote PPP flux by dephosphorylating the PFK activator fructose 2,6-bisphosphate and/or the downstream glycolytic metabolite 2,3-biphosphoglycerate44. Collectively, these data support the possibility that suppressed glycolysis is a physiological means of turning on the PPP in mammals.

Measuring the PPP

Intermediates of the PPP can now be routinely measured using metabolomic methods based on liquid chromatography–mass spectrometry, with proper choice of quenching solvent crucial for measuring NADP(H). For NADP and NADPH, enzyme assay kits are also available but may be less accurate45.

To measure PPP flux rather than metabolite levels, most methods use isotope-labelled glucose. A classic approach compares release of radioactive CO2 from [1-14C]glucose to [6-14C]glucose, with glucose carbon atoms 1 and 6 metabolically similar except for selective C1 release by the PPP46. This method can measure absolute PPP flux in cell culture and at the whole-body level by detecting expired 14CO247.

Stable isotope tracers offer greater biochemical and tissue specificity without concerns about radioactivity but typically measure relative flux (for example, the ratio of glycolytic to pentose-overflow flux) rather than absolute flux (for example, moles of glucose passing through oxPPP per unit time). The [1,2-13C]glucose isotope tracer distinguishes glycolytic intermediates directly made through glycolysis, which retain both labelled carbon atoms, versus from pentose overflow, in which C1 is lost48,49. It also measures the relative contribution of oxPPP versus non-oxPPP to ribose 5-phosphate and can be applied both in cell culture and in vivo50,51. As a relative measurement, results using this tracer method do not always mirror absolute fluxes; for example, increased glycolysis would misleadingly decrease the relative oxPPP flux. This deficiency can be addressed by making a complementary absolute measurement such as total glucose uptake52,53. OxPPP flux can also come from glycogen stored within cells rather than glucose directly, which can be monitored based on glucose-6-phosphate labelling54. Together, these carbon-tracing strategies provide valuable quantitative tools for probing oxPPP activity.

Deuterium-labelled glucose ([1-2H]glucose or [3-2H]glucose) provides a perspective complementary to carbon tracing by measuring the fraction of NADPH that is produced by the oxPPP, as opposed to by other enzymes such as IDH1, ME1 and aldehyde dehydrogenase 1 family member L1 (ALDH1L1) (refs. 17,55–59). A challenge with this method is that the active hydrogen on NADPH can undergo enzyme-catalysed exchange with water, which leads to loss of labelling, but it is feasible to account for such losses by measuring the exchange rate between NADPH and water60. Deuterium labelling can also be followed downstream into products such as fatty acid molecules synthesized by de novo lipogenesis, the labelling of which specifically reflects cytosolic NADPH and can be measured not only by mass spectrometry but also with subcellular spatial resolution by Raman spectroscopy58,61. The merit of these strategies is in revealing the contribution of the PPP to NADPH in different contexts.

To more directly visualize PPP activity in vivo, nuclear magnetic resonance imaging holds potential but currently lacks the sensitivity to detect the above tracers. To address this, investigators are exploring hyperpolarized [1-13C]gluconolactone62,63, which feeds into the middle of the oxPPP. Up to now, however, in vivo measurements are limited to initial uptake and phosphorylation of this tracer and do not yet probe the pathway’s pivotal redox or ribose-5-phosphate-producing steps62,63. We expect continued technological advances, including more effective in vivo tracing, to drive further understanding of the PPP.

Role in physiology and disease

The role of the PPP in redox regulation and biosynthesis makes it important in many contexts, including in development and differentiation6469. In this Review, we will focus on four settings in which the PPP plays a role relevant to human health and disease: (1) RBCs, (2) the immune system, (3) tissue maintenance, damage and repair, and (4) cancer.

RBCs

RBCs are the cell type most sensitive to oxPPP loss. Mature RBCs live for roughly 120 d without fresh protein synthesis under conditions of high oxygen exposure. Due to their lack of mitochondria, they have limited supplies of many metabolites, including tricarboxylic acid cycle (TCA) intermediates, which are required for NADPH production via the main alternative pathways IDH1 and ME1. Thus, the PPP plays an outsized role in RBC health.

Many mutations in PPP enzymes in humans result in haemolytic anaemia67. The G6PD gene on the X chromosome in humans has over 300 variants and 150 unique mutations that result in G6PD deficiency, the most common human enzyme defect70. The frequency of G6PD mutation geographically overlaps with areas where malaria has been historically endemic, and people who are heterozygous carriers of a G6PD mutation or homozygous deficient are protected from severe disease and mortality from malaria9,7174. G6PD deficiency can be asymptomatic until patients are exposed to oxidative stress from infection, certain drugs or fava beans causing haemolysis75. Mutations that impact protein stability cause G6PD deficiency in RBCs, especially as the red cells age, while other cell types maintain sufficient levels of the enzyme due to continued protein synthesis.

Mutations are broken down into classes based on clinical phenotype and enzyme activity in RBCs: class I mutations have less than 10% activity and result in chronic anaemia; class II mutations also have less than 10% activity and result in intermittent haemolytic anaemia; class III mutations have 10–60% activity with haemolytic anaemia only after oxidative stress; and class IV mutations have more than 60% enzymatic activity and are asymptomatic76 (Fig. 4). Consistent with G6pdx knockout being embryonic lethal in mice77, no null mutations have been observed clinically. In vitro studies have found that oxPPP activity is important for the emergence of erythroid progenitors from the haemogenic endothelium, which may contribute to the lethality of null mutations78.

Fig. 4 |. G6PD deficiency leads to RBC and immune dysfunction.

Fig. 4 |

The best-studied mutations in G6PD and their locations within the protein. Mutations are coloured according to their clinical phenotype from most to least severe: class I mutations in red, class II in purple, class III in blue and class IV in beige. The most severe class 1 mutations cluster around the glucose-6-phosphate (G6P) binding site, the dimer interface and the NADP+ structural site that is involved in allosteric activation and homotetramer formation.

G6PD activity is highest in the homotetramer form; it is active as a homodimer and inactive as a monomer79. Tetramers and dimers are also more stable80. Many of the class I mutations fall along the dimer interface and in the allosteric NADPH-binding site13,81,82. Class II and III mutations are more randomly distributed throughout the protein and impact stability and activity83 (Fig. 4). Mice with a common class II mutation, the Mediterranean mutation, have been generated, and they recapitulate the human phenotype84. Thus, across species, G6PD deficiency manifests in RBCs.

Immune system

The PPP plays a key role in the immune system, impacting immune cell fate and functions, such as cytokine production and the oxidative burst. Different types of immune cells have unique metabolic requirements. Phagocytic immune cells, which include macrophages and neutrophils, produce an oxidative burst for killing pathogens. T cells, on the other hand, require a large amount of biosynthesis and energy for rapid proliferation and cytokine production upon activation. This leads to different roles of the PPP in different cell types.

Both intracellular signalling and killing of pathogens during phagocyte inflammatory responses involve the production of superoxide and nitric oxide by the enzymes NADPH oxidase (NOX) and inducible NOS (iNOS), respectively. Both NOX and NOS enzymes use NADPH, with the PPP as a major source. Due to defective pathogen killing in phagocytes, deficiency of NOX causes chronic granulomatous disease, which is characterized by serious recurrent bacterial and fungal infections85. Severe G6PD mutations phenocopy chronic granulomatous disease and put patients at risk for sepsis8688.

Macrophages.

Macrophages are large mononuclear phagocytic cells that play an important role in fighting infection, maintaining tissue homoeostasis and removing foreign substances. They can be classified along a spectrum from classically activated pro-inflammatory macrophages (‘M1’ like) to alternatively activated macrophages that support tissue repair and wound healing (‘M2’ like)8991.

In response to a classical activation signal such as lipopolysaccharide, the polarization of naive macrophages into a pro-inflammatory state requires coupled metabolic remodelling92,93. Within 1 h of activation, glucose uptake and glycolytic flux increase. The contribution of glucose to PPP metabolites via the oxidative branch also increases, as determined by [1,2-13C]glucose tracing94. This increased flux supports NADPH production for NOX and iNOS activity (Fig. 5a). Perturbing oxPPP activity modulates ROS and RNS production11,95,96. In some studies but not others, perturbation of the oxPPP also had broader impacts such as changes in expression of iNOS or cytokines such as interleukin (IL)-6 (refs. 96–98). The extent to which these broader pro-inflammatory phenotypes depend on ROS, RNS or the changes in redox state is not well understood.

Fig. 5 |. The role of oxPPP-produced NADPH in phagocyte function.

Fig. 5 |

a, In phagocytic cell types including macrophages and neutrophils, NADPH production by the oxPPP supports production of superoxide by NOX and nitric oxide by NOS for killing pathogens in the phagosomes and extracellular space. Ru5P, ribulose 5-phosphate. b, In macrophages involved in haem clearance, NADPH supports the breakdown of haem into biliverdin by HMOX with the help of p450 oxoreductase (POR) and biliverdin into bilirubin by biliverdin reductase (BVR).

Macrophages are also important for haem clearance. Haem-loaded macrophages require the PPP for haem detoxification, probably to provide NADPH as a cofactor for haem oxygenase (HMOX)1, which metabolizes haem to biliverdin and carbon monoxide, and biliverdin reductase, which metabolizes biliverdin to bilirubin (Fig. 5b). Haem loading of macrophages induces glucose uptake and increases oxPPP enzyme activity. This oxPPP increase can be phenocopied by treating cells with the carbon monoxide-producing molecule tricarbonylchloro(glycinatro)ruthenium, with release of carbon monoxide by HMOX1 seeming to promote PPP activity, although the biochemical mechanism remains unknown99.

In contrast to the upregulation of glycolysis upon M1 polarization, M2 polarization favours respiratory ATP generation. In addition, M2 polarization by IL-4 upregulates a recently discovered mammalian enzyme, sedoheptulose kinase (SHPK). SHPK, also known as CARLK, converts sedoheptulose, a seven-carbon sugar that is found in various fruits and vegetables, to sedoheptulose 7-phosphate, a non-oxPPP metabolite100,101. Via an unclear mechanism, SHPK overexpression suppresses the M1 phenotype and glycolytic metabolism while promoting M2 polarization94.

The importance of metabolism in macrophage function raises the question of how common metabolic diseases such as obesity and diabetes may affect macrophages. Hypercholesterolaemia suppresses macrophages’ NRF2 antioxidant response, decreasing 6PGD expression. Thus, hypercholesterolaemia may lower oxPPP activity, impairing the macrophage immune response102. Such interactions may be medically important, given the central role of macrophages in atherosclerotic plaques and the health toll of atherosclerosis103.

Neutrophils.

Neutrophils are abundant, short-lived, mobile phagocytic granulocytes that play an important role in fighting bacterial and fungal infections. As part of their inflammatory response, neutrophils, like macrophages, produce an oxidative burst, releasing large quantities of superoxide anion via NOX, which requires NADPH for its activity. Patients with G6PD deficiency have less ROS release from neutrophils104. Pharmacological G6PD inhibition did not alter neutrophil cytokine production but impaired the oxidative burst98.

During NOX-dependent oxidative burst, which can occur within minutes of stimulation, neutrophils shunt most of their glucose flux toward oxPPP. Remarkably, to maximize NADPH yield from glucose 6-phosphate, flux through the glycolytic enzyme glucose-6-phosphate isomerase is reversed to enable pentose cycling. The main molecular trigger of these events appears to be NADPH oxidation by NOX, which stimulates oxPPP flux11. Similar to directly inhibiting the oxPPP, indirectly disrupting pentose cycling by either knocking out the genes encoding the non-oxPPP enzymes TK or TALDO or pharmacologically activating the glycolytic enzyme PFK suppresses oxidative burst in neutrophils11,40. Pharmacologic inhibition of the neutrophil oxidative burst by PFK activators presents a potential therapeutic strategy for inflammatory diseases.

In addition to release of superoxide, neutrophils can also release chromatin traps to capture and kill microorganisms (‘neutrophil extracellular traps’ (NETs)). Superoxide production triggers NET release. NOX is a main source of NET-inducing superoxide. NADPH from the oxPPP fuels NOX. Consistent with this pathway, inhibiting oxPPP suppresses ROS-dependent NET release11,105. In severe coronavirus disease 2019 (COVID-19) infection, decreased neutrophil glyceraldehyde-3-phosphate dehydrogenase activity and increased oxPPP flux may contribute to aberrantly increased NET release and pathogenic inflammation106.

The importance of oxPPP-derived NADPH during the oxidative burst and NET release is now becoming clear. These connections raise the question of whether diet or other upstream steps impacting glucose metabolism can affect neutrophil function107. Diabetes, a disease that causes high levels of circulating glucose, impairs wound healing. While neutrophils play an important role in enabling wound healing, overactivity of neutrophil NETs can also result in tissue damage. Neutrophils from patients with either type 1 diabetes or type 2 diabetes are primed for NET release, which probably contributes to impaired wound healing108. These observations lead to the intriguing hypothesis that increased glucose levels may increase PPP flux and cause the observed phenotype. It will be interesting to determine whether low-carbohydrate or ketogenic diets can prevent unwanted inflammatory responses such as impaired wound healing, gout or infection-induced cytokine storms and whether the PPP may be one pathway moderating these effects.

T cells.

T cells exist in a variety of subpopulations with different immune functions. One fundamental division is between cytotoxic CD8+ T cells that kill pathogen-infected or malignant cells and helper CD4+ T cells. Either can exist in an immunosuppressive regulatory T cell (Treg) form, in addition to the traditional invader-fighting form. Upon antigen stimulation, all these T cell types switch from a quiescent to a highly proliferative state. This switch requires corresponding metabolic changes, including upregulation of both glycolysis and mitochondrial functions109111. Similar to immunosuppressive macrophages, anti-inflammatory Treg cells rely more on fatty acid oxidation than their infection-fighting counterparts112114.

Similar to macrophages and neutrophils, cytotoxic and helper T cells both increase oxPPP flux in response to activation98,115. ROS levels are increased in T cells with G6PD inhibition98, and glucose restriction in CD8+ T cells is sufficient to cause a decrease in the glutathione/glutathione disulfide ratio. These observations suggest a major role for oxPPP in T cell antioxidant defence116.

Consistent with these findings, the oxPPP is functionally important in T cells. G6PD inhibition during T cell activation does not block proliferation but leads to decreased production of pro-inflammatory cytokines including IFNγ99. T cells from patients with rheumatoid arthritis are hyperproliferative and manifest increased G6PD expression and decreased levels of ROS, consistent with upregulation of oxPPP flux. Such flux may facilitate ongoing proliferation and promote differentiation into pro-inflammatory type 1 and 17 helper T cells that cause excessive inflammation in rheumatoid arthritis117,118. Thus, the oxPPP plays an important role in infection-fighting and cancer-killing CD4+ and CD8+ T cells as well as in T cells driving autoimmunity.

The functional importance of the oxPPP carries over into Treg cells. Such cells express the transcription factor FOXP3, which is required for their immunosuppressive function. The FOXP3 transcriptional programme inhibits glycolytic and oxPPP flux, measured by [1-14C]glucose, and promotes oxidative metabolism113. While this might suggest limited importance of the oxPPP in Treg cells, knockout of Pgd specifically in Treg cells (driven by Foxp3-Cre) causes severe Treg dysfunction in mice, which die because of autoimmunity between days 20 and 30 of life119. Similarly, Treg-specific knockout of the gene encoding an enzyme from non-oxPPP, TK, also causes lethal autoimmunity120. Moreover, knockout of Pgd across all CD4+ and CD8+ T cells promoted antitumour immunity (rather than suppressed it, as one might expect if the defect was primarily in pro-inflammatory T cells). Data from this pan-T cell knockout raise the possibility that the in vivo effect of oxPPP loss is mediated primarily through its effect on Treg cells and outweighs the effects of its loss in pro-inflammatory T cells121.

Currently, interpretation of these results is complicated by different oxPPP enzymes being modulated and different aspects of immunity being probed. For example, severe systemic G6PD deficiency predisposes to bacterial and fungal infections, while Pgd knockout in T cells putatively promotes antitumour immunity121. Is the difference due to G6PD’s role also in phagocytes? Is it because G6PD is pro-immune and 6PDG is immunosuppressive, even though they are both oxPPP enzymes? Resolving these questions is pressing because the PPP could potentially be an appealing target for new immunosuppressive or immuno-oncology agents.

Tissue maintenance, damage and repair

Tissue maintenance and repair involve both redox balance and biosynthesis. The PPP probably plays a role in the homoeostasis of many vital organs, including heart, kidney, brain, skin and intestine67,122127. Here, we focus on the latter two as well as ischaemia–reperfusion injury, a medically important tissue-damage process with a large redox component.

Skin.

As the body’s primary barrier, the skin is exposed to damaging agents such as chemicals and UV radiation128. These can cause oxidative stress, the control of which depends in part on the PPP. Some patients with G6PD deficiency can have haemolysis with exposure to normally tolerable chemicals such as those found in the plant-derived dye henna129, probably due to oxidant chemicals interacting with RBCs in skin capillaries. But the role of the PPP in skin extends beyond its vasculature.

G6PD levels in the skin decrease in psoriasis and vitiligo130132. Treatment of psoriasis with 6-aminonicotinamide, which inhibits the 6PGD reaction in the oxPPP, decreased itching and cleared some lesions133. While these effects may be through interactions with immune cells within the skin microenvironment, the PPP also seems to play a role in skin fibroblasts and keratinocytes. Kinetic 13C-glucose experiments revealed that PPP flux increases in fibroblasts and keratinocytes in response to oxidative stress from hydrogen peroxide or UV exposure. This flux was dependent on non-oxPPP enzymes; thus, oxidative stress activates pentose overflow in skin128. The importance of the PPP in the many cell types within the skin may be due to the hazardous nature of this microenvironment.

Intestine.

The PPP can play an important role in homoeostasis of proliferative tissues due to the requirement of nucleotides for cell replication. The intestine is one of the most replicative, high-turnover tissues in the body. The PPP is upregulated during intestinal regeneration. Whole-body Tigar knockout, which may decrease PPP flux, causes no obvious developmental problems but leads to reduction in intestinal crypt regeneration43,134. Crypt formation could be rescued by either antioxidants or nucleotides, suggesting that knockout cells lacked both NADPH and ribose 5-phosphate from the PPP43.

Inflammatory bowel disease involves chronic autoimmune inflammation of the gastrointestinal tract, such as colitis. In mice, knockout of the gene encoding the non-oxPPP enzyme TK in intestinal epithelial cells results in spontaneous colitis with massive immune cell infiltration. Intestinal epithelial cells lacking TK are hyperproliferative and prone to apoptosis. These phenotypic changes occur without alteration of the NADP+/NADPH ratio but with accumulation of the terminal oxPPP product (and non-oxPPP substrate) ribose 5-phosphate as well with decreased levels of glycolytic metabolites, possibly due to impaired pentose overflow135. Knockout of Tigar in intestinal epithelial cells also confers greater sensitivity to colitis43. Thus, both arms of the PPP and their putative regulator TIGAR are important for intestinal health.

Ischaemia–reperfusion injury.

Ischaemia occurs when blood flow and thus oxygen supply to a tissue is compromised. If blood flow is then restored, exposure to oxygen during reperfusion causes a burst of ROS production that damages the tissue. Such damage can be further exacerbated by neutrophil activation at the injury site.

Ischaemia–reperfusion injury is a major clinical problem, contributing to the pathogenesis of both heart attack and stroke. In mouse models of ischaemic brain injury, G6PD overexpression decreases oxidative damage and improves survival and recovery136. As prophylactic G6PD overexpression is not clinically feasible, scientists have searched for ways to drive such expression. One strategy involves ‘post-conditioning’ after stroke with bouts of mild hypoxia. Such post-conditioning increased G6PD activity, NADPH and glutathione levels and neuronal cell survival137.

The oxPPP appears to suppress ischaemia–reperfusion injury also in the heart, as knockout of G6pd results in impaired cardiac relaxation and contractile performance after ischaemia–reperfusion injury in mice138. Glucose flux was also seen to increase through both glycolysis and the PPP in mouse hearts during low-flow ischaemia139. Thus, oxPPP flux protects against ischaemia–reperfusion injury.

Cancer

A key role for the PPP in cancer cells has long been postulated due to the proliferative nature of cancer and the biosynthetic role of the PPP. Epidemiologic evidence suggests that patients with G6PD mutations are protected from certain types of cancers, including endometrial and colon cancers, but not others140,141. This complexity parallels the dual roles of oxidative stress in cancer. ROS can cause DNA damage and thus cancer. After transformation, however, ROS can suppress both tumour growth and metastasis142,143. In addition, the PPP can support biosynthesis and proliferation through both NADPH and ribose-5-phosphate production.

To more specifically assess the oxPPP’s role in cancer, recent studies have turned to genetics, finding that the oxPPP (specifically G6PD) can, remarkably, transform cells when overexpressed but nevertheless is not essential for tumour growth144,145. Upon oxPPP loss, other cytosolic NADPH-producing enzymes, ME1 and IDH1, increase NADPH production to compensate. These alternative routes, however, are insufficient to maintain as high an NADPH/NADP ratio as the oxPPP, leading to downstream consequences including altered folate redox status due to impairment of dihydrofolate reductase activity by NADP57. Thus, among various NADPH-production routes, the PPP particularly effectively supports nucleotide synthesis in proliferative cells such cancer cells57.

Alternative NADPH-production routes to the oxPPP also probably lack the dynamic flexibility to turn up NADPH production quickly. As a result, cancer cells lacking the oxPPP may fare poorly during bouts of high oxidative stress such as during metastasis146,147. Levels of PPP enzymes and metabolites are increased at metastatic sites relative to primary tumours148,149. In mouse models of melanoma metastasis, G6PD deficiency decreases circulating melanoma cells and the metastatic burden149. Efficient metastasis is also associated with higher levels of the lactate transporter MCT1. For reasons that remain unclear, MCT1 inhibition led in melanoma xenografts to decreased oxPPP flux (relative to glycolysis) and increased ROS levels50. Furthermore, the rapid PPP activation upon anchorage independent growth, which involves the production of increased levels of oxidant, might support metastasis formation37. Combined, these studies suggest that inhibition of the oxPPP may be effective in decreasing metastasis.

Oncogenic context.

The functional importance of the oxPPP is increased in tumours that upregulate the master antioxidant defence transcription factor NRF2. Such upregulation most frequently occurs via loss-of-function mutations in KEAP1, the E3 ubiquitin ligase that degrades NRF2. KEAP1 mutations occur in about 20% of non-small-cell lung carcinoma, where they are associated with poor outcomes including immunotherapy resistance150153. NRF2 induces expression of antioxidant genes including those encoding PPP enzymes26,154. In a CRISPR screen of metabolic enzymes, G6PD stood out for being selectively essential in KEAP1-mutant cancer cells26 (Fig. 6a). In agreement with the screen data, G6PD knockdown suppressed preferentially KEAP1-mutant lung tumour growth. Thus, the oxPPP is a validated target in a type of lung cancer with high unmet need.

Fig. 6 |. Oncogenic contexts for targeting the PPP.

Fig. 6 |

a, KEAP1 mutations lead to stabilization of NRF2, which promotes transcription of PPP genes and leads to dependency on their enzyme activity. Ub, ubiquitin. b, Mutations in IDH1 convert IDH from an NADPH producer into a consumer. Therefore, the oxPPP becomes a more important source of NADPH with mutant IDH.

There are additional contexts in which inhibiting the PPP may be advantageous. In acute myeloid leukaemia cell lines harbouring mutations in the receptor tyrosine kinase FLT3, knockdown of G6PD was observed to overcome resistance to FLT3 inhibitors155. More generally, certain oncogenes increase demand for cytosolic NADPH and therefore would logically be expected to augment oxPPP dependency. For example, phosphoinositide-3-kinase (PI3K)–Akt signalling can increase NRF2 nuclear availability, promoting increased PPP enzyme expression25. Similarly, oncogenic mutations in IDH1 (mtIDH1) change the protein from being an NADPH producer to an NADPH consumer that makes the oncometabolite 2-hydroxyglutarate. This NADPH stress suppresses the rate of lipogenesis and increases sensitivity to oxidative stress in mtIDH1 cells in culture156, despite mtIDH1 cells exhibiting increased oxPPP flux157. Experiments are needed to assess whether these flux changes lead to a functional dependence on the oxPPP (Fig. 6b).

Another logical way to inhibit tumour growth is to induce ribose limitation by inhibiting the PPP. The oncogene BCR-ABL1, which is a fusion of two genes (breakpoint cluster region (BCR) and abelson 1 (ABL1)), can decrease oxPPP flux, leading to dependence on non-oxPPP for ribose-5-phosphate synthesis. A thiamine analogue, oxythiamine, which inhibits the thiamine-dependent enzyme TK, is effective in mouse models of BCR-ABL1-driven cancer158. Oxythiamine has also been seen to be effective in multiple types of metastatic cancers. Further work is needed to determine whether this activity occurs via ribose limitation or redox effects159161.

Combination therapies.

Cancer treatments, including radiation, chemotherapy and immunotherapy, have the potential to cause oxidative stress. Chemotherapeutic agents have a wide variety of mechanisms of action and therefore generate various levels of ROS. Anthracyclines, which include doxorubicin and daunorubicin, generate high ROS levels162. Increased G6PD activity is observed in cancer cells exposed to doxorubicin, and knockdown of G6PD kills doxorubicin-resistant cells163,164. Alkylating agents and platinum coordination complexes, including cisplatin and oxaliplatin, also cause ROS production165. Decreased PPP activity, induced by a variety of indirect methods, enhances killing of colorectal cancer cells by platinum agents166169.

Radiotherapy causes the production of ROS and RNS. There is evidence that levels of PPP intermediates increase in response to radiation in culture170. But, despite many papers suggesting that radiotherapy should be used in combination with PPP inhibition, experimental evidence is missing. Experimental testing of the safety and efficacy of oxPPP inhibition in combination with radiotherapy is merited.

In addition to chemotherapy and radiotherapy, targeted metabolic inhibitors may synergize with oxPPP blockade. Logical synthetic lethal targets include the non-oxPPP to cut off its contribution to ribose supply during pentose insufficiency or IDH1 and ME1, the enzymes that compensate for loss of NADPH produced by oxPPP. IDH1 inhibitors were developed as part of the process of targeting mutant IDH1, with the Food and Drug Administration-approved mutant IDH1 inhibitor ivosidenib having activity also against the wild-type enzyme. Ivosidenib has more potent cytotoxic effects under conditions of low glucose, consistent with cytotoxic activity occurring when IDH is blocked and the PPP is limited by substrate availability. In mouse models of pancreatic cancer, ivosidenib prevented tumour growth, and this was reversed by oral glucose supplementation171.

Unfortunately, quality ME1 inhibitors for use in vivo are not yet available. But increased ME1 flux can be targeted indirectly. ME1 activation consumes TCA cycle four-carbon units. In both KEAP1-mutant tumours and in some metastatic melanomas, loss of G6PD led to depletion of TCA cycle intermediates. A major means of generating TCA cycle four-carbon units is glutamine catabolism, and its importance increases with G6PD inhibition. This leads to increased sensitivity to the glutaminase inhibitor CB-839, with the combination of G6PD loss and glutaminase inhibitor being synergistic149,172. Thus, combination regimens incorporating agents targeting the PPP hold potential as cancer treatments.

Perspective and future directions

Current perspective

Despite alternative NADPH-production routes, the PPP plays a key role in redox metabolism in a variety of biological contexts. This reflects its often being the fastest NADPH producer, with the greatest acute responsiveness to increased NADPH demand and with the capacity to generate the highest NADPH/NADP ratio57. In addition to supporting biosynthesis, NADPH produced in the oxPPP can be either antioxidant or pro-oxidant. NADPH’s antioxidant function involves the reduction and removal of ROS, RNS and their downstream products, such as oxidized and nitrosylated thiols in proteins173. Its pro-oxidant role involves purposeful production of ROS and RNS for signalling, antimicrobial killing and other purposes174178. This dual role of the PPP and NADPH mirrors the duelling biological impact of free radicals. For instance, ROS at lower doses stimulates cancer cell migration but at higher doses is inhibitory179. ROS can promote tumorigenesis by increasing genetic instability during cancer initiation, but, after the tumour is established, ROS can limit cancer cell survival and growth.

To make sense of the PPP and of redox metabolism more generally, it is key to move from generalizations (for example, ROS cause cancer) to time-dependent, location-dependent and molecule-specific roles. For example, soon after classical activation, macrophages produce large amounts of ROS powered by the oxPPP and NOX. This ROS is largely directed to the phagosome and extracellular space. This localization promotes pathogen killing and limits host oxidative stress. After the initial response resolves, repair of oxidatively damaged cellular components also requires NADPH. Accordingly, manipulating PPP at different times may have vastly different effects. Thus, to develop therapies, spatial and temporal understanding will be critical.

Moving forward

How can we achieve spatial and temporal understanding? Progress rests on three pillars: genetics, pharmacology and analytics. Genetics and pharmacology enable PPP manipulation (Box 1). Genetic methods for cell type-specific enzyme manipulation in mice are readily available. Their systematic application to the PPP, for example, to manipulate G6PD and 6PGD in T cells, should be highly informative, especially as initial data suggest that these oxPPP enzymes may sometimes have opposite immunological functions. Genetics can also help to inform the roles of non-oxPPP reactions, including newly discovered orphan reactions connected to non-oxPPP such as sedoheptulose kinase.

BOX 1. Manipulating the PPP.

Both genetics and pharmacology can be used to manipulate the PPP. The development of CRISPR–Cas9 has enabled easy knockout of genes encoding various PPP enzymes in cultured cells and a faster way to make genetically engineered mouse models. In addition to targeting enzymes in the PPP, NADPH levels can be genetically manipulated through an engineered enzyme called triphosphopyridine nucleotide oxidase (TPNOX)189, which converts NADPH and oxygen to NADP and water. NADPH can similarly be depleted by a small molecule, phenazine methosulfate. TPNOX and phenazine methosulfate are valuable tools for determining the importance of NADPH in regulating PPP flux.

Several inhibitors are also available to modulate the oxPPP. Dehydroepiandrosterone has been used extensively to target G6PD, but its cellular effects may be largely off target98,190. The molecule 6-aminonicotinamide broadly targets NAD(P) enzymes, with strong engagement of 6PDG in the oxPPP. Despite its lack of specificity, it is a useful tool. A new G6PD inhibitor (G6PDi1) with effective cellular target engagement has recently been developed98. A small-molecule activator (AG1) has also been identified191,192. Development of improved tool compounds, including ones with better specificity and with pharmacokinetics enabling in vivo application, will be valuable in advancing our understanding of the pathway.

Pharmacology is an important complement to genetics, due to its capacity for partial enzyme inhibition, dynamic target engagement and eventual clinical translation. Given the shifting roles of the PPP during disease processes, timed pharmacological interventions could be particularly informative, but unfortunately many PPP enzymes continue to lack specific inhibitors, and existing inhibitors require further optimization to enable routine in vivo application. Better compounds are needed.

The ability to make dynamic and spatially resolved in vivo measurements is also critical. Historically, such measurements of metabolism have been limited, but capabilities are rapidly advancing. The major intermediates of the PPP as well as NADP and NADPH can now be routinely measured from tissue extracts by liquid chromatography–mass spectrometry. Excitingly, imaging mass spectrometry, in which a tissue slice is coated with matrix and then ionized by a laser pulse (MALDI), offers the prospect of visualizing them at single-cell resolution180,181, although proof of principle for PPP metabolites remains to be established. Other technologies, such as water gas cluster ion beam secondary ion mass spectrometry hold potential for subcellular resolution but remain farther from widespread application181183. Mass spectrometry approaches can also be coupled to isotope tracing to gain insights into flux, for example, using [1,2-13C]glucose and searching by imaging mass spectrometry for glycolytic intermediates with one 13C atom to capture oxPPP overflow flux at the single-cell level.

Fluorescent methods are an important complement to mass spectrometry. Fluorescent sensors of NADP+ and NADPH can be used to study the PPP at the subcellular level184186, with potential also for dynamic intravital analyses184,187. Sensors also exist for glucose but not yet for any dedicated PPP metabolite188. The development of such sensors would be valuable. Better measurements of specific ROS and RNS species and their covalent modifications of lipids and macromolecules is another important frontier. Ultimately, systematic application of genetics combined with improved pharmacological and measurement tools will help drive understanding of the PPP temporally and spatially. In so doing, it will also hopefully open the doors to new diagnostics and therapeutics.

Acknowledgements

This work was funded by NIH R01CA163591, NIH DP1DK113643, Ludwig Cancer Research and Stand Up to Cancer SU2CAACR-DT-20-16 to J.D.R; NIH R35GM147014 to J.F.

Footnotes

Competing interests

J.D.R. is an advisor and stockholder in Colorado Research Partners, LEAF Pharmaceuticals, Empress Therapeutics and Bantam Pharmaceutical; a paid consultant of Pfizer and Third Rock Ventures; a founder, director and stockholder of Farber Partners, Raze Therapeutics and Sofro Pharmaceuticals; a cofounder and stockholder in Marea Therapeutics; and a director of the Princeton University–PKU Shenzhen collaboration. All other authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Nissim Hay and the other, anonymous, reviewer for their contribution to the peer review of this work. Primary Handling Editor: Alfredo Giménez-Cassina, in collaboration with the Nature Metabolism team.

References

  • 1.Hostetler KY & Landau BR Estimation of the pentose cycle contribution to glucose metabolism in tissue in vivo. Biochemistry 6, 2961–2964 (1967). [DOI] [PubMed] [Google Scholar]
  • 2.Park JO et al. Metabolite concentrations, fluxes and free energies imply efficient enzyme usage. Nat. Chem. Biol 12, 482–489 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Christodoulou D et al. Reserve flux capacity in the pentose phosphate pathway by NADPH binding is conserved across kingdoms. iScience 19, 1133–1144 (2019). This study revealed that the reserve capacity of the PPP is evolutionarily conserved.
  • 4.Masi A, Mach RL & Mach-Aigner AR The pentose phosphate pathway in industrially relevant fungi: crucial insights for bioprocessing. Appl. Microbiol. Biotechnol 105, 4017–4031 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Liguori I et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 13, 757–772 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Ralser M et al. Dynamic rerouting of the carbohydrate flux is key to counteracting oxidative stress. J. Biol 6, 10 (2007). This paper demonstrated that decreasing glycolytic flux can cause carbons to be redirected through the PPP.
  • 7.Winterbourn CC, Kettle AJ & Hampton MB Reactive oxygen species and neutrophil function. Annu. Rev. Biochem 85, 765–792 (2016). [DOI] [PubMed] [Google Scholar]
  • 8.Sies H & Jones DP Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol 21, 363–383 (2020). [DOI] [PubMed] [Google Scholar]
  • 9.Luzzatto L, Ally M & Notaro R Glucose-6-phosphate dehydrogenase deficiency. Blood 136, 1225–1240 (2020). [DOI] [PubMed] [Google Scholar]
  • 10.Lemons JMS et al. Quiescent fibroblasts exhibit high metabolic activity. PLoS Biol 8, e1000514 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Britt EC et al. Switching to the cyclic pentose phosphate pathway powers the oxidative burst in activated neutrophils. Nat. Metab 4, 389–403 (2022). This paper revealed that partial pentose cycling enables neutrophil oxidative burst by increasing NADPH production.
  • 12.Wrigley NG, Heather JV, Bonsignore A & De Flora A Human erythrocyte glucose 6-phosphate dehydrogenase: electron microscope studies on structure and interconversion of tetramers, dimers and monomers. J. Mol. Biol 68, 483–499 (1972). [DOI] [PubMed] [Google Scholar]
  • 13.Garcia AA et al. Stabilization of glucose-6-phosphate dehydrogenase oligomers enhances catalytic activity and stability of clinical variants. J. Biol. Chem 298, 101610 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wasylenko TM, Ahn WS & Stephanopoulos G The oxidative pentose phosphate pathway is the primary source of NADPH for lipid overproduction from glucose in Yarrowia lipolytica. Metab. Eng 30, 27–39 (2015). [DOI] [PubMed] [Google Scholar]
  • 15.Cabezas H, Raposo RR & Meléndez-Hevia E Activity and metabolic roles of the pentose phosphate cycle in several rat tissues. Mol. Cell. Biochem 201, 57–63 (1999). [DOI] [PubMed] [Google Scholar]
  • 16.Jung SM et al. In vivo isotope tracing reveals the versatility of glucose as a brown adipose tissue substrate. Cell Rep 36, 109459 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang Z et al. Serine catabolism generates liver NADPH and supports hepatic lipogenesis. Nat. Metab 3, 1608–1620 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Amemiya-Kudo M et al. Transcriptional activities of nuclear SREBP-1a, -1c, and -2 to different target promoters of lipogenic and cholesterogenic genes. J. Lipid Res 43, 1220–1235 (2002). [PubMed] [Google Scholar]
  • 19.Dentin R, Girard J & Postic C Carbohydrate responsive element binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c): two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie 87, 81–86 (2005). [DOI] [PubMed] [Google Scholar]
  • 20.Tsutomu K, Takumi K, Wadzinski BE & Kosaku U Xylulose 5-phosphate mediates glucose-induced lipogenesis by xylulose 5-phosphate-activated protein phosphatase in rat liver. Proc. Natl Acad. Sci. USA 100, 5107–5112 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Doiron B, Cuif M-H, Chen R & Kahn A Transcriptional glucose signaling through the glucose response element is mediated by the pentose phosphate pathway. J. Biol. Chem 271, 5321–5324 (1996). [DOI] [PubMed] [Google Scholar]
  • 22.Sanchez-Gurmaches J et al. Brown fat AKT2 is a cold-induced kinase that stimulates ChREBP-mediated de novo lipogenesis to optimize fuel storage and thermogenesis. Cell Metab 27, 195–209 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ralser M et al. Metabolic reconfiguration precedes transcriptional regulation in the antioxidant response. Nat. Biotechnol 27, 604–605 (2009). [DOI] [PubMed] [Google Scholar]
  • 24.Kitamura H & Motohashi H NRF2 addiction in cancer cells. Cancer Sci 109, 900–911 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Mitsuishi Y et al. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 22, 66–79 (2012). This paper showed that enzymes in the PPP can be transcriptionally activated by NRF2.
  • 26.Zhao D et al. Combinatorial CRISPR–Cas9 metabolic screens reveal critical redox control points dependent on the KEAP1–NRF2 regulatory axis. Mol. Cell 69, 699–708 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ma Q Role of Nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol 53, 401–426 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yan L-J et al. Mouse heat shock transcription factor 1 deficiency alters cardiac redox homeostasis and increases mitochondrial oxidative damage. EMBO J 21, 5164–5172 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cosentino C, Grieco D & Costanzo V ATM activates the pentose phosphate pathway promoting anti-oxidant defence and DNA repair. EMBO J 30, 546–555 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Préville X et al. 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 247, 61–78 (1999). [DOI] [PubMed] [Google Scholar]
  • 31.Yichong Z et al. Mitochondrial redox sensing by the kinase ATM maintains cellular antioxidant capacity. Sci. Signal 11, eaaq0702 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Christodoulou D et al. Reserve flux capacity in the pentose phosphate pathway enables Escherichia coli’s rapid response to oxidative stress. Cell Syst 6, 569–578 (2018). [DOI] [PubMed] [Google Scholar]
  • 33.Yoshida A & Lin M Regulation of glucose-6-phosphate dehydrogenase activity in red blood cells from hemolytic and nonhemolytic variant subjects. Blood 41, 877–891 (1973). [PubMed] [Google Scholar]
  • 34.Janero DR, Hreniuk D & Sharif HM Hydroperoxide-induced oxidative stress impairs heart muscle cell carbohydrate metabolism. Am. J. Physiol 266, C179–C188 (1994). [DOI] [PubMed] [Google Scholar]
  • 35.Eaton P, Wright N, Hearse DJ & Shattock MJ Glyceraldehyde phosphate dehydrogenase oxidation during cardiac ischemia and reperfusion. J. Mol. Cell. Cardiol 34, 1549–1560 (2002). [DOI] [PubMed] [Google Scholar]
  • 36.Peralta D et al. A proton relay enhances H2O2 sensitivity of GAPDH to facilitate metabolic adaptation. Nat. Chem. Biol 11, 156–163 (2015). [DOI] [PubMed] [Google Scholar]
  • 37. Talwar D et al. The GAPDH redox switch safeguards reductive capacity and enables survival of stressed tumour cells. Nat. Metab 5, 660–676 (2023). This paper demonstrated in mammalian cells that glucose carbon flux can be redirected from glycolysis into the PPP during oxidative stress by oxidizing GADPH.
  • 38.Anastasiou D et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334, 1278–1283 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Long CP, Gonzalez JE, Feist AM, Palsson BO & Antoniewicz MR Dissecting the genetic and metabolic mechanisms of adaptation to the knockout of a major metabolic enzyme in Escherichia coli. Proc. Natl Acad. Sci. USA 115, 222–227 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Amara N et al. Selective activation of PFKL suppresses the phagocytic oxidative burst. Cell 184, 4480–4494 (2021). This paper demonstrated that a small-molecule activator of glycolysis can limit inflammation through inhibition of the neutrophil oxidative burst.
  • 41.Graham DB et al. Functional genomics identifies negative regulatory nodes controlling phagocyte oxidative burst. Nat. Commun 6, 7838 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bensaad K et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006). [DOI] [PubMed] [Google Scholar]
  • 43.Cheung EC et al. TIGAR is required for efficient intestinal regeneration and tumorigenesis. Dev. Cell 25, 463–477 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gerin I et al. Identification of TP53-induced glycolysis and apoptosis regulator (TIGAR) as the phosphoglycolate-independent 2,3-bisphosphoglycerate phosphatase. Biochem. J 458, 439–448 (2014). [DOI] [PubMed] [Google Scholar]
  • 45.Lu W, Wang L, Chen L, Hui S & Rabinowitz JD Extraction and quantitation of nicotinamide adenine dinucleotide redox cofactors. Antioxid. Redox Signal 28, 167–179 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Katz J & Wood HG The use of C14O2 yields from glucose-1- and -6-C14 for the evaluation of the pathways of glucose metabolism. J. Biol. Chem 238, 517–523 (1963). This paper established use of [1-14C]glucose and [6–14C]glucose in combination for quantification of oxPPP flux.
  • 47.Burki HR & Okita GT Effect of mammary tumor virus infection on in vivo oxidation of glucose-1-14C and glucose-6-14C in C3H mice. Cancer Res 31, 1955–1961 (1971). [PubMed] [Google Scholar]
  • 48. Lee WNP et al. Mass isotopomer study of the nonoxidative pathways of the pentose cycle with [1,2-13C2]glucose. Am. J. Physiol 274, E843–E851 (1998). This paper established use of the [1,2-13C]glucose tracer for measurement of relative use of glucose in the PPP versus glycolysis.
  • 49.Metallo CM, Walther JL & Stephanopoulos G Evaluation of 13C isotopic tracers for metabolic flux analysis in mammalian cells. J. Biotechnol 144, 167–174 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tasdogan A et al. Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ghergurovich JM et al. Local production of lactate, ribose phosphate, and amino acids by human triple-negative breast cancer. Med 2, 736–754 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bartman CR et al. Slow TCA flux and ATP production in primary solid tumours but not metastases. Nature 614, 349–357 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.TeSlaa T & Teitell MA Techniques to monitor glycolysis. Methods Enzymol 542, 91–114 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.TeSlaa T et al. The source of glycolytic intermediates in mammalian tissues. Cell Metab 33, 367–378 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ben‐Yoseph O, Kingsley PB & Ross BD Metabolic loss of deuterium from isotopically labeled glucose. Magn. Reson. Med 32, 405–409 (1994). [DOI] [PubMed] [Google Scholar]
  • 56.Lewis CA et al. Article tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol. Cell 55, 253–263 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chen L et al. NADPH production by the oxidative pentosephosphate pathway supports folate metabolism. Nat. Metab 1, 404–415 (2019). [PMC free article] [PubMed] [Google Scholar]
  • 58.Liu L et al. Malic enzyme tracers reveal hypoxia-induced switch in adipocyte NADPH pathway usage. Nat. Chem. Biol 12, 345–352 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fan J et al. Quantitative flux analysis reveals folate-dependent NADPH production. Nature 510, 298–302 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang Z, Chen L, Liu L, Su X & Rabinowitz JD Chemical basis for deuterium labeling of fat and NADPH. J. Am. Chem. Soc 139, 14368–14371 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hong S et al. Live-cell imaging of NADPH production from specific pathways. CCS Chem 3, 1642–1648 (2021). [Google Scholar]
  • 62.Batsios G et al. Imaging 6-phosphogluconolactonase activity in brain tumors in vivo using hyperpolarized δ-[1-13C] gluconolactone. Front. Oncol 11, 1194 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Moreno KX et al. Hyperpolarized δ-[1-13C]gluconolactone as a probe of the pentose phosphate pathway. NMR Biomed 30, e3713 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Maraldi T, Angeloni C, Prata C & Hrelia S NADPH oxidases: redox regulators of stem cell fate and function. Antioxidants 10, 973 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nugud A, Sandeep D & El-Serafi AT Two faces of the coin: minireview for dissecting the role of reactive oxygen species in stem cell potency and lineage commitment. J. Adv. Res 14, 73–79 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhang J, Nuebel E, Daley GQ, Koehler CM & Teitell MA Metabolic regulation in pluripotent stem cells during reprogramming and self-renewal. Cell Stem Cell 11, 589–595 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Stincone A et al. The return of metabolism: biochemistry and physiology of the pentose phosphate pathway. Biol. Rev. Camb. Philos. Soc 90, 927–963 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Folmes CDL, Dzeja PP, Nelson TJ & Terzic A Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 11, 596–606 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Perales-Clemente E, Folmes CDL & Terzic A Metabolic regulation of redox status in stem cells. Antioxid. Redox Signal 21, 1648–1659 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cappellini MD & Fiorelli G Glucose-6-phosphate dehydrogenase deficiency. Lancet 371, 64–74 (2008). [DOI] [PubMed] [Google Scholar]
  • 71.Ruwende C et al. Natural selection of hemi- and heterozygotes for G6PD deficiency in Africa by resistance to severe malaria. Nature 376, 246–249 (1995). [DOI] [PubMed] [Google Scholar]
  • 72.Siniscalco M et al. Population genetics of haemoglobin variants, thalassaemia and glucose-6-phosphate dehydrogenase deficiency, with particular reference to the malaria hypothesis. Bull. World Health Organ 34, 379–393 (1966). [PMC free article] [PubMed] [Google Scholar]
  • 73.Bienzle U, Ayeni O, Lucas AO & Luzzatto L Glucose-6-phosphate dehydrogenase and malaria: greater resistance of females heterozygous for enzyme deficiency and of males with non-deficient variant. Lancet 299, 107–110 (1972). [DOI] [PubMed] [Google Scholar]
  • 74. Allison AC Glucose-6-phosphate dehydrogenase deficiency in red blood cells of East Africans. Nature 186, 531–532 (1960). This paper was among the first to connect G6PD deficiency with resistance to malaria.
  • 75.Luzzatto L, Nannelli C & Notaro R Glucose-6-phosphate dehydrogenase deficiency. Hematol. Oncol. Clin. North Am 30, 373–393 (2016). [DOI] [PubMed] [Google Scholar]
  • 76.WHO Working Group. Glucose-6-phosphate dehydrogenase deficiency. Bull. World Health Organ 67, 601–611 (1989). [PMC free article] [PubMed] [Google Scholar]
  • 77. Longo L et al. Maternally transmitted severe glucose 6-phosphate dehydrogenase deficiency is an embryonic lethal. EMBO J 21, 4229–4239 (2002). This paper demonstrated that complete loss of G6PD is embryonic lethal.
  • 78.Monsalve A, Canals I & Oburoglu L FOXO1 regulates pentose phosphate pathway-mediated induction of developmental erythropoiesis. Front. Cell Dev. Biol 10, 1039636 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ranzani AT & Cordeiro AT Mutations in the tetramer interface of human glucose‐6‐phosphate dehydrogenase reveals kinetic differences between oligomeric states. FEBS Lett 591, 1278–1284 (2017). [DOI] [PubMed] [Google Scholar]
  • 80.Gómez-Manzo S et al. The stability of G6PD is affected by mutations with different clinical phenotypes. Int. J. Mol. Sci 15, 21179–21201 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Naoki H et al. Long-range structural defects by pathogenic mutations in most severe glucose-6-phosphate dehydrogenase deficiency. Proc. Natl Acad. Sci. USA 118, e2022790118 (2021). This paper found that the most severe observed G6PD mutations interfere with dimerization and allosteric binding sites.
  • 82.Mason PJ, Bautista JM & Gilsanz F G6PD deficiency: the genotype–phenotype association. Blood Rev 21, 267–283 (2007). [DOI] [PubMed] [Google Scholar]
  • 83.Cunningham AD, Colavin A, Huang KC & Mochly-Rosen D Coupling between protein stability and catalytic activity determines pathogenicity of G6PD variants. Cell Rep 18, 2592–2599 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.D’Alessandro A et al. Hematologic and systemic metabolic alterations due to Mediterranean class II G6PD deficiency in mice. JCI Insight 6, e147056 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Holland SM Chronic granulomatous disease. Clin. Rev. Allergy Immunol 38, 3–10 (2010). [DOI] [PubMed] [Google Scholar]
  • 86. Cooper MR et al. Complete deficiency of leukocyte glucose-6-phosphate dehydrogenase with defective bactericidal activity. J. Clin. Invest 51, 769–778 (1972). This paper described a patient with G6PD deficiency who had features of chronic granulomatous disease.
  • 87. Roos D et al. Molecular basis and enzymatic properties of glucose 6-phosphate dehydrogenase volendam, leading to chronic nonspherocytic anemia, granulocyte dysfunction, and increased susceptibility to infections. Blood 94, 2955–2962 (1999). This paper described a patient with G6PD deficiency who experienced chronic infections and had decreased neutrophil respiratory burst.
  • 88.van Bruggen R et al. Deletion of leucine 61 in glucose-6-phosphate dehydrogenase leads to chronic nonspherocytic anemia, granulocyte dysfunction, and increased susceptibility to infections. Blood 100, 1026–1030 (2002). [DOI] [PubMed] [Google Scholar]
  • 89.Martinez FO & Gordon S The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6, 13 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sica A & Mantovani A Macrophage plasticity and polarization: in vivo veritas. J. Clin. Invest 122, 787–795 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mosser DM & Edwards JP Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol 8, 958–969 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Galván-Peña S & O’Neill LAJ Metabolic reprograming in macrophage polarization. Front. Immunol 5, 420 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nagy C & Haschemi A Time and demand are two critical dimensions of immunometabolism: the process of macrophage activation and the pentose phosphate pathway. Front. Immunol 6, 164 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Haschemi A et al. The sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metab 15, 813–826 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Koo S, Szczesny B, Wan X, Putluri N & Garg NJ Pentose phosphate shunt modulates reactive oxygen species and nitric oxide production controlling Trypanosoma cruzi in macrophages. Front. Immunol 9, 202 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ma J et al. Glycogen metabolism regulates macrophage-mediated acute inflammatory responses. Nat. Commun 11, 1769 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mira H et al. Macrophage glucose-6-phosphate dehydrogenase stimulates proinflammatory responses with oxidative stress. Mol. Cell. Biol 33, 2425–2435 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Ghergurovich JM et al. A small molecule G6PD inhibitor reveals immune dependence on pentose phosphate pathway. Nat. Chem. Biol 16, 731–739 (2020). This paper developed a more specific small-molecule G6PD inhibitor and found a role for the oxPPP in T cell function.
  • 99. Bories GFP et al. Macrophage metabolic adaptation to heme detoxification involves CO-dependent activation of the pentose phosphate pathway. Blood 136, 1535–1548 (2020). This paper demonstrated that NADPH from the oxPPP is important for haem detoxification in macrophages.
  • 100.Wamelink MMC et al. Sedoheptulokinase deficiency due to a 57-kb deletion in cystinosis patients causes urinary accumulation of sedoheptulose: elucidation of the CARKL gene. Hum. Mutat 29, 532–536 (2008). [DOI] [PubMed] [Google Scholar]
  • 101.Kardon T, Stroobant V, Veiga-da-Cunha M & Van Schaftingen E Characterization of mammalian sedoheptulokinase and mechanism of formation of erythritol in sedoheptulokinase deficiency. FEBS Lett 582, 3330–3334 (2008). [DOI] [PubMed] [Google Scholar]
  • 102.Baardman J et al. A defective pentose phosphate pathway reduces inflammatory macrophage responses during hypercholesterolemia. Cell Rep 25, 2044–2052 (2018). [DOI] [PubMed] [Google Scholar]
  • 103.Moore KJ, Sheedy FJ & Fisher EA Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol 13, 709–721 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kindzelskii AL et al. Pregnancy alters glucose-6-phosphate dehydrogenase trafficking, cell metabolism, and oxidant release of maternal neutrophils. J. Clin. Invest 110, 1801–1811 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Azevedo EP et al. A metabolic shift toward pentose phosphate pathway is necessary for amyloid fibril- and phorbol 12-myristate 13-acetate-induced neutrophil extracellular trap (NET) formation. J. Biol. Chem 290, 22174–22183 (2015). This paper found that the PPP supports NET formation in neutrophils.
  • 106.Li Y et al. Neutrophil metabolomics in severe COVID-19 reveal GAPDH as a suppressor of neutrophil extracellular trap formation. Nat. Commun 14, 2610 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Woody RC, Steele RW, Knapple WL & Pilkington NS Jr. Impaired neutrophil function in children with seizures treated with the ketogenic diet. J. Pediatr 115, 427–430 (1989). [DOI] [PubMed] [Google Scholar]
  • 108.Wong SL et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med 21, 815–819 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gerriets VA & Rathmell JC Metabolic pathways in T cell fate and function. Trends Immunol 33, 168–173 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Klein Geltink RI, Kyle RL & Pearce EL Unraveling the complex interplay between T cell metabolism and function. Annu. Rev. Immunol 36, 461–488 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ron-Harel N et al. Mitochondrial biogenesis and proteome remodeling promote one-carbon metabolism for T cell activation. Cell Metab 24, 104–117 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Field CS et al. Mitochondrial integrity regulated by lipid metabolism is a cell-intrinsic checkpoint for Treg suppressive function. Cell Metab 31, 422–437 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Gerriets VA et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat. Immunol 17, 1459–1466 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Michalek RD et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J. Immunol 186, 3299–3303 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wang R et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Klein Geltink RI et al. Metabolic conditioning of CD8+ effector T cells for adoptive cell therapy. Nat. Metab 2, 703–716 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yang Z et al. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med 8, 331ra38 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Weyand CM, Shen Y & Goronzy JJ Redox-sensitive signaling in inflammatory T cells and in autoimmune disease. Free Radic. Biol. Med 125, 36–43 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Daneshmandi S, Cassel T, Higashi RM, Fan TW-M & Seth P 6-Phosphogluconate dehydrogenase (6PGD), a key checkpoint in reprogramming of regulatory T cells metabolism and function. eLife 10, e67476 (2021). This paper found that mice with 6PGD-deficient regulatory T cells die at an early age owing to autoimmune disease.
  • 120.Liu Q et al. Non-oxidative pentose phosphate pathway controls regulatory T cell function by integrating metabolism and epigenetics. Nat. Metab 4, 559–574 (2022). [DOI] [PubMed] [Google Scholar]
  • 121.Daneshmandi S et al. Blockade of 6-phosphogluconate dehydrogenase generates CD8+ effector T cells with enhanced anti-tumor function. Cell Rep 34, 108831 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Xu Y et al. Glucose-6-phosphate dehydrogenase-deficient mice have increased renal oxidative stress and increased albuminuria. FASEB J 24, 609–616 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jain M et al. Glucose-6-phosphate dehydrogenase modulates cytosolic redox status and contractile phenotype in adult cardiomyocytes. Circ. Res 93, e9–e16 (2003). [DOI] [PubMed] [Google Scholar]
  • 124.Hecker PA et al. Glucose 6-phosphate dehydrogenase deficiency increases redox stress and moderately accelerates the development of heart failure. Circ. Heart Fail 6, 118–126 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Thomas JE et al. Glucose-6-phosphate dehydrogenase deficiency is associated with cardiovascular disease in U.S. military centers. Tex. Heart Inst. J 45, 144–150 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Spencer NY & Stanton RC Glucose 6-phosphate dehydrogenase and the kidney. Curr. Opin. Nephrol. Hypertens 26, 43–49 (2017). [DOI] [PubMed] [Google Scholar]
  • 127.Tiwari M Glucose 6 phosphatase dehydrogenase (G6PD) and neurodegenerative disorders: mapping diagnostic and therapeutic opportunities. Genes Dis 4, 196–203 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kuehne A et al. Acute activation of oxidative pentose phosphate pathway as first-line response to oxidative stress in human skin cells. Mol. Cell 59, 359–371 (2015). [DOI] [PubMed] [Google Scholar]
  • 129.Saghi A, Gao J-L, Giampietro L, letchuman S & Amiri-Ardekani E A mechanistic analysis of henna in G6PD deficiency patients. Toxicol. Anal. Clin 35, 23–32 (2023). [Google Scholar]
  • 130.Weber G & Korting G Glucose-6-phosphate dehydrogenase in human skin. J. Invest. Dermatol 42, 167–169 (1964). [DOI] [PubMed] [Google Scholar]
  • 131.Agrawal D, Shajil EM, Marfatia YS & Begum R Study on the antioxidant status of vitiligo patients of different age groups in Baroda. Pigment Cell Res 17, 289–294 (2004). [DOI] [PubMed] [Google Scholar]
  • 132.Farahi-Jahromy A et al. Decreased glucose-6-phosphate dehydrogenase levels in vitiligo patients: further evidence of oxidative stress. Adv. Biomed. Res 3, 34 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Zackheim HS Treatment of psoriasis with 6-aminonicotinamide. Arch. Dermatol 111, 880–882 (1975). [PubMed] [Google Scholar]
  • 134.Nakayama H & Weser E Adaptation of small bowel after intestinal resection: increase in the pentose phosphate pathway. Biochem. Biophys. Acta Gen. Subj 279, 416–423 (1972). [DOI] [PubMed] [Google Scholar]
  • 135.Tian N et al. TKT maintains intestinal ATP production and inhibits apoptosis-induced colitis. Cell Death Dis 12, 853 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Cao L et al. G6PD plays a neuroprotective role in brain ischemia through promoting pentose phosphate pathway. Free Radic. Biol. Med 112, 433–444 (2017). [DOI] [PubMed] [Google Scholar]
  • 137.Vetrovoy O et al. Neuroprotective mechanism of hypoxic post-conditioning involves HIF1-associated regulation of the pentose phosphate pathway in rat brain. Neurochem. Res 44, 1425–1436 (2019). [DOI] [PubMed] [Google Scholar]
  • 138.Jain M et al. Increased myocardial dysfunction after ischemia–reperfusion in mice lacking glucose-6-phosphate dehydrogenase. Circulation 109, 898–903 (2004). [DOI] [PubMed] [Google Scholar]
  • 139.Fan W, Lu D, Shi X & He H Flux of glucose through pentose phosphate pathway and glycolysis in mouse hearts subjected to low-flow ischemia. FASEB J 34, 29–38 (2020). [Google Scholar]
  • 140.Pes GM, Errigo A, Soro S, Longo NP & Dore MP Glucose-6-phosphate dehydrogenase deficiency reduces susceptibility to cancer of endodermal origin. Acta Oncol 58, 1205–1211 (2019). [DOI] [PubMed] [Google Scholar]
  • 141.Dore MP, Davoli A, Longo N, Marras G & Pes GM Glucose-6-phosphate dehydrogenase deficiency and risk of colorectal cancer in northern Sardinia: a retrospective observational study. Medicine 95, e5254 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Gill JG, Piskounova E & Morrison SJ Cancer, oxidative stress, and metastasis. Cold Spring Harb. Symp. Quant. Biol 81, 163–175 (2016). [DOI] [PubMed] [Google Scholar]
  • 143.Chandel NS & Tuveson DA The promise and perils of antioxidants for cancer patients. N. Engl. J. Med 371, 177–178 (2014). [DOI] [PubMed] [Google Scholar]
  • 144. Zhang Y et al. Upregulation of antioxidant capacity and nucleotide precursor availability suffices for oncogenic transformation. Cell Metab 33, 94–109 (2021). This paper found that overexpression of G6PD is sufficient for oncogenic transformation.
  • 145. Ghergurovich JM et al. Glucose-6-phosphate dehydrogenase is not essential for K-Ras-driven tumor growth or metastasis. Cancer Res 80, 3820–3829 (2020). This paper demonstrated that the oxPPP is not required for tumour growth in several cancer models.
  • 146.Piskounova E et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Kristell LG et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med 7, 308re8 (2015). [DOI] [PubMed] [Google Scholar]
  • 148.Cha YJ, Jung WH & Koo JS Differential site-based expression of pentose phosphate pathway-related proteins among breast cancer metastases. Dis. Markers 2017, 7062517 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Aurora AB et al. Loss of glucose 6-phosphate dehydrogenase function increases oxidative stress and glutaminolysis in metastasizing melanoma cells. Proc. Natl Acad. Sci. USA 119, e2120617119 (2022). This paper found that the oxPPP promotes tumour metastasis by combating associated oxidative stress.
  • 150.Consortium TAPG et al. AACR Project GENIE: powering precision medicine through an international consortium. Cancer Discov 7, 818–831 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Di Federico A, De Giglio A, Parisi C & Gelsomino F STK11/LKB1 and KEAP1 mutations in non-small cell lung cancer: prognostic rather than predictive? Eur. J. Cancer 157, 108–113 (2021). [DOI] [PubMed] [Google Scholar]
  • 152.Marinelli D et al. KEAP1-driven co-mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Ann. Oncol 31, 1746–1754 (2020). [DOI] [PubMed] [Google Scholar]
  • 153.Dempke WCM & Reck M KEAP1/NRF2 (NFE2L2) mutations in NSCLC—fuel for a superresistant phenotype? Lung Cancer 159, 10–17 (2021). [DOI] [PubMed] [Google Scholar]
  • 154.Best SA et al. Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma. Nat. Commun 10, 4190 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Bhanot H et al. Acute myeloid leukemia cells require 6-phosphogluconate dehydrogenase for cell growth and NADPH-dependent metabolic reprogramming. Oncotarget 8, 67639–67650 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Badur MG et al. Oncogenic R132 IDH1 mutations limit NADPH for de novo lipogenesis through (D)2-hydroxyglutarate production in fibrosarcoma cells. Cell Rep 25, 1018–1026 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Gelman SJ et al. Consumption of NADPH for 2-HG synthesis increases pentose phosphate pathway flux and sensitizes cells to oxidative stress. Cell Rep 22, 512–522 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhao F et al. Imatinib resistance associated with BCR-ABL upregulation is dependent on HIF-1α-induced metabolic reprograming. Oncogene 29, 2962–2972 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Tseng C-W et al. Transketolase regulates the metabolic switch to control breast cancer cell metastasis via the α-ketoglutarate signaling pathway. Cancer Res 78, 2799–2812 (2018). [DOI] [PubMed] [Google Scholar]
  • 160.Ricciardelli C et al. Transketolase is upregulated in metastatic peritoneal implants and promotes ovarian cancer cell proliferation. Clin. Exp. Metastasis 32, 441–455 (2015). [DOI] [PubMed] [Google Scholar]
  • 161.Li M et al. Transketolase promotes colorectal cancer metastasis through regulating AKT phosphorylation. Cell Death Dis 13, 99 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Huang J et al. Understanding anthracycline cardiotoxicity from mitochondrial aspect. Front. Pharmacol 13, 811406 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Ma L & Cheng Q Inhibiting 6-phosphogluconate dehydrogenase reverses doxorubicin resistance in anaplastic thyroid cancer via inhibiting NADPH-dependent metabolic reprogramming. Biochem. Biophys. Res. Commun 498, 912–917 (2018). [DOI] [PubMed] [Google Scholar]
  • 164.Polimeni M et al. Modulation of doxorubicin resistance by the glucose-6-phosphate dehydrogenase activity. Biochem. J 439, 141–149 (2011). [DOI] [PubMed] [Google Scholar]
  • 165.Kleih M et al. Direct impact of cisplatin on mitochondria induces ROS production that dictates cell fate of ovarian cancer cells. Cell Death Dis 10, 851 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ju HQ et al. Disrupting G6PD-mediated redox homeostasis enhances chemosensitivity in colorectal cancer. Oncogene 36, 6282–6292 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wang X et al. lncRNA-encoded pep-AP attenuates the pentose phosphate pathway and sensitizes colorectal cancer cells to oxaliplatin. EMBO Rep 23, e53140 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Gao WC et al. Targeting oxidative pentose phosphate pathway prevents recurrence in mutant Kras colorectal carcinomas. PLoS Biol 17, e3000425 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Li Q et al. Rac1 activates non-oxidative pentose phosphate pathway to induce chemoresistance of breast cancer. Nat. Commun 11, 1456 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Gunda V et al. MUC1-mediated metabolic alterations regulate response to radiotherapy in pancreatic cancer. Clin. Cancer Res 23, 5881–5891 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Vaziri-Gohar A et al. Limited nutrient availability in the tumor microenvironment renders pancreatic tumors sensitive to allosteric IDH1 inhibitors. Nat. Cancer 3, 852–865 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Ding H et al. Activation of the NRF2 antioxidant program sensitizes tumors to G6PD inhibition. Sci. Adv 7, eabk1023 (2021). This paper found that KEAP1-mutant tumours are sensitive to oxPPP inhibition.
  • 173.Benhar M, Forrester MT & Stamler JS Protein denitrosylation: enzymatic mechanisms and cellular functions. Nat. Rev. Mol. Cell Biol 10, 721–732 (2009). [DOI] [PubMed] [Google Scholar]
  • 174.Gamper N & Ooi L Redox and nitric oxide-mediated regulation of sensory neuron ion channel function. Antioxid. Redox Signal 22, 486–504 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Tejero J, Shiva S & Gladwin MT Sources of vascular nitric oxide and reactive oxygen species and their regulation. Physiol. Rev 99, 311–379 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Block K & Gorin Y Aiding and abetting roles of NOX oxidases in cellular transformation. Nat. Rev. Cancer 12, 627–637 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Moloney JN & Cotter TG ROS signalling in the biology of cancer. Semin. Cell Dev. Biol 80, 50–64 (2018). [DOI] [PubMed] [Google Scholar]
  • 178.Michaelson LP, Iler C & Ward CW ROS and RNS signaling in skeletal muscle: critical signals and therapeutic targets. Annu. Rev. Nurs. Res 31, 367–387 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tochhawng L, Deng S, Pervaiz S & Yap CT Redox regulation of cancer cell migration and invasion. Mitochondrion 13, 246–253 (2013). [DOI] [PubMed] [Google Scholar]
  • 180.Rappez L et al. SpaceM reveals metabolic states of single cells. Nat. Methods 18, 799–805 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Tian H et al. Multi-modal mass spectrometry imaging reveals single-cell metabolic states in mammalian liver. Preprint at bioRxiv 10.1101/2022.09.26.508878 (2022). [DOI] [Google Scholar]
  • 182.Tian H, Sheraz née Rabbani S, Vickerman JC & Winograd N Multiomics imaging using high-energy water gas cluster ion beam secondary ion mass spectrometry [(H2O)n-GCIB-SIMS] of frozen-hydrated cells and tissue. Anal. Chem 93, 7808–7814 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Pareek V, Tian H, Winograd N & Benkovic SJ Metabolomics and mass spectrometry imaging reveal channeled de novo purine synthesis in cells. Science 368, 283–290 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Tao R et al. Genetically encoded fluorescent sensors reveal dynamic regulation of NADPH metabolism. Nat. Methods 14, 720–728 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cameron WD et al. Apollo-NADP+: a spectrally tunable family of genetically encoded sensors for NADP+. Nat. Methods 13, 352–358 (2016). [DOI] [PubMed] [Google Scholar]
  • 186.Zhao F-L, Zhang C, Zhang C, Tang Y & Ye B-C A genetically encoded biosensor for in vitro and in vivo detection of NADP. Biosens. Bioelectron 77, 901–906 (2016). [DOI] [PubMed] [Google Scholar]
  • 187.Zhao Y et al. In vivo monitoring of cellular energy metabolism using SoNar, a highly responsive sensor for NAD+/NADH redox state. Nat. Protoc 11, 1345–1359 (2016). [DOI] [PubMed] [Google Scholar]
  • 188.Zhang Z, Cheng X, Zhao Y & Yang Y Lighting up live-cell and in vivo central carbon metabolism with genetically encoded fluorescent sensors. Annu. Rev. Anal. Chem 13, 293–314 (2020). [DOI] [PubMed] [Google Scholar]
  • 189.Cracan V, Titov DV, Shen H, Grabarek Z & Mootha VK A genetically encoded tool for manipulation of NADP+/NADPH in living cells. Nat. Chem. Biol 13, 1088–1095 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Köhler E, Barrach H-J & Neubert D Inhibition of NADP dependent oxidoreductases by the 6‐aminonicotinamide analogue of NADP. FEBS Lett 6, 225–228 (1970). [DOI] [PubMed] [Google Scholar]
  • 191. Hwang S et al. Correcting glucose-6-phosphate dehydrogenase deficiency with a small-molecule activator. Nat. Commun 9, 4045 (2018). This paper found a small-molecule G6PD activator that can increase activity and thermostability of Canton mutant R459L, opening the door for a potential therapeutic for G6PD deficiency.
  • 192.Raub AG et al. Small-molecule activators of glucose-6-phosphate dehydrogenase (G6PD) bridging the dimer interface. ChemMedChem 14, 1321–1324 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES