Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2019

Open Access 01-12-2019 | Parkinson's Disease | Research

The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration

Authors: Dezhen Tu, Yun Gao, Ru Yang, Tian Guan, Jau-Shyong Hong, Hui-Ming Gao

Published in: Journal of Neuroinflammation | Issue 1/2019

Login to get access

Abstract

Background

Metabolic dysfunction and neuroinflammation are increasingly implicated in Parkinson’s disease (PD). The pentose phosphate pathway (PPP, a metabolic pathway parallel to glycolysis) converts glucose-6-phosphate into pentoses and generates ribose-5-phosphate and NADPH thereby governing anabolic biosynthesis and redox homeostasis. Brains and immune cells display high activity of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the PPP. A postmortem study reveals dysregulation of G6PD enzyme in brains of PD patients. However, spatial and temporal changes in activity/expression of G6PD in PD remain undetermined. More importantly, it is unclear how dysfunction of G6PD and the PPP affects neuroinflammation and neurodegeneration in PD.

Methods

We examined expression/activity of G6PD and its association with microglial activation and dopaminergic neurodegeneration in multiple chronic PD models generated by an intranigral/intraperitoneal injection of LPS, daily subcutaneous injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) for 6 days, or transgenic expression of A53T α-synuclein. Primary microglia were transfected with G6PD siRNAs and treated with lipopolysaccharide (LPS) to examine effects of G6PD knockdown on microglial activation and death of co-cultured neurons. LPS alone or with G6PD inhibitor(s) was administrated to mouse substantia nigra or midbrain neuron-glia cultures. While histological and biochemical analyses were conducted to examine microglial activation and dopaminergic neurodegeneration in vitro and in vivo, rotarod behavior test was performed to evaluate locomotor impairment in mice.

Results

Expression and activity of G6PD were elevated in LPS-treated midbrain neuron-glia cultures (an in vitro PD model) and the substantia nigra of four in vivo PD models. Such elevation was positively associated with microglial activation and dopaminergic neurodegeneration. Furthermore, inhibition of G6PD by 6-aminonicotinamide and dehydroepiandrosterone and knockdown of microglial G6PD attenuated LPS-elicited chronic dopaminergic neurodegeneration. Mechanistically, microglia with elevated G6PD activity/expression produced excessive NADPH and provided abundant substrate to over-activated NADPH oxidase (NOX2) leading to production of excessive reactive oxygen species (ROS). Knockdown and inhibition of G6PD ameliorated LPS-triggered production of ROS and activation of NF-кB thereby dampening microglial activation.

Conclusions

Our findings indicated that G6PD-mediated PPP dysfunction and neuroinflammation exacerbated each other mediating chronic dopaminergic neurodegeneration and locomotor impairment. Insight into metabolic-inflammatory interface suggests that G6PD and NOX2 are potential therapeutic targets for PD.
Appendix
Available only for authorised users
Literature
1.
go back to reference Olanow CW, Tatton WG. Etiology and pathogenesis of Parkinson’s disease. Annu Rev Neurosci. 1999;22:123–44.CrossRefPubMed Olanow CW, Tatton WG. Etiology and pathogenesis of Parkinson’s disease. Annu Rev Neurosci. 1999;22:123–44.CrossRefPubMed
3.
5.
go back to reference Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 2008;29:357–65.CrossRefPubMedPubMedCentral Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 2008;29:357–65.CrossRefPubMedPubMedCentral
6.
go back to reference Wahner AD, Sinsheimer JS, Bronstein JM, Ritz B. Inflammatory cytokine gene polymorphisms and increased risk of Parkinson disease. Arch Neurol. 2007;64:836–40.CrossRefPubMed Wahner AD, Sinsheimer JS, Bronstein JM, Ritz B. Inflammatory cytokine gene polymorphisms and increased risk of Parkinson disease. Arch Neurol. 2007;64:836–40.CrossRefPubMed
7.
go back to reference Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet. 2010;42:781–5.CrossRefPubMedPubMedCentral Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet. 2010;42:781–5.CrossRefPubMedPubMedCentral
8.
go back to reference McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.CrossRefPubMed McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.CrossRefPubMed
9.
go back to reference Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.CrossRefPubMed Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.CrossRefPubMed
10.
11.
go back to reference Zhang F, Shi JS, Zhou H, Wilson B, Hong JS, Gao HM. Resveratrol protects dopamine neurons against lipopolysaccharide-induced neurotoxicity through its anti-inflammatory actions. Mol Pharmacol. 2010;78:466–77.CrossRefPubMedPubMedCentral Zhang F, Shi JS, Zhou H, Wilson B, Hong JS, Gao HM. Resveratrol protects dopamine neurons against lipopolysaccharide-induced neurotoxicity through its anti-inflammatory actions. Mol Pharmacol. 2010;78:466–77.CrossRefPubMedPubMedCentral
12.
go back to reference Wu DC, Teismann P, Tieu K, Vila M, Jackson-Lewis V, Ischiropoulos H, Przedborski S. NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2003;100:6145–50.CrossRefPubMedPubMedCentral Wu DC, Teismann P, Tieu K, Vila M, Jackson-Lewis V, Ischiropoulos H, Przedborski S. NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2003;100:6145–50.CrossRefPubMedPubMedCentral
13.
go back to reference Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, Hargreaves IP, Pope S, Holton JL, Lees A, et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiol Aging. 2014;35:1111–5.CrossRefPubMedPubMedCentral Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, Hargreaves IP, Pope S, Holton JL, Lees A, et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiol Aging. 2014;35:1111–5.CrossRefPubMedPubMedCentral
14.
go back to reference Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H, Hoehn M, Fink GR, Schroeter M, Graf R. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. Neuroimage. 2016;128:54–62.CrossRefPubMed Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H, Hoehn M, Fink GR, Schroeter M, Graf R. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. Neuroimage. 2016;128:54–62.CrossRefPubMed
15.
go back to reference Anandhan A, Jacome MS, Lei S, Hernandez-Franco P, Pappa A, Panayiotidis MI, Powers R, Franco R. Metabolic dysfunction in Parkinson’s disease: bioenergetics, redox homeostasis and central carbon metabolism. Brain Res Bull. 2017;133:12–30.CrossRefPubMedPubMedCentral Anandhan A, Jacome MS, Lei S, Hernandez-Franco P, Pappa A, Panayiotidis MI, Powers R, Franco R. Metabolic dysfunction in Parkinson’s disease: bioenergetics, redox homeostasis and central carbon metabolism. Brain Res Bull. 2017;133:12–30.CrossRefPubMedPubMedCentral
16.
go back to reference Perl A, Hanczko R, Telarico T, Oaks Z, Landas S. Oxidative stress, inflammation and carcinogenesis are controlled through the pentose phosphate pathway by transaldolase. Trends Mol Med. 2011;17:395–403.CrossRefPubMedPubMedCentral Perl A, Hanczko R, Telarico T, Oaks Z, Landas S. Oxidative stress, inflammation and carcinogenesis are controlled through the pentose phosphate pathway by transaldolase. Trends Mol Med. 2011;17:395–403.CrossRefPubMedPubMedCentral
17.
go back to reference Battistuzzi G, D’Urso M, Toniolo D, Persico GM, Luzzatto L. Tissue-specific levels of human glucose-6-phosphate dehydrogenase correlate with methylation of specific sites at the 3′ end of the gene. Proc Natl Acad Sci U S A. 1985;82:1465–9.CrossRefPubMedPubMedCentral Battistuzzi G, D’Urso M, Toniolo D, Persico GM, Luzzatto L. Tissue-specific levels of human glucose-6-phosphate dehydrogenase correlate with methylation of specific sites at the 3′ end of the gene. Proc Natl Acad Sci U S A. 1985;82:1465–9.CrossRefPubMedPubMedCentral
18.
go back to reference D’Urso M, Mareni C, Toniolo D, Piscopo M, Schlessinger D, Luzzatto L. Regulation of glucose 6-phosphate dehydrogenase expression in CHO-human fibroblast somatic cell hybrids. Somatic Cell Genet. 1983;9:429–43.CrossRefPubMed D’Urso M, Mareni C, Toniolo D, Piscopo M, Schlessinger D, Luzzatto L. Regulation of glucose 6-phosphate dehydrogenase expression in CHO-human fibroblast somatic cell hybrids. Somatic Cell Genet. 1983;9:429–43.CrossRefPubMed
19.
go back to reference Herrero-Mendez A, Almeida A, Fernandez E, Maestre C, Moncada S, Bolanos JP. The bioenergetic and antioxidant status of neurons is controlled by continuous degradation of a key glycolytic enzyme by APC/C-Cdh1. Nat Cell Biol. 2009;11:747–52.CrossRefPubMed Herrero-Mendez A, Almeida A, Fernandez E, Maestre C, Moncada S, Bolanos JP. The bioenergetic and antioxidant status of neurons is controlled by continuous degradation of a key glycolytic enzyme by APC/C-Cdh1. Nat Cell Biol. 2009;11:747–52.CrossRefPubMed
20.
go back to reference Gao L, Mir P, Diaz-Corrales FJ, Mejias R, Carrillo F, Vime PJ, Diaz-Martin J, Palomino A, Carballo M, Pintado E, et al. Glucose-6-phosphate dehydrogenase activity in Parkinson’s disease. J Neurol. 2008;255:1850–1.CrossRefPubMed Gao L, Mir P, Diaz-Corrales FJ, Mejias R, Carrillo F, Vime PJ, Diaz-Martin J, Palomino A, Carballo M, Pintado E, et al. Glucose-6-phosphate dehydrogenase activity in Parkinson’s disease. J Neurol. 2008;255:1850–1.CrossRefPubMed
21.
go back to reference Abraham S, Soundararajan CC, Vivekanandhan S, Behari M. Erythrocyte antioxidant enzymes in Parkinson’s disease. Indian J Med Res. 2005;121:111–5.PubMed Abraham S, Soundararajan CC, Vivekanandhan S, Behari M. Erythrocyte antioxidant enzymes in Parkinson’s disease. Indian J Med Res. 2005;121:111–5.PubMed
22.
go back to reference Yin F, Sancheti H, Patil I, Cadenas E. Energy metabolism and inflammation in brain aging and Alzheimer’s disease. Free Radic Biol Med. 2016. Yin F, Sancheti H, Patil I, Cadenas E. Energy metabolism and inflammation in brain aging and Alzheimer’s disease. Free Radic Biol Med. 2016.
23.
go back to reference Zhang W, Wang T, Qin L, Gao HM, Wilson B, Ali SF, Zhang W, Hong JS, Liu B. Neuroprotective effect of dextromethorphan in the MPTP Parkinson’s disease model: role of NADPH oxidase. Faseb j. 2004;18:589–91.CrossRefPubMed Zhang W, Wang T, Qin L, Gao HM, Wilson B, Ali SF, Zhang W, Hong JS, Liu B. Neuroprotective effect of dextromethorphan in the MPTP Parkinson’s disease model: role of NADPH oxidase. Faseb j. 2004;18:589–91.CrossRefPubMed
24.
go back to reference Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci. 2008;28:7687–98.CrossRefPubMedPubMedCentral Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM. Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci. 2008;28:7687–98.CrossRefPubMedPubMedCentral
25.
26.
go back to reference Gao HM, Liu B, Hong JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2003;23:6181–7.CrossRefPubMedPubMedCentral Gao HM, Liu B, Hong JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci. 2003;23:6181–7.CrossRefPubMedPubMedCentral
27.
go back to reference Zhou H, Zhang F, Chen SH, Zhang D, Wilson B, Hong JS, Gao HM. Rotenone activates phagocyte NADPH oxidase by binding to its membrane subunit gp91phox. Free Radic Biol Med. 2012;52:303–13.CrossRefPubMed Zhou H, Zhang F, Chen SH, Zhang D, Wilson B, Hong JS, Gao HM. Rotenone activates phagocyte NADPH oxidase by binding to its membrane subunit gp91phox. Free Radic Biol Med. 2012;52:303–13.CrossRefPubMed
28.
go back to reference Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55:2320–32.CrossRefPubMed Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55:2320–32.CrossRefPubMed
29.
go back to reference Kohler E, Barrach H, Neubert D. Inhibition of NADP dependent oxidoreductases by the 6-aminonicotinamide analogue of NADP. FEBS Lett. 1970;6:225–8.CrossRefPubMed Kohler E, Barrach H, Neubert D. Inhibition of NADP dependent oxidoreductases by the 6-aminonicotinamide analogue of NADP. FEBS Lett. 1970;6:225–8.CrossRefPubMed
30.
go back to reference Hofgaard JP, Sigurdardottir KS, Treiman M. Protection by 6-aminonicotinamide against oxidative stress in cardiac cells. Pharmacol Res. 2006;54:303–10.CrossRefPubMed Hofgaard JP, Sigurdardottir KS, Treiman M. Protection by 6-aminonicotinamide against oxidative stress in cardiac cells. Pharmacol Res. 2006;54:303–10.CrossRefPubMed
31.
go back to reference Raineri R, Levy HR. On the specificity of steroid interaction with mammary glucose 6-phosphate dehydrogenase. Biochemistry. 1970;9:2233–43.CrossRefPubMed Raineri R, Levy HR. On the specificity of steroid interaction with mammary glucose 6-phosphate dehydrogenase. Biochemistry. 1970;9:2233–43.CrossRefPubMed
32.
go back to reference Park J, Choe SS, Choi AH, Kim KH, Yoon MJ, Suganami T, Ogawa Y, Kim JB. Increase in glucose-6-phosphate dehydrogenase in adipocytes stimulates oxidative stress and inflammatory signals. Diabetes. 2006;55:2939–49.CrossRefPubMed Park J, Choe SS, Choi AH, Kim KH, Yoon MJ, Suganami T, Ogawa Y, Kim JB. Increase in glucose-6-phosphate dehydrogenase in adipocytes stimulates oxidative stress and inflammatory signals. Diabetes. 2006;55:2939–49.CrossRefPubMed
33.
go back to reference Krum JM. Age-dependent susceptibility of CNS glial populations in situ to the antimetabolite 6-aminonicotinamide. Mol Chem Neuropathol. 1995;26:79–94.CrossRefPubMed Krum JM. Age-dependent susceptibility of CNS glial populations in situ to the antimetabolite 6-aminonicotinamide. Mol Chem Neuropathol. 1995;26:79–94.CrossRefPubMed
34.
go back to reference Penkowa M, Camats J, Hadberg H, Quintana A, Rojas S, Giralt M, Molinero A, Campbell IL, Hidalgo J. Astrocyte-targeted expression of interleukin-6 protects the central nervous system during neuroglial degeneration induced by 6-aminonicotinamide. J Neurosci Res. 2003;73:481–96.CrossRefPubMed Penkowa M, Camats J, Hadberg H, Quintana A, Rojas S, Giralt M, Molinero A, Campbell IL, Hidalgo J. Astrocyte-targeted expression of interleukin-6 protects the central nervous system during neuroglial degeneration induced by 6-aminonicotinamide. J Neurosci Res. 2003;73:481–96.CrossRefPubMed
35.
go back to reference Sanna F, Bonatesta RR, Frongia B, Uda S, Banni S, Melis MP, Collu M, Madeddu C, Serpe R, Puddu S, et al. Production of inflammatory molecules in peripheral blood mononuclear cells from severely glucose-6-phosphate dehydrogenase-deficient subjects. J Vasc Res. 2007;44:253–63.CrossRefPubMed Sanna F, Bonatesta RR, Frongia B, Uda S, Banni S, Melis MP, Collu M, Madeddu C, Serpe R, Puddu S, et al. Production of inflammatory molecules in peripheral blood mononuclear cells from severely glucose-6-phosphate dehydrogenase-deficient subjects. J Vasc Res. 2007;44:253–63.CrossRefPubMed
36.
go back to reference Ahsan H. 3-Nitrotyrosine: a biomarker of nitrogen free radical species modified proteins in systemic autoimmunogenic conditions. Hum Immunol. 2013;74:1392–9.CrossRefPubMed Ahsan H. 3-Nitrotyrosine: a biomarker of nitrogen free radical species modified proteins in systemic autoimmunogenic conditions. Hum Immunol. 2013;74:1392–9.CrossRefPubMed
37.
go back to reference Jomova K, Vondrakova D, Lawson M, Valko M. Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem. 2010;345:91–104.CrossRefPubMed Jomova K, Vondrakova D, Lawson M, Valko M. Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem. 2010;345:91–104.CrossRefPubMed
38.
go back to reference Edison P, Ahmed I, Fan Z, Hinz R, Gelosa G, Ray Chaudhuri K, Walker Z, Turkheimer FE, Brooks DJ. Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38:938–49.CrossRefPubMedPubMedCentral Edison P, Ahmed I, Fan Z, Hinz R, Gelosa G, Ray Chaudhuri K, Walker Z, Turkheimer FE, Brooks DJ. Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38:938–49.CrossRefPubMedPubMedCentral
39.
go back to reference Ahmed SS, Santosh W, Kumar S, Christlet HT. Metabolic profiling of Parkinson’s disease: evidence of biomarker from gene expression analysis and rapid neural network detection. J Biomed Sci. 2009;16:63.CrossRefPubMedPubMedCentral Ahmed SS, Santosh W, Kumar S, Christlet HT. Metabolic profiling of Parkinson’s disease: evidence of biomarker from gene expression analysis and rapid neural network detection. J Biomed Sci. 2009;16:63.CrossRefPubMedPubMedCentral
40.
go back to reference Eberling JL, Richardson BC, Reed BR, Wolfe N, Jagust WJ. Cortical glucose metabolism in Parkinson’s disease without dementia. Neurobiol Aging. 1994;15:329–35.CrossRefPubMed Eberling JL, Richardson BC, Reed BR, Wolfe N, Jagust WJ. Cortical glucose metabolism in Parkinson’s disease without dementia. Neurobiol Aging. 1994;15:329–35.CrossRefPubMed
41.
go back to reference Mejias R, Villadiego J, Pintado CO, Vime PJ, Gao L, Toledo-Aral JJ, Echevarria M, Lopez-Barneo J. Neuroprotection by transgenic expression of glucose-6-phosphate dehydrogenase in dopaminergic nigrostriatal neurons of mice. J Neurosci. 2006;26:4500–8.CrossRefPubMedPubMedCentral Mejias R, Villadiego J, Pintado CO, Vime PJ, Gao L, Toledo-Aral JJ, Echevarria M, Lopez-Barneo J. Neuroprotection by transgenic expression of glucose-6-phosphate dehydrogenase in dopaminergic nigrostriatal neurons of mice. J Neurosci. 2006;26:4500–8.CrossRefPubMedPubMedCentral
42.
go back to reference Anandhan A, Lei S, Levytskyy R, Pappa A, Panayiotidis MI, Cerny RL, Khalimonchuk O, Powers R, Franco R. Glucose metabolism and AMPK signaling regulate dopaminergic cell death induced by gene (alpha-Synuclein)-environment (Paraquat) interactions. Mol Neurobiol. 2017;54:3825–42.CrossRefPubMed Anandhan A, Lei S, Levytskyy R, Pappa A, Panayiotidis MI, Cerny RL, Khalimonchuk O, Powers R, Franco R. Glucose metabolism and AMPK signaling regulate dopaminergic cell death induced by gene (alpha-Synuclein)-environment (Paraquat) interactions. Mol Neurobiol. 2017;54:3825–42.CrossRefPubMed
43.
go back to reference Lei S, Zavala-Flores L, Garcia-Garcia A, Nandakumar R, Huang Y, Madayiputhiya N, Stanton RC, Dodds ED, Powers R, Franco R. Alterations in energy/redox metabolism induced by mitochondrial and environmental toxins: a specific role for glucose-6-phosphate-dehydrogenase and the pentose phosphate pathway in paraquat toxicity. ACS Chem Biol. 2014;9:2032–48.CrossRefPubMedPubMedCentral Lei S, Zavala-Flores L, Garcia-Garcia A, Nandakumar R, Huang Y, Madayiputhiya N, Stanton RC, Dodds ED, Powers R, Franco R. Alterations in energy/redox metabolism induced by mitochondrial and environmental toxins: a specific role for glucose-6-phosphate-dehydrogenase and the pentose phosphate pathway in paraquat toxicity. ACS Chem Biol. 2014;9:2032–48.CrossRefPubMedPubMedCentral
45.
go back to reference Jain M, Brenner DA, Cui L, Lim CC, Wang B, Pimentel DR, Koh S, Sawyer DB, Leopold JA, Handy DE, et al. Glucose-6-phosphate dehydrogenase modulates cytosolic redox status and contractile phenotype in adult cardiomyocytes. Circ Res. 2003;93:e9–16.CrossRefPubMed Jain M, Brenner DA, Cui L, Lim CC, Wang B, Pimentel DR, Koh S, Sawyer DB, Leopold JA, Handy DE, et al. Glucose-6-phosphate dehydrogenase modulates cytosolic redox status and contractile phenotype in adult cardiomyocytes. Circ Res. 2003;93:e9–16.CrossRefPubMed
46.
go back to reference Leopold JA, Zhang YY, Scribner AW, Stanton RC, Loscalzo J. Glucose-6-phosphate dehydrogenase overexpression decreases endothelial cell oxidant stress and increases bioavailable nitric oxide. Arterioscler Thromb Vasc Biol. 2003;23:411–7.CrossRefPubMed Leopold JA, Zhang YY, Scribner AW, Stanton RC, Loscalzo J. Glucose-6-phosphate dehydrogenase overexpression decreases endothelial cell oxidant stress and increases bioavailable nitric oxide. Arterioscler Thromb Vasc Biol. 2003;23:411–7.CrossRefPubMed
47.
go back to reference Nicol CJ, Zielenski J, Tsui LC, Wells PG. An embryoprotective role for glucose-6-phosphate dehydrogenase in developmental oxidative stress and chemical teratogenesis. Faseb j. 2000;14:111–27.CrossRefPubMed Nicol CJ, Zielenski J, Tsui LC, Wells PG. An embryoprotective role for glucose-6-phosphate dehydrogenase in developmental oxidative stress and chemical teratogenesis. Faseb j. 2000;14:111–27.CrossRefPubMed
48.
go back to reference Park J, Chung JJ, Kim JB. New evaluations of redox regulating system in adipose tissue of obesity. Diabetes Res Clin Pract. 2007;77(Suppl 1):S11–6.CrossRefPubMed Park J, Chung JJ, Kim JB. New evaluations of redox regulating system in adipose tissue of obesity. Diabetes Res Clin Pract. 2007;77(Suppl 1):S11–6.CrossRefPubMed
49.
go back to reference Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V, Floyd BC, Ojaimi C, Bellomo M, Wolin MS, Recchia FA. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J Mol Cell Cardiol. 2006;41:340–9.CrossRefPubMed Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V, Floyd BC, Ojaimi C, Bellomo M, Wolin MS, Recchia FA. Glucose-6-phosphate dehydrogenase-derived NADPH fuels superoxide production in the failing heart. J Mol Cell Cardiol. 2006;41:340–9.CrossRefPubMed
50.
go back to reference Matsui R, Xu S, Maitland KA, Hayes A, Leopold JA, Handy DE, Loscalzo J, Cohen RA. Glucose-6 phosphate dehydrogenase deficiency decreases the vascular response to angiotensin II. Circulation. 2005;112:257–63.CrossRefPubMed Matsui R, Xu S, Maitland KA, Hayes A, Leopold JA, Handy DE, Loscalzo J, Cohen RA. Glucose-6 phosphate dehydrogenase deficiency decreases the vascular response to angiotensin II. Circulation. 2005;112:257–63.CrossRefPubMed
51.
go back to reference Matsui R, Xu S, Maitland KA, Mastroianni R, Leopold JA, Handy DE, Loscalzo J, Cohen RA. Glucose-6-phosphate dehydrogenase deficiency decreases vascular superoxide and atherosclerotic lesions in apolipoprotein E(-/-) mice. Arterioscler Thromb Vasc Biol. 2006;26:910–6.CrossRefPubMed Matsui R, Xu S, Maitland KA, Mastroianni R, Leopold JA, Handy DE, Loscalzo J, Cohen RA. Glucose-6-phosphate dehydrogenase deficiency decreases vascular superoxide and atherosclerotic lesions in apolipoprotein E(-/-) mice. Arterioscler Thromb Vasc Biol. 2006;26:910–6.CrossRefPubMed
52.
go back to reference Park J, Rho HK, Kim KH, Choe SS, Lee YS, Kim JB. Overexpression of glucose-6-phosphate dehydrogenase is associated with lipid dysregulation and insulin resistance in obesity. Mol Cell Biol. 2005;25:5146–57.CrossRefPubMedPubMedCentral Park J, Rho HK, Kim KH, Choe SS, Lee YS, Kim JB. Overexpression of glucose-6-phosphate dehydrogenase is associated with lipid dysregulation and insulin resistance in obesity. Mol Cell Biol. 2005;25:5146–57.CrossRefPubMedPubMedCentral
53.
go back to reference Serpillon S, Floyd BC, Gupte RS, George S, Kozicky M, Neito V, Recchia F, Stanley W, Wolin MS, Gupte SA. Superoxide production by NAD(P)H oxidase and mitochondria is increased in genetically obese and hyperglycemic rat heart and aorta before the development of cardiac dysfunction. The role of glucose-6-phosphate dehydrogenase-derived NADPH. Am J Physiol Heart Circ Physiol. 2009;297:H153–62.CrossRefPubMedPubMedCentral Serpillon S, Floyd BC, Gupte RS, George S, Kozicky M, Neito V, Recchia F, Stanley W, Wolin MS, Gupte SA. Superoxide production by NAD(P)H oxidase and mitochondria is increased in genetically obese and hyperglycemic rat heart and aorta before the development of cardiac dysfunction. The role of glucose-6-phosphate dehydrogenase-derived NADPH. Am J Physiol Heart Circ Physiol. 2009;297:H153–62.CrossRefPubMedPubMedCentral
54.
go back to reference Gupte RS, Floyd BC, Kozicky M, George S, Ungvari ZI, Neito V, Wolin MS, Gupte SA. Synergistic activation of glucose-6-phosphate dehydrogenase and NAD(P)H oxidase by Src kinase elevates superoxide in type 2 diabetic, Zucker fa/fa, rat liver. Free Radic Biol Med. 2009;47:219–28.CrossRefPubMedPubMedCentral Gupte RS, Floyd BC, Kozicky M, George S, Ungvari ZI, Neito V, Wolin MS, Gupte SA. Synergistic activation of glucose-6-phosphate dehydrogenase and NAD(P)H oxidase by Src kinase elevates superoxide in type 2 diabetic, Zucker fa/fa, rat liver. Free Radic Biol Med. 2009;47:219–28.CrossRefPubMedPubMedCentral
55.
go back to reference Lee JW, Choi AH, Ham M, Kim JW, Choe SS, Park J, Lee GY, Yoon KH, Kim JB. G6PD up-regulation promotes pancreatic beta-cell dysfunction. Endocrinology. 2011;152:793–803.CrossRefPubMed Lee JW, Choi AH, Ham M, Kim JW, Choe SS, Park J, Lee GY, Yoon KH, Kim JB. G6PD up-regulation promotes pancreatic beta-cell dysfunction. Endocrinology. 2011;152:793–803.CrossRefPubMed
56.
go back to reference Ham M, Lee JW, Choi AH, Jang H, Choi G, Park J, Kozuka C, Sears DD, Masuzaki H, Kim JB. Macrophage glucose-6-phosphate dehydrogenase stimulates proinflammatory responses with oxidative stress. Mol Cell Biol. 2013;33:2425–35.CrossRefPubMedPubMedCentral Ham M, Lee JW, Choi AH, Jang H, Choi G, Park J, Kozuka C, Sears DD, Masuzaki H, Kim JB. Macrophage glucose-6-phosphate dehydrogenase stimulates proinflammatory responses with oxidative stress. Mol Cell Biol. 2013;33:2425–35.CrossRefPubMedPubMedCentral
57.
go back to reference Ham M, Choe SS, Shin KC, Choi G, Kim JW, Noh JR, Kim YH, Ryu JW, Yoon KH, Lee CH, Kim JB. Glucose-6-phosphate dehydrogenase deficiency improves insulin resistance with reduced adipose tissue inflammation in obesity. Diabetes. 2016;65:2624–38.CrossRefPubMed Ham M, Choe SS, Shin KC, Choi G, Kim JW, Noh JR, Kim YH, Ryu JW, Yoon KH, Lee CH, Kim JB. Glucose-6-phosphate dehydrogenase deficiency improves insulin resistance with reduced adipose tissue inflammation in obesity. Diabetes. 2016;65:2624–38.CrossRefPubMed
58.
go back to reference Voloboueva LA, Emery JF, Sun X, Giffard RG. Inflammatory response of microglial BV-2 cells includes a glycolytic shift and is modulated by mitochondrial glucose-regulated protein 75/mortalin. FEBS Lett. 2013;587:756–62.CrossRefPubMedPubMedCentral Voloboueva LA, Emery JF, Sun X, Giffard RG. Inflammatory response of microglial BV-2 cells includes a glycolytic shift and is modulated by mitochondrial glucose-regulated protein 75/mortalin. FEBS Lett. 2013;587:756–62.CrossRefPubMedPubMedCentral
59.
go back to reference Gimeno-Bayon J, Lopez-Lopez A, Rodriguez MJ, Mahy N. Glucose pathways adaptation supports acquisition of activated microglia phenotype. J Neurosci Res. 2014;92:723–31.CrossRefPubMed Gimeno-Bayon J, Lopez-Lopez A, Rodriguez MJ, Mahy N. Glucose pathways adaptation supports acquisition of activated microglia phenotype. J Neurosci Res. 2014;92:723–31.CrossRefPubMed
60.
go back to reference Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173:649–65.CrossRefPubMed Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173:649–65.CrossRefPubMed
61.
62.
go back to reference Falkowska A, Gutowska I, Goschorska M, Nowacki P, Chlubek D, Baranowska-Bosiacka I. Energy metabolism of the brain, including the cooperation between astrocytes and neurons, especially in the context of glycogen metabolism. Int J Mol Sci. 2015;16:25959–81.CrossRefPubMedPubMedCentral Falkowska A, Gutowska I, Goschorska M, Nowacki P, Chlubek D, Baranowska-Bosiacka I. Energy metabolism of the brain, including the cooperation between astrocytes and neurons, especially in the context of glycogen metabolism. Int J Mol Sci. 2015;16:25959–81.CrossRefPubMedPubMedCentral
64.
go back to reference Volkenhoff A, Weiler A, Letzel M, Stehling M, Klambt C, Schirmeier S. Glial glycolysis is essential for neuronal survival in drosophila. Cell Metab. 2015;22:437–47.CrossRefPubMed Volkenhoff A, Weiler A, Letzel M, Stehling M, Klambt C, Schirmeier S. Glial glycolysis is essential for neuronal survival in drosophila. Cell Metab. 2015;22:437–47.CrossRefPubMed
65.
go back to reference Lee Y, Morrison BM, Li Y, Lengacher S, Farah MH, Hoffman PN, Liu Y, Tsingalia A, Jin L, Zhang PW, et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature. 2012;487:443–8.CrossRefPubMedPubMedCentral Lee Y, Morrison BM, Li Y, Lengacher S, Farah MH, Hoffman PN, Liu Y, Tsingalia A, Jin L, Zhang PW, et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature. 2012;487:443–8.CrossRefPubMedPubMedCentral
66.
go back to reference Bubp J, Jen M, Matuszewski K. Caring for glucose-6-phosphate dehydrogenase (G6PD)-deficient patients: implications for pharmacy. P t. 2015;40:572–4.PubMedPubMedCentral Bubp J, Jen M, Matuszewski K. Caring for glucose-6-phosphate dehydrogenase (G6PD)-deficient patients: implications for pharmacy. P t. 2015;40:572–4.PubMedPubMedCentral
Metadata
Title
The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration
Authors
Dezhen Tu
Yun Gao
Ru Yang
Tian Guan
Jau-Shyong Hong
Hui-Ming Gao
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2019
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1659-1

Other articles of this Issue 1/2019

Journal of Neuroinflammation 1/2019 Go to the issue