Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

Open Access 01-12-2022 | Research

Lactate attenuates astrocytic inflammation by inhibiting ubiquitination and degradation of NDRG2 under oxygen–glucose deprivation conditions

Authors: Jinying Xu, Tong Ji, Guichen Li, Haiying Zhang, Yangyang Zheng, Meiying Li, Jie Ma, Yulin Li, Guangfan Chi

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Background

Brain lactate concentrations are enhanced in response to cerebral ischemia and promote the formation of reactive astrocytes, which are major components of the neuroinflammatory response and functional recovery, following cerebral ischemia. NDRG2 is upregulated during reactive astrocyte formation. However, its regulation and function are unclear. We studied the relationship between lactate and NDRG2 in astrocytes under conditions of ischemia or oxygen–glucose deprivation (OGD).

Methods

We examined astrocytic NDRG2 expression after middle cerebral artery occlusion (MCAO) using western blot and immunofluorescence staining. Under hypoxia conditions, we added exogenous L-lactate sodium (lactate) to cultured primary astrocytes to explore the effects of lactate on the ubiquitination modification of NDRG2. We profiled the transcriptomic features of NDRG2 silencing in astrocytes after 8 h of OGD conditions as well as exogenous lactate treatment by performing RNA-seq. Finally, we evaluated the molecular mechanisms of NDRG2 in regulating TNFα under OGD conditions using western blot and immunohistochemistry.

Results

Reactive astrocytes strongly expressed NDRG2 in a rat model of MCAO. We also showed that lactate stabilizes astrocytic NDRG2 by inhibiting its ubiquitination. NDRG2 inhibition in astrocytes increased inflammation and upregulated immune-associated genes and signaling pathways. NDRG2 knockdown induced TNFα expression and secretion via c-Jun phosphorylation.

Conclusions

We revealed that under OGD conditions, lactate plays an important anti-inflammatory role and inhibits TNFα expression by stabilizing NDRG2, which is beneficial for neurological functional recovery. NDRG2 may be a new therapeutic target for cerebral ischemia.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jin R, Liu L, Zhang S, Nanda A, Li G. Role of inflammation and its mediators in acute ischemic stroke. J Cardiovasc Transl Res. 2013;6:834–51.CrossRef Jin R, Liu L, Zhang S, Nanda A, Li G. Role of inflammation and its mediators in acute ischemic stroke. J Cardiovasc Transl Res. 2013;6:834–51.CrossRef
2.
go back to reference Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation. 2019;16:142.CrossRef Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation. 2019;16:142.CrossRef
3.
go back to reference Giovannoni F, Quintana FJ. The role of astrocytes in CNS inflammation. Trends Immunol. 2020;41:805–19.CrossRef Giovannoni F, Quintana FJ. The role of astrocytes in CNS inflammation. Trends Immunol. 2020;41:805–19.CrossRef
4.
go back to reference Sims NR, Yew WP. Reactive astrogliosis in stroke: contributions of astrocytes to recovery of neurological function. Neurochem Int. 2017;107:88–103.CrossRef Sims NR, Yew WP. Reactive astrogliosis in stroke: contributions of astrocytes to recovery of neurological function. Neurochem Int. 2017;107:88–103.CrossRef
5.
go back to reference Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119:7–35.CrossRef Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119:7–35.CrossRef
6.
go back to reference Rakers C, Schleif M, Blank N, Matuskova H, Ulas T, Handler K, Torres SV, Schumacher T, Tai K, Schultze JL, et al. Stroke target identification guided by astrocyte transcriptome analysis. Glia. 2019;67:619–33.CrossRef Rakers C, Schleif M, Blank N, Matuskova H, Ulas T, Handler K, Torres SV, Schumacher T, Tai K, Schultze JL, et al. Stroke target identification guided by astrocyte transcriptome analysis. Glia. 2019;67:619–33.CrossRef
7.
go back to reference Banerjee A, Ghatak S, Sikdar SK. l-Lactate mediates neuroprotection against ischaemia by increasing TREK1 channel expression in rat hippocampal astrocytes in vitro. J Neurochem. 2016;138:265–81.CrossRef Banerjee A, Ghatak S, Sikdar SK. l-Lactate mediates neuroprotection against ischaemia by increasing TREK1 channel expression in rat hippocampal astrocytes in vitro. J Neurochem. 2016;138:265–81.CrossRef
8.
go back to reference Xu J, Zheng Y, Lv S, Kang J, Yu Y, Hou K, Li Y, Chi G. Lactate promotes reactive astrogliosis and confers axon guidance potential to astrocytes under oxygen-glucose deprivation. Neuroscience. 2020;442:54–68.CrossRef Xu J, Zheng Y, Lv S, Kang J, Yu Y, Hou K, Li Y, Chi G. Lactate promotes reactive astrogliosis and confers axon guidance potential to astrocytes under oxygen-glucose deprivation. Neuroscience. 2020;442:54–68.CrossRef
9.
go back to reference Bohn T, Rapp S, Luther N, Klein M, Bruehl TJ, Kojima N, Aranda Lopez P, Hahlbrock J, Muth S, Endo S, et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat Immunol. 2018;19:1319–29.CrossRef Bohn T, Rapp S, Luther N, Klein M, Bruehl TJ, Kojima N, Aranda Lopez P, Hahlbrock J, Muth S, Endo S, et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat Immunol. 2018;19:1319–29.CrossRef
10.
go back to reference Liu N, Luo J, Kuang D, Xu S, Duan Y, Xia Y, Wei Z, Xie X, Yin B, Chen F, et al. Lactate inhibits ATP6V0d2 expression in tumor-associated macrophages to promote HIF-2alpha-mediated tumor progression. J Clin Invest. 2019;129:631–46.CrossRef Liu N, Luo J, Kuang D, Xu S, Duan Y, Xia Y, Wei Z, Xie X, Yin B, Chen F, et al. Lactate inhibits ATP6V0d2 expression in tumor-associated macrophages to promote HIF-2alpha-mediated tumor progression. J Clin Invest. 2019;129:631–46.CrossRef
11.
go back to reference Lin K, Yin A, Yao L, Li Y. N-myc downstream-regulated gene 2 in the nervous system: from expression pattern to function. Acta Biochim Biophys Sin. 2015;47:761–6.CrossRef Lin K, Yin A, Yao L, Li Y. N-myc downstream-regulated gene 2 in the nervous system: from expression pattern to function. Acta Biochim Biophys Sin. 2015;47:761–6.CrossRef
12.
go back to reference Flügge G, Araya-Callis C, Garea-Rodriguez E, Stadelmann-Nessler C, Fuchs E. NDRG2 as a marker protein for brain astrocytes. Cell Tissue Res. 2014;357:31–41.CrossRef Flügge G, Araya-Callis C, Garea-Rodriguez E, Stadelmann-Nessler C, Fuchs E. NDRG2 as a marker protein for brain astrocytes. Cell Tissue Res. 2014;357:31–41.CrossRef
13.
go back to reference Zhang Z, Ma Z, Zou W, Zhang L, Li Y, Zhang J, Liu M, Hou W, Ma Y. N-myc downstream-regulated gene 2 controls astrocyte morphology via Rho-GTPase signaling. J Cell Physiol. 2019;234:20847–58.CrossRef Zhang Z, Ma Z, Zou W, Zhang L, Li Y, Zhang J, Liu M, Hou W, Ma Y. N-myc downstream-regulated gene 2 controls astrocyte morphology via Rho-GTPase signaling. J Cell Physiol. 2019;234:20847–58.CrossRef
14.
go back to reference Takarada-Iemata M, Kezuka D, Takeichi T, Ikawa M, Hattori T, Kitao Y, Hori O. Deletion of N-myc downstream-regulated gene 2 attenuates reactive astrogliosis and inflammatory response in a mouse model of cortical stab injury. J Neurochem. 2014;130:374–87.CrossRef Takarada-Iemata M, Kezuka D, Takeichi T, Ikawa M, Hattori T, Kitao Y, Hori O. Deletion of N-myc downstream-regulated gene 2 attenuates reactive astrogliosis and inflammatory response in a mouse model of cortical stab injury. J Neurochem. 2014;130:374–87.CrossRef
15.
go back to reference Takarada-Iemata M, Yoshikawa A, Ta HM, Okitani N, Nishiuchi T, Aida Y, Kamide T, Hattori T, Ishii H, Tamatani T, et al. N-myc downstream-regulated gene 2 protects blood-brain barrier integrity following cerebral ischemia. Glia. 2018;66:1432–46.CrossRef Takarada-Iemata M, Yoshikawa A, Ta HM, Okitani N, Nishiuchi T, Aida Y, Kamide T, Hattori T, Ishii H, Tamatani T, et al. N-myc downstream-regulated gene 2 protects blood-brain barrier integrity following cerebral ischemia. Glia. 2018;66:1432–46.CrossRef
16.
go back to reference Yin A, Guo H, Tao L, Cai G, Wang Y, Yao L, Xiong L, Zhang J, Li Y. NDRG2 protects the brain from excitotoxicity by facilitating interstitial glutamate uptake. Transl Stroke Res. 2020;11:214–27.CrossRef Yin A, Guo H, Tao L, Cai G, Wang Y, Yao L, Xiong L, Zhang J, Li Y. NDRG2 protects the brain from excitotoxicity by facilitating interstitial glutamate uptake. Transl Stroke Res. 2020;11:214–27.CrossRef
17.
go back to reference von Karstedt S. NDRG2 programs tumor-associated macrophages for tumor support. Cell Death Dis. 2018;9:294.CrossRef von Karstedt S. NDRG2 programs tumor-associated macrophages for tumor support. Cell Death Dis. 2018;9:294.CrossRef
18.
go back to reference Li M, Lai X, Zhao Y, Zhang Y, Li M, Li D, Kong J, Zhang Y, Jing P, Li H, et al. Loss of NDRG2 in liver microenvironment inhibits cancer liver metastasis by regulating tumor associate macrophages polarization. Cell Death Dis. 2018;9:248.CrossRef Li M, Lai X, Zhao Y, Zhang Y, Li M, Li D, Kong J, Zhang Y, Jing P, Li H, et al. Loss of NDRG2 in liver microenvironment inhibits cancer liver metastasis by regulating tumor associate macrophages polarization. Cell Death Dis. 2018;9:248.CrossRef
19.
go back to reference Wang L, Liu N, Yao L, Li F, Zhang J, Deng Y, Liu J, Ji S, Yang A, Han H, et al. NDRG2 is a new HIF-1 target gene necessary for hypoxia-induced apoptosis in A549 cells. Cell Physiol Biochem. 2008;21:239–50.CrossRef Wang L, Liu N, Yao L, Li F, Zhang J, Deng Y, Liu J, Ji S, Yang A, Han H, et al. NDRG2 is a new HIF-1 target gene necessary for hypoxia-induced apoptosis in A549 cells. Cell Physiol Biochem. 2008;21:239–50.CrossRef
20.
go back to reference Liu J, Zhang J, Wang X, Li Y, Chen Y, Li K, Zhang J, Yao L, Guo G. HIF-1 and NDRG2 contribute to hypoxia-induced radioresistance of cervical cancer Hela cells. Exp Cell Res. 2010;316:1985–93.CrossRef Liu J, Zhang J, Wang X, Li Y, Chen Y, Li K, Zhang J, Yao L, Guo G. HIF-1 and NDRG2 contribute to hypoxia-induced radioresistance of cervical cancer Hela cells. Exp Cell Res. 2010;316:1985–93.CrossRef
21.
go back to reference Munoz Maniega S, Cvoro V, Chappell FM, Armitage PA, Marshall I, Bastin ME, Wardlaw JM. Changes in NAA and lactate following ischemic stroke: a serial MR spectroscopic imaging study. Neurology. 2008;71:1993–9.CrossRef Munoz Maniega S, Cvoro V, Chappell FM, Armitage PA, Marshall I, Bastin ME, Wardlaw JM. Changes in NAA and lactate following ischemic stroke: a serial MR spectroscopic imaging study. Neurology. 2008;71:1993–9.CrossRef
22.
go back to reference Li Y, Wang T, Zhang T, Lin Z, Li Y, Guo R, Zhao Y, Meng Z, Liu J, Yu X, et al. Fast high-resolution metabolic imaging of acute stroke with 3D magnetic resonance spectroscopy. Brain. 2020;143:3225–33.CrossRef Li Y, Wang T, Zhang T, Lin Z, Li Y, Guo R, Zhao Y, Meng Z, Liu J, Yu X, et al. Fast high-resolution metabolic imaging of acute stroke with 3D magnetic resonance spectroscopy. Brain. 2020;143:3225–33.CrossRef
23.
go back to reference Rehncrona S, Rosen I, Siesjo BK. Brain lactic acidosis and ischemic cell damage: 1. Biochemistry and neurophysiology. J Cereb Blood Flow Metab. 1981;1:297–311.CrossRef Rehncrona S, Rosen I, Siesjo BK. Brain lactic acidosis and ischemic cell damage: 1. Biochemistry and neurophysiology. J Cereb Blood Flow Metab. 1981;1:297–311.CrossRef
24.
go back to reference Hillered L, Hallstrom A, Segersvard S, Persson L, Ungerstedt U. Dynamics of extracellular metabolites in the striatum after middle cerebral artery occlusion in the rat monitored by intracerebral microdialysis. J Cereb Blood Flow Metab. 1989;9:607–16.CrossRef Hillered L, Hallstrom A, Segersvard S, Persson L, Ungerstedt U. Dynamics of extracellular metabolites in the striatum after middle cerebral artery occlusion in the rat monitored by intracerebral microdialysis. J Cereb Blood Flow Metab. 1989;9:607–16.CrossRef
25.
go back to reference Harada K, Honmou O, Liu H, Bando M, Houkin K, Kocsis JD. Magnetic resonance lactate and lipid signals in rat brain after middle cerebral artery occlusion model. Brain Res. 2007;1134:206–13.CrossRef Harada K, Honmou O, Liu H, Bando M, Houkin K, Kocsis JD. Magnetic resonance lactate and lipid signals in rat brain after middle cerebral artery occlusion model. Brain Res. 2007;1134:206–13.CrossRef
26.
go back to reference Chaudhari P, Madaan A, Rivera JC, Charfi I, Habelrih T, Hou X, Nezhady M, Lodygensky G, Pineyro G, Muanza T, Chemtob S. Neuronal GPR81 regulates developmental brain angiogenesis and promotes brain recovery after a hypoxic ischemic insult. J Cereb Blood Flow Metab. 2022;42:1294–308.CrossRef Chaudhari P, Madaan A, Rivera JC, Charfi I, Habelrih T, Hou X, Nezhady M, Lodygensky G, Pineyro G, Muanza T, Chemtob S. Neuronal GPR81 regulates developmental brain angiogenesis and promotes brain recovery after a hypoxic ischemic insult. J Cereb Blood Flow Metab. 2022;42:1294–308.CrossRef
27.
go back to reference Hoque R, Farooq A, Ghani A, Gorelick F, Mehal WZ. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology. 2014;146:1763–74.CrossRef Hoque R, Farooq A, Ghani A, Gorelick F, Mehal WZ. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology. 2014;146:1763–74.CrossRef
28.
go back to reference Zhai X, Li J, Li L, Sun Y, Zhang X, Xue Y, Lv J, Gao Y, Li S, Yan W, et al. L-lactate preconditioning promotes plasticity-related proteins expression and reduces neurological deficits by potentiating GPR81 signaling in rat traumatic brain injury model. Brain Res. 2020;1746: 146945.CrossRef Zhai X, Li J, Li L, Sun Y, Zhang X, Xue Y, Lv J, Gao Y, Li S, Yan W, et al. L-lactate preconditioning promotes plasticity-related proteins expression and reduces neurological deficits by potentiating GPR81 signaling in rat traumatic brain injury model. Brain Res. 2020;1746: 146945.CrossRef
29.
go back to reference Lauritzen KH, Morland C, Puchades M, Holm-Hansen S, Hagelin EM, Lauritzen F, Attramadal H, Storm-Mathisen J, Gjedde A, Bergersen LH. Lactate receptor sites link neurotransmission, neurovascular coupling, and brain energy metabolism. Cereb Cortex. 2014;24:2784–95.CrossRef Lauritzen KH, Morland C, Puchades M, Holm-Hansen S, Hagelin EM, Lauritzen F, Attramadal H, Storm-Mathisen J, Gjedde A, Bergersen LH. Lactate receptor sites link neurotransmission, neurovascular coupling, and brain energy metabolism. Cereb Cortex. 2014;24:2784–95.CrossRef
30.
go back to reference Lee DC, Sohn HA, Park ZY, Oh S, Kang YK, Lee KM, Kang M, Jang YJ, Yang SJ, Hong YK, et al. A lactate-induced response to hypoxia. Cell. 2015;161:595–609.CrossRef Lee DC, Sohn HA, Park ZY, Oh S, Kang YK, Lee KM, Kang M, Jang YJ, Yang SJ, Hong YK, et al. A lactate-induced response to hypoxia. Cell. 2015;161:595–609.CrossRef
31.
go back to reference Melotte V, Qu X, Ongenaert M, van Criekinge W, de Bruine AP, Baldwin HS, van Engeland M. The N-myc downstream regulated gene (NDRG) family: diverse functions, multiple applications. FASEB J. 2010;24:4153–66.CrossRef Melotte V, Qu X, Ongenaert M, van Criekinge W, de Bruine AP, Baldwin HS, van Engeland M. The N-myc downstream regulated gene (NDRG) family: diverse functions, multiple applications. FASEB J. 2010;24:4153–66.CrossRef
32.
go back to reference Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–80.CrossRef Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–80.CrossRef
33.
go back to reference Yu J, Chai P, Xie M, Ge S, Ruan J, Fan X, Jia R. Histone lactylation drives oncogenesis by facilitating m(6)A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 2021;22:85.CrossRef Yu J, Chai P, Xie M, Ge S, Ruan J, Fan X, Jia R. Histone lactylation drives oncogenesis by facilitating m(6)A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 2021;22:85.CrossRef
34.
go back to reference Hagihara H, Shoji H, Otabi H, Toyoda A, Katoh K, Namihira M, Miyakawa T. Protein lactylation induced by neural excitation. Cell Rep. 2021;37: 109820.CrossRef Hagihara H, Shoji H, Otabi H, Toyoda A, Katoh K, Namihira M, Miyakawa T. Protein lactylation induced by neural excitation. Cell Rep. 2021;37: 109820.CrossRef
35.
go back to reference Guo H, Yin A, Ma Y, Fan Z, Tao L, Tang W, Ma Y, Hou W, Cai G, Zhuo L, et al. Astroglial N-myc downstream-regulated gene 2 protects the brain from cerebral edema induced by stroke. Glia. 2021;69:281–95.CrossRef Guo H, Yin A, Ma Y, Fan Z, Tao L, Tang W, Ma Y, Hou W, Cai G, Zhuo L, et al. Astroglial N-myc downstream-regulated gene 2 protects the brain from cerebral edema induced by stroke. Glia. 2021;69:281–95.CrossRef
36.
go back to reference Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.CrossRef Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.CrossRef
37.
go back to reference Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, Barres BA. Genomic analysis of reactive astrogliosis. J Neurosci. 2012;32:6391–410.CrossRef Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, Barres BA. Genomic analysis of reactive astrogliosis. J Neurosci. 2012;32:6391–410.CrossRef
38.
go back to reference Dembic Z, Loetscher H, Gubler U, Pan Y, Lahm H, Gentz R, Brockhaus M, Lesslauer W. Two human TNF receptors have similar extracellular, but distinct intracellular, domain sequences. Cytokine. 1990;2:231–7.CrossRef Dembic Z, Loetscher H, Gubler U, Pan Y, Lahm H, Gentz R, Brockhaus M, Lesslauer W. Two human TNF receptors have similar extracellular, but distinct intracellular, domain sequences. Cytokine. 1990;2:231–7.CrossRef
39.
go back to reference Vandenabeele P, Declercq W, Beyaert R, Fiers W. Two tumour necrosis factor receptors: structure and function. Trends Cell Biol. 1995;5:392–9.CrossRef Vandenabeele P, Declercq W, Beyaert R, Fiers W. Two tumour necrosis factor receptors: structure and function. Trends Cell Biol. 1995;5:392–9.CrossRef
40.
go back to reference Hsu H, Xiong J, Goeddel D. The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell. 1995;81:495–504.CrossRef Hsu H, Xiong J, Goeddel D. The TNF receptor 1-associated protein TRADD signals cell death and NF-kappa B activation. Cell. 1995;81:495–504.CrossRef
41.
go back to reference Sedger LM, McDermott MF. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev. 2014;25:453–72.CrossRef Sedger LM, McDermott MF. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev. 2014;25:453–72.CrossRef
42.
go back to reference Wang LW, Chang YC, Chen SJ, Tseng CH, Tu YF, Liao NS, Huang CC, Ho CJ. TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain. J Neuroinflammation. 2014;11:215.CrossRef Wang LW, Chang YC, Chen SJ, Tseng CH, Tu YF, Liao NS, Huang CC, Ho CJ. TNFR1-JNK signaling is the shared pathway of neuroinflammation and neurovascular damage after LPS-sensitized hypoxic-ischemic injury in the immature brain. J Neuroinflammation. 2014;11:215.CrossRef
43.
go back to reference Maddahi A, Kruse LS, Chen QW, Edvinsson L. The role of tumor necrosis factor-alpha and TNF-alpha receptors in cerebral arteries following cerebral ischemia in rat. J Neuroinflammation. 2011;8:107.CrossRef Maddahi A, Kruse LS, Chen QW, Edvinsson L. The role of tumor necrosis factor-alpha and TNF-alpha receptors in cerebral arteries following cerebral ischemia in rat. J Neuroinflammation. 2011;8:107.CrossRef
44.
go back to reference Canedo T, Portugal CC, Socodato R, Almeida TO, Terceiro AF, Bravo J, Silva AI, Magalhaes JD, Guerra-Gomes S, Oliveira JF, et al. Astrocyte-derived TNF and glutamate critically modulate microglia activation by methamphetamine. Neuropsychopharmacology. 2021;46:2358–70.CrossRef Canedo T, Portugal CC, Socodato R, Almeida TO, Terceiro AF, Bravo J, Silva AI, Magalhaes JD, Guerra-Gomes S, Oliveira JF, et al. Astrocyte-derived TNF and glutamate critically modulate microglia activation by methamphetamine. Neuropsychopharmacology. 2021;46:2358–70.CrossRef
45.
go back to reference Kim S, Son Y. Astrocytes stimulate microglial proliferation and m2 polarization in vitro through crosstalk between astrocytes and microglia. Int J Mol Sci. 2021;22:8800.CrossRef Kim S, Son Y. Astrocytes stimulate microglial proliferation and m2 polarization in vitro through crosstalk between astrocytes and microglia. Int J Mol Sci. 2021;22:8800.CrossRef
46.
go back to reference Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, Olson AJ. Autodock4 and AutoDockTools4: automated docking with selective receptor flexiblity. J Comput Chem. 2009;16:2785–91.CrossRef Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, Olson AJ. Autodock4 and AutoDockTools4: automated docking with selective receptor flexiblity. J Comput Chem. 2009;16:2785–91.CrossRef
Metadata
Title
Lactate attenuates astrocytic inflammation by inhibiting ubiquitination and degradation of NDRG2 under oxygen–glucose deprivation conditions
Authors
Jinying Xu
Tong Ji
Guichen Li
Haiying Zhang
Yangyang Zheng
Meiying Li
Jie Ma
Yulin Li
Guangfan Chi
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02678-6

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue